Skip to main content

The contribution of pattern recognition receptor signalling in the development of age related macular degeneration: the role of toll-like-receptors and the NLRP3-inflammasome

Abstract

Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss, characterised by the dysfunction and death of the photoreceptors and retinal pigment epithelium (RPE). Innate immune cell activation and accompanying para-inflammation have been suggested to contribute to the pathogenesis of AMD, although the exact mechanism(s) and signalling pathways remain elusive. Pattern recognition receptors (PRRs) are essential activators of the innate immune system and drivers of para-inflammation. Of these PRRs, the two most prominent are (1) Toll-like receptors (TLR) and (2) NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3)-inflammasome have been found to modulate the progression of AMD. Mutations in TLR2 have been found to be associated with an increased risk of developing AMD. In animal models of AMD, inhibition of TLR and NLRP3 has been shown to reduce RPE cell death, inflammation and angiogenesis signalling, offering potential novel treatments for advanced AMD. Here, we examine the evidence for PRRs, TLRs2/3/4, and NLRP3-inflammasome pathways in macular degeneration pathogenesis.

Introduction: innate immunity, inflammation and PRRs in AMD

Age-related macular degeneration is a disease that affects central vision and is strongly associated with the activation of the innate immune system. It is the leading cause of irreversible visual impairment in developed nations [1], affecting approximately 196 million people, and likely to grow to 288 million by 2040 [2]. AMD progresses slowly and is characterized by the loss of photoreceptors and their support cells, the retinal pigment epithelium (RPE). In its early and intermediate stages, AMD is characterised by changes in RPE pigmentation and by the presence of drusen, waste deposits between the RPE and Bruch’s membrane [3]. Advanced or late-stage AMD presents in two distinct forms: geographic atrophy (GA), where patches of the RPE and photoreceptors die, and neovascular AMD (nAMD), which involves abnormal growth of blood vessels from sub-RPE, subretinal or intraretinal locations. The current treatment for nAMD involves regular intravitreal injections of anti-vascular endothelial growth factor (VEGF) agents, and recently, injection of a combination of anti-VEGF together with an anti-angiopoietin-2 (Ang2) has been introduced to stabilize vision loss [4,5,6]. However, despite the effectiveness of anti-VEGF agents, approximately half of treated individuals experience significant vision loss over five to seven years [7, 8]. Consequently, new treatments are needed for AMD. Those that target the innate immune system are particularly promising given the role of immunity in AMD [9].

The innate immune system is a non-specific host defence system that aims to eliminate foreign pathogens prior to activation of an adaptive immune response. There is compelling evidence for the role of innate immunity, and more recently, for the role of the adaptive immune system in the development and progression of AMD [10]. While the mounting evidence for the adaptive immune system involvement is reviewed in [10, 11], the focus of the present review will be on the innate immune system and the importance of para-inflammation in the development of AMD. Para-inflammation is a theory that describes a response of the immune system to low levels of stress [12]. In the ageing eye, the trigger for para-inflammation is the accumulation of oxidized lipids and proteins and other altered metabolic products that occurs with years of oxidative stress [12]. With respect to AMD, para-inflammation has been postulated to be important in the formation of drusen because it may develop with age and/or a dysfunctional RPE [13]. Drusen also contain mediators of inflammation (e.g. C-reactive protein) including many complement related proteins (e.g. C3a and C5a) suggesting chronic activation of the innate immune system in the development of early stage disease [12, 13].

One of the most notable links between AMD and the innate immune system is the prevalence of inherited mutations in genes related to innate immunity, particularly genes encoding the complement factor pathways including complement factor H (CFH), complement factor I (CFI), complement factor B (CFB), complement component 2 (C2), and complement component 3 (C3) [14, 15]. One variant of the CFH gene (the Y402H allele) is present in at least 60% of individuals with AMD and increases the risk of developing AMD by 2.0-to-7.3-fold [16]. Innate immune cells are attracted to the retina during early-stage AMD by the immunogenic material within drusen that then stimulate local inflammation [17,18,19,20]. The recently approved antibody injection (Pegcetacoplan) for treating GA targets C3 and has been shown to decrease GA lesion size and, in one study, to reduce drusen load and slow progression [21, 22]. In the past 25 years of AMD clinical trials, this is the first approved treatment for GA, demonstrating the potential of immune cell targets for treating AMD progression.

The primary innate immune cells associated with AMD are the mononuclear phagocytes. These cells are phagocytic and can promote inflammation within the eye [23]. Mononuclear phagocytes can be blood-derived monocytes, dendritic cells, tissue resident macrophages, or microglia (see Fig. 1). Under normal aging conditions, mononuclear phagocytes are not typically observed in the RPE-Bruch’s membrane nor within the subretinal space (between the RPE and photoreceptors). In AMD, mononuclear phagocytes have been detected in AMD lesions, both in specific forms of early-stage disease as well as late-stage AMD lesions, including both GA and nAMD [23,24,25,26,27,28,29,30,31]. Moreover, these mononuclear phagocytes cells have been demonstrated to exacerbate inflammation and, in some cases, promote proangiogenic signalling [32, 33].

Central to understanding the role of innate immune activation in AMD is identifying the causes of immune system activation. One potential mechanism is via activation of pattern recognition receptors (PRRs), which detect pathogens and general cell damage [34]. Importantly, innate immune cells within AMD lesions, including mononuclear phagocytes and neutrophils, all express high levels of PRRs [23,24,25,26,27,28,29,30,31]. PRRs sense molecular patterns of damage, known as danger-associated molecular patterns (DAMPs), and patterns associated with pathogens, such as viruses, bacteria, and parasites (called pathogen-associated molecular patterns or PAMPs). PRR activation by DAMPs occurs in the absence of infections when tissue is damaged or stressed, as in AMD.

The DAMPs that activate pattern recognition receptors are produced by stressed cells in the retina and by drusen, a pathological feature of AMD. Factors such as reactive oxidative species (ROS) [35], glycation end products [36], lysosomal damage to RPE [37], Alu RNA [38], damaged mitochondrial DNA (mtDNA; [39] and inflammatory drusen components, such as amyloid-β oligomer (Aβ-oligomer) can mediate the activation of PRRs by DAMPs [40], which in turn can lead to the potentiation of inflammation and para-inflammation via release of pro-inflammatory cytokines. Alongside the many types of DAMPs, there are also several different classes of PRRs that mediate innate immune cell signalling. Some prominent PRRs include the Toll-like receptors (TLRs), Retinoic acid-inducible gene (RIG)-I-like receptors, and NOD-like receptors (NLRs) [41]. Examples of DAMP and PAMP signals and expression of TLRs and NLRP3 in the retina are depicted in Fig. 1. This review provides an overview of the mechanisms by which DAMPs regulate innate immune cell activation via the different PRRs. In particular, the role of TLRs and NLRP3 pattern recognition receptors will be discussed, and the potential for targeting these PRRs for reducing the symptoms of AMD highlighted.

Fig. 1
figure 1

PAMP and DAMP stimulation of pattern recognition receptors within the retina. An overview of the different pathogen-associated molecular patterns and damage-associated molecular patterns that can stimulate pattern recognition receptors in the retina. Pattern recognition receptors are critical for host defence responses to pathogen infections. Pathogens such as bacteria, viruses, and fungi present pathogen-associated molecular patterns that stimulate toll-like and NOD-like receptors. In age-related macular degeneration and experimental models of macular degeneration, damage-associated molecular patterns have been shown to injure retinal pigment cells and amplify inflammatory responses. Damage-associated molecular patterns of Alu RNA, fibrinogen, and Aβ-oligomer found within drusen, ROS, ATP, HMBG-1 released by damaged/dying photoreceptors and cathepsin from retinal pigment cells have been shown to stimulate either toll-like receptors and NOD-like receptors, on retinal pigment cells or innate immune cells that are found within the subretinal space. (Amyloid β-oligomer – Aβ-oligomer, ATP – Adenosine Tri Phosphate, BM – Bruch’s Membrane, CC – Choriocapillaris, DAMP – Danger Associated Molecular Pattern, HMBG-1 - High mobility group box 1 protein, PAMP – Pattern Associated Molecular Pattern, PR – Photoreceptor – PRR – Pattern Recognition Receptors – RPE – Retinal Pigment Epithelium ROS – Reactive Oxidative Species – SR – Subretinal space)

Subtypes of pattern recognition receptors and their signalling mechanisms

TLRs and NLRP3 are the most extensively studied pattern recognition receptors (PRRs). TLRs play a crucial role in the host defence against infections by detecting evolutionarily conserved structures expressed by various pathogens. However, TLRs also respond to DAMP ligands released by damaged, dead, or dying cells, such as fibrinogen, lipid oxidation products, and high mobility group box 1 (HMGB1) protein [42,43,44].

NLRP3 is activated by DAMPs found in drusen, such as Aβ-oligomer [45,46,47]. In addition to TLRs and NLRP3, recent discoveries have revealed other PRRs that sense cellular RNA or DNA, including cyclic GMP-AMP synthase (cGAS), a DNA sensor, and C-type lectin receptors that recognize glycans [48]. The purinergic P2 × 7 receptor responds to high levels of ATP, a DAMP signal released from dying cells and a co-receptor for NLRP3 [49]. Among these receptors, TLRs 1–9, NLRP3, and P2 × 7 have been extensively studied in the context of AMD. The expression of TLRs, NLRP3, and P2 × 7 receptors in the posterior eye, including the outer retina, choroid, and RPE, is summarized in Table 1.

Table 1 A summary of the cell type in retina and choroid that express pattern recognition receptors

There are ten known human TLRs (TLR1-TLR10) and 12 mouse TLRs (TLR1-TLR9, TLR11-TLR13) [50]. These receptors respond to structures found in bacteria, fungi, mycoplasma, protozoa, and nucleic acids expressed by viruses. The cellular location of TLRs differs, with nucleic acid sensing TLRs (TLR3, TLR7/8, TLR9) located within the endoplasmic reticulum to prevent abnormal activation by self-derived nucleic acids. In contrast, the “anti-microorganism” TLRs (TLR1, TLR2, TLR4, TLR5, TLR6) are expressed in the plasma membrane to detect pathogens within extracellular spaces. TLR4 is unique because it can be internalised by cells to endosomes following bacterial lipopolysaccharides (LPS) stimulation.

TLR stimulation by PAMPs/DAMPs ultimately results in inflammation. However, the intracellular signalling is complex and interwoven, much like a ‘choose your own adventure’ novel. The intracellular signalling and receptor-proximal proteins below will be described for TLRs and depicted in Fig. 2. Upon binding of a DAMP/PAMP, TLRs initiate intracellular signalling via adaptor proteins, of which there are five types (Fig. 2). The adaptor proteins are MyD88 (myeloid differentiation primary-response gene 88), MyD88-adapter-like (MAL), TRIF-related adaptor molecule (TIR-domain-containing adapter-inducing interferon-β, also referred to as TICAM1), and TRAM (TRIF-related adaptor molecule). MyD88 is a universal adaptor utilized by all TLRs except TLR3. MAL is used by TLR2, TLR4, TLR6, TLR7/8, and TLR9 [51]. MyD88 interacts with interleukin-1 receptor-associated kinases (IRAK), IRAK4, and IRAK1/2, while TRAF6 stimulates the transcription factors NF-κB, Activator protein 1 (AP-1), and CREB (cAMP response element). These three (NF-κB, AP-1 and CREB) nuclear transcription factors produce cytokines and chemokines such as tumour necrosis factor (TNF-α), interleukins (ILs) IL-1β, IL-6, IL-18, and chemokines MCP-1 and CXCL10 [52]. TRAF6 or TRIF can stimulate type I interferon responses via IRF3/5/7 [52]. The transcription factor NF-κB, in combination with a second trigger (e.g., ATP or viral RNA), can activate the NLRP3 inflammasome, resulting in the activation of caspase-1 and the release of pro-inflammatory cytokines (e.g., IL-1β) [53, 54]. The endosome located TLRs (TLR3 and TLR4 endocytosed) utilize TRIF to initiate signalling. TLR4, when endocytosed, requires TRAM as its proximal protein and not MyD88. These endosome-located receptors (TLR3/4) stimulate TRIF to either interact with FAS-associated death domain protein (FADD) to induce apoptosis, TRAF6 to produce cytokines and chemokines, or TRAF3 to result in type 1 interferon responses [51].

Fig. 2
figure 2

The complexity of TLR signalling and different types of inflammation, apoptosis and antiviral responses. Depending on the receptor activated, TLR stimulation leads to two parallel outcomes: cytokine and chemokine release (via MyD88, orange) or interferon responses (via TRAF, TRIF, IRF, green). Inflammasome activation can occur by directly stimulating NLRP3 (not shown) or NF-κB. TLRs can be either monomers or dimers that have co-receptors (CD14 for TLR4) or adaptor protein MAL for TLR2, TLR4, TLR7/8 and TLR9, or TRAM for endocytosed TLR4. The pathway has been adapted from [52] and simplified not to include the full extent of intracellular signalling proteins

The NLRs (nucleotide-binding oligomerization domain and leucine-rich repeat-containing receptors) include NLRP3, AIM2, IFI16, and pyrin. DAMP stimulates NLRP3 directly, or following TLR activation [54]. Activating the NLRP3 inflammasome complex involves multiple steps, including priming and activation. Priming requires the upregulation of NLRP3 or elevation of pro-IL-1β (via NF-κB), while activation involves oligomerization and assembly of the inflammasome. The priming step can be induced by a broad range of factors, including PAMPs or DAMPs, (e.g., ATP), lysosomal destabilization products, (e.g., cathepsin B) [53], and components of drusen such as Aβ-oligomer [55]. The oligomerized and assembled inflammasome activates caspase-1 to cleave pro-IL-1β and pro-IL-18 into mature forms [56]. The NLRP3 inflammasome can be activated via canonical signalling that requires caspase-1 activation and non-canonical signalling which utilises caspase-4/5 (caspase-11 in mice) and gasdermin [57]. The subsequent activation of the NLRP3 inflammasome (either canonical or non-canonical) is the release of inflammatory mediators (i.e. IL-1-β or IL-18) that recruits immune cells and potential to induce pyroptosis, a form of cell death.

In summary, TLR and NLRP3 signalling is complex with numerous proteins capable of crosstalk to produce cytokines, chemokines, interferons, or apoptosis processes. The stimulation of chemokines and cytokines recruits immune cells, regulates cell death in inflammation, and can affect vascular endothelial cell permeability [56]. The upregulation of IRF proteins stimulates the transcription and translation of type-1-interferons such as IFN-α and IFN-β, which are essential in antiviral responses and modulating immune responses [57]. The expression of TLRs and NLRP3 is throughout ocular tissues and the responses they initiate in the eye and in AMD are considered below.

Ocular expression of TLRs and NLRP3

To understand how the pattern recognition receptors, TLRs and NLRP3 contribute to AMD, it is first necessary to have a firm understanding of the cell types that express these PRRs. Ocular surfaces interface with the external environment and provide an active barrier against pathogens, and correspondingly TLRs are highly expressed in the eye in immune and non-immune cells [58]. Within the posterior eye, TLRs and NLRP3 are highly expressed in mononuclear phagocytes [59]. In the retina, expression of TLRs and NLRP3 is high on cells that form the blood retinal barrier including RPE [60,61,62], Müller glia, microglia, and endothelial cells. In addition, some retinal neurons including bipolar, amacrine, and ganglion cells also express TLRs [63,64,65]. TLRs may support vision-related processes beyond immunity in the RPE. For example, TLR4 expressed by RPE cells is thought to contribute to photoreceptor outer segment renewal [66]. The following section will consider the expression and function of each subtype of TLR and NLRP3 within the retina and choroid under healthy conditions.

Innate immune cells of the retina, choroid and blood

TLR signalling initiates inflammatory responses in circulating and resident myeloid derived immune cells (i.e., neutrophils and monocytes) in the choroid. Within the posterior eye, circulating immune cells are usually restricted from entering the retina due to tight junctions at the RPE that form the outer retinal blood barrier and endothelial cells of the inner retinal barrier. However, circulating immune cells can migrate to retina and choroid following damage, inflammation or infection [67]. The circulating myeloid derived cells (e.g., monocyte macrophages) and resident myeloid derived cells in the choroid; dendritic cells, macrophages and mast cells all express TLRs (notably TLR2/3/4/9) with variation between myeloid type and NLRP3 [68,69,70,71].

In the retina, mononuclear phagocytes derived from circulating myeloid cells or residential in origin highly express TLRs and NLRP3 with some variation in the subtypes [72]. Microglia are the resident macrophages of the retina and are derived from primitive myeloid precursors that arise in the embryonic yolk sac [73]. Microglia are distributed across the retina to monitor and contribute to retinal homeostasis [74]. Microglia can respond to inflammation and sense foreign pathogens and damage (e.g., DAMPs) and retinal studies have shown they express TLR1/2/3/4/6/7/13 mRNA and protein expression for TLR2 and TLR4 [75,76,77,78]. Studies in vitro report that retinal microglia stimulated directly by pathogens or LPS produce high levels of TLR2 and TLR4 and proinflammatory mediators, TNF-α and MIP-2 [76, 79, 80]. Retinal microglia express NLRP3 and in vitro studies have demonstrated they form inflammasomes following NLRP3 stimulation and produce pro-inflammatory cytokines such as TNF-α, CXCL-1, CSF-1, IL-6, IL-1β, and IL-18 [81]. Consequently, mononuclear phagocytes, whether resident or derived from monocytes in circulation plays a critical role in inflammation via TLR and NLRP3 signalling.

Retinal pigment epithelial cells

As the interface and barrier between the choroidal vasculature and neural retina, the RPE expresses many TLRs and NLRP3 as part of the innate immune defence system. Cultured human RPE cells have been shown to express all TLR1-10 mRNA, except for TLR7/8 [61] while TLR1-6 and TLR9 have been detected at the protein level [82]. One study shows the expression of TLR7 in RPE cells in mice, suggesting a role in mediating pro-cytokine release in small mammals [83]. TLR2 protein expression has been detected in ex-vivo human tissue and mice [84]. TLRs, as immune regulatory receptors, have been found to directly respond to pathogens or TLR2-4 agonists in human and porcine RPE cultures, producing pro-inflammatory cytokines (e.g., MCP-1, IL-6, IL-8). This cytokine release can influence RPE barrier function and contribute to inflammation in the outer retina [85,86,87]. The co-receptor of TLR4, CD14, shows expression in adult human RPE and function as demonstrated by responding to LPS in culture [88]. Blocking TLR3, TLR4 or CD14 in cultured human RPE reduced cytokines (e.g. IL-8) in response to LPS and reduction of IFN-β in response to poly I:C treatment [61, 89]. These findings demonstrate that RPE cells can robustly respond to, and initiate TLR-driven inflammatory processes triggered by DAMPs or PAMPs.

RPE are also phagocytes that engulf photoreceptor segments to maintain normal vision. An antibody inhibitor of TLR4 was shown to block phagocytosis of human photoreceptor outer segments, not bovine outer segments [90]. This study by Kindzelskii et al. showed that TLR4 stimulation by photoreceptor outer segments initiated calcium signals in human RPE [90]. A subsequent study in TLR4 knockout mice showed reduced photoreceptor function using the ERG [66]. These studies show that TLR4 plays either a small role in RPE phagocytosis or is needed for normal vision.

The presence of NLRP3 inflammasome in RPE cells has been debated in age-related macular degeneration research. Investigations of NLRP3 expression in single-cell human data for the RPE-choroid tissues show low expression of NLRP3 compared to macrophages [91, 92]. In support of this finding, a study of human RPE cell lines found no mRNA, protein, or evidence of NLRP3 inflammasome activation in the RPE [93]. Regardless of the low mRNA expression of NLRP3, several studies have demonstrated the presence of functional NLRP3 protein in human RPE and using genetic knockdown in mice [62, 94,95,96,97]. Stimulation of NLRP3 in RPE cells triggers the formation and activation of the NLRP3 inflammasome, leading to the formation of cytokines. NLRP3 stimulations have been observed with agonists such as A2E, iron, and complement proteins [98,99,100]. The NLRP3 inflammasome can process different cytokines, including IL-1β, IL-6 and IL-18, with the specific cytokines produced in RPE cells depending on the stimulus [97, 98, 100, 101]. For instance, IL-18 is produced following 7-Ketocholesterol application, IL-1β is induced by ultraviolet B irradiation, and both IL-18 and IL-1β are generated in response to A2E [94, 98, 102]. These contradictory findings may be explained by differing levels of functional protein produced in the ARPE-19 cell line, which forms the bulk of studies of NLRP3 in RPE and human primary RPE culture study by Kosmidou et al. [93]. Although the immortalized ARPE-19 cell line is widely used in retinal research, there are proteome differences between these cultures of primary RPE and ARPE-19 [103]. ARPE-19 expresses higher levels of proliferation and cell-death proteins, including IL-18 compared to primary RPE, which may explain the difference in observations in human single-cell data and human RPE cultures [103].

Endothelial cells and melanocytes

Like RPE, vascular endothelial cells interface between the circulating blood and neural tissues. In the human and mouse choroid and retina, vascular endothelial cells express high levels of TLR3 with lower expression of TLR2, TLR4 and TLR6-10 [64, 65, 104]. Moreover, activation of TLR4 in retinal endothelial cells has been demonstrated to mediate inflammatory effects in animal models [105, 106]. Melanocytes within the choroid have been reported to express mRNA for TLRs1-10 and the TLR adaptor protein MyD88 in human tissue [107]. Human melanocytes respond to TLR agonists to produce pro-inflammatory cytokines (MCP-1 and IL-8) and chemokines (CXCL1 and CXCL2) indicating the TLRs are functional and likely to contribute to choroidal inflammatory responses from either melanocytes or vascular endothelial cells [107, 108].

Müller glia and astrocytes in the retina

Müller glia and astrocytes are macroglial cells in addition to microglia, the resident immune cells of the retina. Müller glia tile the retina and contact photoreceptors, neuronal soma, neurites and blood vessels throughout the retina. The Müller glia functions to support the neurons and respond to virtually all pathogenic stimuli [109]. Müller glia express TLR5 mRNA and show protein expression for TLR2, TLR3, and TLR4 [110]. Astrocytes express TLR2, TLR3 and TLR4 with TLR3 expression being the predominant form identified in human diseased eyes [77]. Müller glia TLRs are functional, as demonstrated by their expression of TLR adaptor proteins (MyD88 and TRAF6) and interferon intracellular signalling (IRF) following stimulation [63, 110]. Stimulation of TLR in cultured Müller glia produces cytokines (e.g., IL-6) and interferons (e.g., IFN-α and IFN-β). Cultured retinal astrocytes respond to TLR agonists by upregulating major histocompatibility complex II (MHCII), production of cytokines (IL-6, IL-12 and IL-23) and may alter autoimmunity pathology in vivo [77, 111]. Müller glia could be capable of NLRP3 inflammasome activation because they express IL-1β and caspase-1. However, no observations of NLRP3 expression in Müller glia have been reported [112]. Astrocytes have been shown to express NLRP3 protein and produce inflammatory cytokines following NLRP3 inflammasome activation in the retina [113]. In summary, Müller glia and astrocytes act as resident immune cells to signal via TLR2/3/4 (and NLRP3 in astrocytes) to produce inflammatory responses to damage or infection.

Retinal neurons

There is evidence to show that TLRs are expressed in retinal neurons although no studies support a functional role in sensing foreign pathogens or damage. Of the retinal neurons, mRNA and protein of TLRs (TLR4/5/6/7) have been detected in bipolar, amacrine and ganglion cells [63, 110, 114, 115]. TLR4 may have an essential role in bipolar cell function, because bipolar cell dendrites were reduced with a lower microglial density in the retinae of TLR4 knockout mice [66]. This reduction in bipolar dendrites may have been due to microglial expression rather than bipolar cells as microglia can influence the neuronal and synapse development in the retina [74]. TLR3 and TLR4 expression has been reported in an immortalized mouse cone cell line (661 W) and rodent rods, although no supporting evidence for expression in photoreceptors has been demonstrated in primates [115,116,117,118]. NLRP3 expression has been detected in cone photoreceptors, cells of the inner retina (amacrine or bipolar cells) and ganglion cells [113, 119,120,121]. No studies have demonstrated whether NLRP3 and TLRs on retinal neurons respond to damage or pathogen signals. It has yet to be determined how functional neuronal PRRs in the retina are.

In summary, there is a broad expression of TLRs and NLRP3 in cells of the posterior eye, particularly the mononuclear phagocytes, endothelial cells and the RPE. The activation of these pattern recognition receptors has been associated with the release of proinflammatory cytokines in response to PAMP or DAMP stimuli. In the next section, we consider their role in the development of AMD.

The contribution of toll-like receptors to AMD

Genetic associations and AMD

TLR mutations have been linked with the development of AMD based on ​Genome Wide Association Studies (GWAS) [122,123,124]. TLR single nucleotide polymorphisms can either result in a loss or gain in function in the receptors and depending on the SNP range from rare variants (e.g., TLR4 polymorphism D299G) to more prevalent variants (e.g., TLR3 polymorphism L412F) [125]. Each TLR subtype will be reviewed separately.

TLR2 polymorphisms have been associated with an increased risk of AMD [123]. The TLR2 polymorphism (rs5743708) increases the risk of developing geographic atrophy and nAMD in the Turkish population in 383 patients [123]. This TLR2 polymorphism is a loss of function variant in transfected HEK cells [126]. However, additional studies are needed in other populations to confirm this association between TLR2 and AMD.

TLR3 has been reported to be associated with AMD risk with differences based on ethnicity reported between Caucasian and Asian populations [124]. A link between TLR3 and AMD was first reported in a GWAS of 2055 patients, with the rs3775291 polymorphism associated with protection from AMD [124]. This variant (rs3775291) showed a reduced likelihood of developing geographic atrophy in Caucasians but not in Han Chinese patients [124]. The rs3775291 polymorphism is a loss of function variant that shows reduced binding to double stranded RNA (dsRNA) and subsequently smaller production of NF-κB (a transcription factor that regulates inflammation) in transfected ARPE19 cells, as well as protection from Poly I: C induced apoptosis in cultured human RPE cells [124, 127]. Despite rs3772591 supposed protection, subsequent studies that included the rs3772591 allele and other common TLR3 variants found no association with AMD in Caucasian cohorts in the USA, nor three case controls in Caucasian cohorts from the USA and Australia, or for nAMD and PCV in North Indian or ethnic Chinese population [125, 128,129,130]. In summary, the evidence of TLR3 variants and reducing AMD risk is limited.

TLR4 polymorphisms have conflicting evidence to be linked to AMD susceptibility, with differences reported across global populations. A GWAS (1106 patients) reported the association of a rare SNP (rs498690) with an increased risk of AMD in Caucasians [131]. One small study (223 patients) showed an association for both rs4986790 and rs4986791 in a Greek population for nAMD [132]. These results have not been replicated, with subsequent GWAS finding no association between TLR4 (rs498690) and AMD risk in Caucasian and Indian populations [125, 133,134,135]. Meta-analysis that includes the studies mentioned above did not find an increased risk of developing AMD for the TLR4 SNPs rs4986790 nor rs4986791 [136]. Studies of the rs4986790 SNP in monocytes have shown that the function of TLR4 is impaired with reduced binding of the bacterial analogue lipopolysaccharide (LPS) [137]. Despite the loss of function for this SNP (rs4986790), insufficient evidence currently supports an association between TLR4 polymorphism and AMD risk.

In summary, there is weak and conflicting evidence to link SNPs in TLRs to the development of AMD. Despite this, there is substantial cause to continue researching TLRs and AMD, based on their complexity and importance to innate immunity. For example, the TLR2 mutation was shown in one study to increase risk of both GA and nAMD [123]. The rs5743708 mutation is a loss of function SNP meaning that TLR2 would have a diminished response to PAMP or DAMP stimuli. If para-inflammation drives AMD, would not a reduced inflammatory response resulting from a loss of function SNP benefit the eye? The issue could be understood in the redundancy in the system within TLR signalling. Stimulation by PAMPs or DAMPs activates multiple TLRs that ensure crucial stimuli are not missed by either TLRs or the innate immune system. A single TLR2 mutation, for example, could be compensated by the other functional bacterial TLRs (e.g., TLR1/4/5/6). Another consideration is the subtly of SNP mutation compared to monogenic diseases of TLR genes on protein function [71]. TLR monogenic diseases severely alter the immune system as TLRs play a key role in regulating the development of the immune system and function in adulthood [71]. TLRs are essential to innate immunity. While the association between SNPs and AMD is limited, robust evidence links TLR signalling to AMD pathology and suggests that targeting TLRs can preserve vision in AMD.

TLR activation and AMD studies in humans and animal models

TLRs are linked to AMD based on studies in humans and animals where inhibition benefits vision. The RPE highly expresses TLR2/TLR3 and targeting TLR2 and TLR4 are effective for treating RPE degeneration and choroidal neovascularization in pre-clinical models. DAMP ligands that stimulate TLRs are associated with AMD such as Alu RNA and tenascin-C, which are expressed at higher levels in humans with AMD. Evidence for the expression in humans, and treatments targeting TLRs for neovascularisation and preserving the RPE will be discussed.

TLRs can be stimulated by DAMPs, some of which have been linked to AMD. The following DAMPs can activate TLRs: heat-shock proteins (e.g., HSP70 [138], HMGB1 [139], extracellular matrix (ECM) molecules such as tenascin-C [140], and fragments of ECM molecules that are found within the Bruch’s membrane, retina and vitreous (e.g., hyaluronic acid (HA) [141] and heparan sulphate (HS) [142]. All of these TLR stimuli have been detected within the retina, however only one study (tenascin-C) has directly linked a change in these DAMP stimuli within AMD eyes [143,144,145,146,147]. The stimuli for TLR3, ssRNA in the form of Alu RNA has been detected in the drusen and lesions of AMD eyes (as discussed in the NLRP3 section) [38] with no differences in systemic Alu RNA expression [148].

In AMD patients, TLRs expression is higher and more responsive to TLR ligands than age matched controls. Expression of TLR2 and TLR3 is higher in AMD patients’ peripheral blood monocytes (PBMCs) compared to age-matched controls [149]. Stimulation of isolated PBMC using agonist for TLR3, poly(I:C), produced higher levels of IL-6 and IL-8 in nAMD patients indicating that systemic para-inflammation is likely higher in nAMD patients. Studies of TLR expression in excised CNV membranes show that TLR3 expression is exclusively found within the RPE with no expression on endothelial cells or fibroblasts [150]. The RPE was also the site of TLR2 expression in formalin fixed samples of age-matched and nAMD retinas [44]. The primary downstream target of TLR2, NF-κB was elevated in early and intermediate AMD in the RPE and cells of the choroid; endothelial cells and leukocytes and not in nAMD [44]. These studies indicate that the RPE is a crucial site for TLR2/TLR3 activation in early and intermediate AMD.

Targeting TLR2 in animal studies has shown that suppression can preserve visual function in retinal degeneration and reduced neovascularization. Gene expression of TLR2 and its downstream effector proteins (Myd88 and IL-1β) in the retina increases in models of retinal degeneration such as rd10 and P23H [151]. Genetic ablation of TLR2 in these two retinal degeneration models preserved vision [151]. Consistent with this study, knockout of MyD88 in rd1 mouse model was shown to reduce microglia, reduce retinal expression of chemokines (e.g. CXC10) and preserve vision [152]. In a mouse model of nAMD, stimulating TLR2 using a synthetic agonist (PAM3CSK4) increased the size of choroid neovascular lesions and was correlated with an increase of macrophages at the lesion sites [153]. In a spontaneous model of nAMD (JR5558) and the control strain (C57/Bl6), TLR2 was exclusively expressed in the RPE/choroid as seen in gene and protein expression data [44]. The JR5558 strain had higher levels of TLR2 expressed within the choroid/RPE and that antagonism of TLR2 using antibodies reduced neovascularisation in this model [44]. These studies agree with the first laser induced CNV study that showed genetic ablation of TLR2 or it is downstream adaptor protein MyD88 combined with TLR stimulation (C. pneumoniae antigen) reduced CNV lesions [154]. The ability of TLR2 to modify angiogenesis, has also been shown in a mouse model of oxygen induced retinopathy whereby genetic ablation of TLR2 resulted in attenuation of angiogenesis [155].

Treatments targeting TLR3 have a controversial role in AMD, whereby stimulation could benefit the eye by reducing neovascularisation or harm healthy RPE. Clinical trials where short interfering RNA (siRNA) was used to block VEGF production in nAMD were halted due to a lack of efficacy compared to the placebo [156]. Later, the siRNA used to block VEGF formed duplexes of dsRNA [104, 157] similar to viral dsRNA and a well-described ligand of TLR3. Although dsRNA could benefit an animal model of CNV (via TLR3) the clinical trials in humans saw no improvements in vision [158]. In additional mouse studies, nucleotides were injected into healthy young animals, either as the nucleotides used in the nAMD clinical studies (siRNA) or poly(I:C) a synthetic long dsRNA TLR3 ligand. Both siRNA and poly(I:C) were neurotoxic to the RPE and photoreceptors [104, 159]. These data suggest TLR3 activation damages RPE and photoreceptors with inhibition showing limited protection in humans to treat nAMD.

Inflammation and angiogenesis can be modified by TLR4 as observed in ocular disease animal model studies. TLR4 has long been understood to play a role in infection-induced angiogenesis when tissues need to repair following infection or inflammation [160]. For example, angiogenesis can be driven by TLR4, as TLR4 stimulated endothelial cells produce VEGF [161, 162]. Consistent with this proangiogenic role of TLR4, an animal model of neovascularization (oxygen-induced retinopathy) was shown to have less neovascularisation, VEGF production and pro-inflammatory mediators IL-1β and NF- κβ production in TLR4 knockdown animals [163]. Although in the anterior eye, the DAMP High-Mobility Group Box-1 (HMGB1) has directly been linked to angiogenesis via TLR4 [164]. Inhibition of TLR4 co-receptor myeloid differentiation protein 2 (MD2) reduced laser induced CNV lesions and accompanying inflammation [165]. TLR4 signalling can trigger myeloid reprogramming to favour a proinflammatory phenotype that worsens retinal angiogenesis (laser induced CNV) in mice [166]. These studies suggest that inhibition of TLR4 could reduce inflammation, alter innate immune cell responses, and ultimately suppress angiogenesis in nAMD.

In summary, TLRs can be stimulated by DAMPs linked to AMD such as Alu RNA and tenascin-C. TLR2 and TLR3 are highly expressed by the RPE, and their expression increase in animal models of retinal degeneration and early and intermediate AMD. Targeting TLR2 and TLR4 has shown to be effective in reducing neovascularization and other models of retinal angiogenesis and these offer the most promising in slowing the progression of nAMD.

The contribution of the NLRP3 inflammasome to AMD

The NLRP3 inflammasome is a host defence response that assists in clearing bacterial, viral, and fungal infections via pyroptosis, a form of programmed cell death. The NLRP3 inflammasome is of interest due to its ability to damage the outer retina when activated by endogenous proteins and intracellular DNA [121]. There have been no genetic risk studies that link polymorphisms of NLRP3 with AMD risk. This section will describe the NLRP3 expression, DAMP stimuli and NLRP3 signalling outcomes reported in preclinical and human studies of AMD eyes.

The NLRP3 inflammasome has been implicated in AMD due to the detection of NLRP3 proteins in the retina, eye, and blood. Drusen, a characteristic of AMD, has been found to contain inflammasome stimulating proteins (NLRP3) and elevated levels of inflammasome processed cytokines (IL-1β and IL-18) [97, 121, 167]. Drusen and drusen components, such as C1q, have been shown to cause NLRP3 activation and secretion of IL-1β and IL-18 from peripheral blood mononuclear cells [168]. The drusen component, Aβ-oligomer, has also been reported to stimulate the NLRP3 inflammasome and induce RPE degeneration by multiple research groups [45,46,47]. Later studies have shown NLRP3 inflammasome formation in the retina due to signalling via the P2 × 7 receptor [169, 170]. Cytokines that can be produced via NLRP3 inflammasome (but also through other immune regulatory pathways) have been detected at higher levels in AMD patients, in the serum (IL-1β, IL-6), the plasma (TNF-α, IL-6) and vitreous (IL-1β) compared to age-matched controls [171,172,173,174]. A recent retinal single-cell RNAseq study found enrichment of the Nod-like signalling pathway in advanced AMD eyes compared to controls for NLRP3 genes and P2 × 7 [175]. These studies suggest that NLRP3 inflammasome activation may be a systemic indicator of disease and a local mediator of damage in the eye.

A wide range of stimuli activates the NLRP3 inflammasome. The most well understood signalling events are reactive oxidative species (ROS), ionic flux responding to extracellular ATP, and lysosomal damage [176]. There is evidence that these stimuli, and others, are present in AMD and in vitro studies have shown that NLRP3 stimulation damages RPE cells [47, 97]. Reactive oxidative species (ROS) are a well-described trigger and regulate NLRP3 formation [177]. ROS has long been implicated in the pathogenesis of AMD and ROS has been shown to directly activate and mediate the NLRP3 inflammasome in the RPE [46, 178, 179]. Extracellular ATP, released by damaged cells or present in the serum of nAMD patients, is a potent activator of the NLRP3 inflammasome [49]. The priming step for P2 × 7 receptor mediated NLRP3 inflammasome formation is high concentrations of extracellular ATP that causes the P2 × 7 receptor to open, allowing K + efflux and this change in cellular osmolarity triggers the NLRP3 inflammasome [180, 181]. In the retina, extracellular ATP has been shown to trigger the NLRP3 inflammasome via the release of ATP from dying cells or in response to mechanical strain [119, 182]. In mechanical strain studies, where intraocular pressure is elevated, it has been shown that retinal astrocytes and microglia activate NLRP3 via P2 × 7 stimulation [120, 179]. The NLRP3 inflammasome activation via the P2 × 7 receptor results in the maturation of IL-1β and consequently increases pro-inflammatory cytokine and chemokine release such as IL-6 in the retina and TNF-α and Chemokine (C-C motif) ligands CCL2 and CCL3 in cultured microglia [183,184,185,186].

The lysosome ruptured process, where phagolysosomes burst and lysosome enzymes such as cathepsin activate the NLRP3 inflammasome, has also been implicated in activating an RPE-driven inflammasome response [40, 187, 188]. The RPE engulfs and phagocytoses the outer segments of the photoreceptors daily in a process that renews photoreceptor function. These engulfed photoreceptor outer segments are combined with lysosomes, to form phagolysosomes as part of this process. In vitro studies of RPE cells have shown that phagolysosomes can burst, stimulating the NLRP3 inflammasome, activating caspase-1 and secretion of IL-1β and IL-18 from the RPE [100, 121, 189].

Double-stranded RNA (dsRNA) has also stimulated the NLRP3 inflammasome in AMD [38]. Endogenous dsRNA was studied in AMD due to studies in mice that showed synthetic dsRNA induced outer retina damage akin to geographic atrophy [190]. Endogenous dsRNA was unexpectedly expressed in the human retina, where copious amounts were detected in the RPE and drusen of AMD eyes with little to none detected in normal aged eyes [38]. The dsRNA was extracted and amplified where it was discovered that the amplicons were Alu fragments, a retrotransposon. Alu RNA is dsRNA encoded by small repetitive Alu elements in the genome, likely derived from parasitic DNA [191]. High expression levels of Alu RNA found within tissues have been associated with human disease [192]. In vitro studies in mice found Alu RNA to induce RPE cell death by activating the NLRP3 inflammasome in the RPE [97, 99]. These studies showed that by preventing the activation of NLRP3 inflammasome, the RPE was protected from damage in multiple animal models of AMD and against sterile inflammation damage in mice [193]. Subsequent studies have shown that Alu RNA can trigger RPE cell death through non-canonical NLRP3 inflammasome signalling via gasdermin D (GSDMD) activation [47, 194, 195].

The NLRP3 inflammasome can be activated via canonical signalling involving caspase-1 and non-canonically via caspase-4/caspase-5 (caspase-11 in mice). Non-canonical NLRP3 inflammasome formation can be triggered by intracellular detection of either LPS by caspase-4/caspase-5 or foreign DNA via cGAS (cyclic GMP–AMP synthase) [57]. Intracellular stimulation by either caspase-4/caspase-5 or cGAS results in activation of gasdermin D, which can form pores on the cell surface [57]. The presence of a pore allows for passive transport of ions and molecules that cause the cell to swell and die, in a form of cell death termed pyroptosis. The presence of GSDMD pores causes secondary activation of the NLRP3 inflammasome leading to release of pro-inflammatory cytokines, IL-18 and IL-1β [196]. GSDMS can also be triggered by cGAS-STING (signalling a second messenger stimulator of interferon genes). cGAS is a cytosolic DNA sensor that can detect foreign DNA, and damaged mitochondrial DNA (mtDNA), that activates STING [197]. While many studies have observed mtDNA damage in AMD lesions, recently, the cGAS-STING pathway has been linked to RPE damage [194, 195, 198]. RPE and photoreceptor death are known to be associated with NLRP3 inflammasome mediated pyropotosis due to GSDMD activation and cGAS signalling [47, 199, 200]. The later studies on RPE degeneration due to AluRNA proposes that AluRNA accumulation causes mtDNA damage and release into the cytosol that triggers non-canonical NLRP3 signalling via gasdermin [47, 194, 195]. This means for NLRP3 inflammasome formation, AluRNA can stimulate either through canonical or non-canonical signalling to bring about RPE degeneration.

Of the many triggers for the NLRP3 inflammasome, activation of the P2 × 7 receptor has consistently been shown to be integral to mediating RPE cell death via NLRP3 in canonical and non-canonical pathways [193, 194]. A schematic of P2 × 7 and NLRP3 inflammasome independent and dependent signalling pathways leading to RPE degeneration and pyroptosis are depicted in Fig. 3.

Fig. 3
figure 3

NLRP3 activation via P2 × 7 signalling within RPE and macrophages. A schematic depicting the cell signalling pathways that leads to RPE degeneration or pyroptosis. In either immune cell (e.g., macrophage) or RPE, multiple stimuli such as Alu RNA, ROS or extracellular ATP can stimulate NLRP3 inflammasome. In canonical stimulation leading to RPE degeneration, NLRP3 is activated and, co-activates the P2 × 7 receptor. Alu RNA causes upregulation of interferon type 1 and NF-κB regulated cytokines (e.g. pro-IL-18). Assembly of the NLRP3 following either ATP, Alu RNA or ROS stimulation allows for processing of pro-interleukins into mature IL-1β and IL-18. Microvesicles can release IL-1β to stimulate inflammatory responses further. IL-18 binds to IL-18 receptor to induce RPE degeneration via MyD88 signalling. In non-canonical stimulation, RPE degeneration occurs due to Alu RNA stimulation of GSDMD via cGAS-STING signalling. The activation of GSDMD results in GSDMD pore formation that causes pyroptosis cell death in RPE cells and secondary activation of the NLRP3 inflammasome. Black arrows indicate a known signalling step. Dashed arrow steps indicate postulated signalling step

There has been some controversy about whether NLRP3 inflammasome formation occurs within the RPE or from other cells within the retina. Some studies implicate the RPE directly [38, 97, 121, 192]. Others have shown that NLRP3 activation in immune cells mediates damage to the RPE [201,202,203]. DAMP stimuli released by the RPE may lead to stimulation of immune cell based NLRP3 inflammasomes or that transport of inflammasome components from the RPE may occur. For example, it has been reported that in cultured RPE, extracellular vesicles that contain inflammasome components (e.g., caspase-1) are secreted [204]. These vesicles could be a method of communication between the RPE and immune cells to initiate the formation of the NLRP3 inflammasome. In co-culturing studies combining RPE with macrophages, it has been confirmed that damaged RPE cells can induce inflammasome activation within macrophages [205]. More work is required to understand the cellular source of the NLRP3-inflammasome processes that contribute to AMD.

In summary, the NLRP3 has been shown to mediate RPE cell damage and is activated by numerous damaging stimuli in the retina. The classic stimuli that activate NLRP3 are high extracellular ATP, which stimulates the P2 × 7 receptor, ROS, and the release of cathepsin following phagosome bursting [40, 119, 178, 182, 187, 188]. Activation of the NLRP3 has been shown to release pro-inflammatory cytokines and damage the RPE [45,46,47, 179]. Damaging stimuli found in AMD eyes, Aβ-oligomers in drusen and Alu RNA in the RPE have both been shown to be potent stimulators of the NLRP3 inflammasome via the P2 × 7 receptor [97, 169, 170]. Activation of NLRP3 inflammasome by these stimuli results (e.g. Aβ-oligomers or Alu RNA) damages RPE cells [47, 97]. Subsequent studies have reported that Alu RNA can affect the outer retina via mechanisms involving non-canonical inflammosome formation via GSDMD [47, 194]. There is some evidence of NLRP3 inflammasome-dependent pyroptosis in RPE and photoreceptors mediated by GSDMD activation [47, 200]. A potential therapy for preventing RPE damage in AMD could involve inhibiting the NRLP3 inflammasome with anti-retroviral agents, as demonstrated in a mouse model of GA where Aβ-oligomers were injected to injure RPE [169]. Alternatively, blockade of NLRP3 inflammasome activation by targeting either intracellular adaptor proteins (i.e. MyD88) or GSDMD a pyroptosis mediator may have utility in reducing RPE loss in AMD [200, 206, 207].

Conclusion

Over the past 20 years there has been increasing evidence for the role of innate immunity in the development of AMD. In particular, the mechanisms by which activation of pattern recognition receptors and their effectors contribute to cell loss and dysfunction in AMD have emerged. As mediators of innate immune activation, PRRs are unique, in that both signals from foreign infectious material and endogenous cellular damage activate them. The outer retina expresses high levels of PRRs, particularly TLRs on the RPE and NLRP3 and TLRs on mononuclear phagocytes that appear at all stages of AMD. In AMD, numerous DAMP signals are present in drusen and the RPE including Alu RNA, Aβ-oligomers, tenascin, ROS and cathepsin that can drive para-inflammation by stimulating PRRs. The ability of the innate immune system to auto-activate by DAMPs can initially be a beneficial response to remove damaged cells, however prolonged auto-activation initiates a cycle that can lead to chronic para-inflammation that is harmful and may contribute to AMD pathology. In GA, there are localised areas where drusen accumulate and RPE atrophies [3]. At these lesion locations DAMP stimuli are present and could activate PRR that then potentiate pro-inflammatory signalling to damage the surrounding tissue. Para-inflammation could also by suppressed by targeting either the PRR stimuli found in drusen (e.g. neutralising Aβ-oligomer), the PRR themselves (e.g. antagonists of TLRs or NLRP3) or their intracellular adaptor proteins (e.g. MyD88). A reduction in para-inflammation, and PRR activation has the potential to preserve the surrounding RPE and prevent further photoreceptor loss. In the case of nAMD, targeting TLR2/4 and it’s adaptor protein MyD88 have shown the most promise in reducing inflammation as well as angiogenesis in preclinical studies. By targeting the innate immune systems’ pattern recognition receptors it may be possible to target the underlying chronic inflammation that occurs in those with AMD, thus preventing further loss of RPE and photoreceptors.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ATP:

Adenosine triphosphate

BM:

Bruch’s membrane

C3:

Complement component 3

CC:

Choroidal capillaries

CCL2:

Chemokine (C-C motif) ligand 2

CFB:

Complement factor B

CFH:

Complement factor H

CFI:

Complement factor I

cGAS:

cyclic GMP-AMP synthase

cGAS-STING:

cGAS-Stimulator of interferon genes

DAMPs:

Damage-associated molecular patterns

dsRNA:

double stranded RNA

ECM:

Extracellular matrix

GA:

Geographic atrophy

GSDMD:

Gasdermin D

HMGB:

High mobility group box

IFN:

Interferon

IFANR:

Type I interferon receptor

IL:

Interleukins

INL:

Inner nuclear layer

IS:

Inner segments of photoreceptors

LPS:

Lipopolysaccharide

Myd88:

Myeloid differentiation primary response 88

mtDNA:

Mitochondrial DNA

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

NLR:

NOD-like receptor

NLRP3:

NLR family pyrin domain containing 3

OS:

Outer segments of photoreceptors

P2 × 7:

P2X purinoceptor 7

PAMPs:

Pathogen-associated molecular patterns

PBMC:

Peripheral blood monocytes

PR:

Photoreceptors

RIG:

Retinoic acid-inducible gene

RLR:

RIG-I-like receptors

ROS:

Reactive oxidative species

RPE:

Retinal pigment epithelium

SNP:

Single nucleotide polymorphism

SR:

Subretinal layer

ssRNA:

Single-strand RNA

TLR:

Toll-like receptor

TNFα:

Tumor necrosis factor

VGLUT:

Vesicular glutamate transporter 1

References

  1. Flaxman SR, Bourne RRA, Resnikoff S, Ackland P, Braithwaite T, Cicinelli MV, et al. Global causes of blindness and distance vision impairment 1990–2020: a systematic review and meta-analysis. Lancet Global Health. 2017;5(12):e1221–e34.

    Article  PubMed  Google Scholar 

  2. Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng CY, et al. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Global Health. 2014;2(2):e106–16.

    Article  PubMed  Google Scholar 

  3. Guymer RH, Campbell TG. Age-related macular degeneration. Lancet. 2023;401(10386):1459–72.

    Article  CAS  PubMed  Google Scholar 

  4. Solomon SD, Lindsley K, Vedula SS, Krzystolik MG, Hawkins BS. Anti-vascular endothelial growth factor for neovascular age-related macular degeneration. Cochrane Database Syst Rev. 2014;8(8):Cd005139.

    PubMed  Google Scholar 

  5. Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK, Chung CY, et al. Ranibizumab for neovascular age-related macular degeneration. N Engl J Med. 2006;355(14):1419–31.

    Article  CAS  PubMed  Google Scholar 

  6. Heier JS, Khanani AM, Quezada Ruiz C, Basu K, Ferrone PJ, Brittain C, et al. Efficacy, durability, and safety of intravitreal faricimab up to every 16 weeks for neovascular age-related macular degeneration (TENAYA and LUCERNE): two randomised, double-masked, phase 3, non-inferiority trials. Lancet (London England). 2022;399(10326):729–40.

    Article  CAS  PubMed  Google Scholar 

  7. Rofagha S, Bhisitkul RB, Boyer DS, Sadda SR, Zhang K. Seven-year outcomes in ranibizumab-treated patients in ANCHOR, MARINA, and HORIZON: a multicenter cohort study (SEVEN-UP). Ophthalmology. 2013;120(11):2292–9.

    Article  PubMed  Google Scholar 

  8. Daniel E, Pan W, Ying G-s, Kim BJ, Grunwald JE, Ferris FL, et al. Development and Course of scars in the comparison of age-related Macular Degeneration treatments trials. Ophthalmology. 2018;125(7):1037–46.

    Article  PubMed  Google Scholar 

  9. Park YG, Park YS, Kim IB. Complement system and potential therapeutics in age-related Macular Degeneration. Int J Mol Sci. 2021;22(13).

  10. DeMaio A, Mehrotra S, Sambamurti K, Husain S. The role of the adaptive immune system and T cell dysfunction in neurodegenerative diseases. J Neuroinflamm. 2022;19(1):251.

    Article  Google Scholar 

  11. Killick J, Morisse G, Sieger D, Astier AL. Complement as a regulator of adaptive immunity. Semin Immunopathol. 2018;40(1):37–48.

    Article  CAS  PubMed  Google Scholar 

  12. Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Prog Retin Eye Res. 2009;28(5):348–68.

    Article  PubMed  Google Scholar 

  13. Nita M, Grzybowski A, Ascaso FJ, Huerva V. Age-related macular degeneration in the aspect of chronic low-grade inflammation (pathophysiological parainflammation). Mediat Inflamm. 2014;2014:930671.

    Article  Google Scholar 

  14. Yates JR, Sepp T, Matharu BK, Khan JC, Thurlby DA, Shahid H, et al. Complement C3 variant and the risk of age-related macular degeneration. N Engl J Med. 2007;357(6):553–61.

    Article  CAS  PubMed  Google Scholar 

  15. Fritsche LG, Igl W, Bailey JN, Grassmann F, Sengupta S, Bragg-Gresham JL, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet. 2016;48(2):134–43.

    Article  CAS  PubMed  Google Scholar 

  16. Magnusson KP, Duan S, Sigurdsson H, Petursson H, Yang Z, Zhao Y, et al. CFH Y402H confers similar risk of soft drusen and both forms of advanced AMD. PLoS Med. 2006;3(1):e5.

    Article  PubMed  Google Scholar 

  17. Nozaki M, Raisler BJ, Sakurai E, Sarma JV, Barnum SR, Lambris JD, et al. Drusen complement components C3a and C5a promote choroidal neovascularization. Proc Natl Acad Sci USA. 2006;103(7):2328–33.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Anderson DH, Mullins RF, Hageman GS, Johnson LV. A role for local inflammation in the formation of drusen in the aging eye. Am J Ophthalmol. 2002;134(3):411–31.

    Article  CAS  PubMed  Google Scholar 

  19. Johnson LV, Ozaki S, Staples MK, Erickson PA, Anderson DH. A potential role for Immune Complex Pathogenesis in Drusen formation. Exp Eye Res. 2000;70(4):441–9.

    Article  CAS  PubMed  Google Scholar 

  20. Crabb JW. The proteomics of drusen. Cold Spring Harbor Perspect Med. 2014;4(7):a017194.

    Article  Google Scholar 

  21. Nittala MG, Metlapally R, Ip M, Chakravarthy U, Holz FG, Staurenghi G, et al. Association of Pegcetacoplan with Progression of Incomplete retinal pigment epithelium and outer retinal atrophy in Age-Related Macular Degeneration: a Post Hoc Analysis of the FILLY Randomized Clinical Trial. JAMA Ophthalmol. 2022;140(3):243–9.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Goldberg R, Heier JS, Wykoff CC, Staurenghi G, Singh RP, Steinle N, et al. Efficacy of intravitreal pegcetacoplan in patients with geographic atrophy (GA): 12-month results from the phase 3 OAKS and DERBY studies. Investig Ophthalmol Vis Sci. 2022;63(7):1500.

    Google Scholar 

  23. Guillonneau X, Eandi CM, Paques M, Sahel JA, Sapieha P, Sennlaub F. On phagocytes and macular degeneration. Prog Retin Eye Res. 2017;61:98–128.

    Article  CAS  PubMed  Google Scholar 

  24. Grossniklaus HE, Ling JX, Wallace TM, Dithmar S, Lawson DH, Cohen C, et al. Macrophage and retinal pigment epithelium expression of angiogenic cytokines in choroidal neovascularization. Mol Vis. 2002;8:119–26.

    CAS  PubMed  Google Scholar 

  25. Grossniklaus HE, Martinez JA, Brown VB, Lambert HM, Sternberg P Jr., Capone A Jr., et al. Immunohistochemical and histochemical properties of surgically excised subretinal neovascular membranes in age-related macular degeneration. Am J Ophthalmol. 1992;114(4):464–72.

    Article  CAS  PubMed  Google Scholar 

  26. Combadière C, Feumi C, Raoul W, Keller N, Rodéro M, Pézard A, et al. CX3CR1-dependent subretinal microglia cell accumulation is associated with cardinal features of age-related macular degeneration. J Clin Investig. 2007;117(10):2920–8.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Gupta N, Brown KE, Milam AH. Activated microglia in human retinitis pigmentosa, late-onset retinal degeneration, and age-related macular degeneration. Exp Eye Res. 2003;76(4):463–71.

    Article  CAS  PubMed  Google Scholar 

  28. Lad EM, Cousins SW, Van Arnam JS, Proia AD. Abundance of infiltrating CD163 + cells in the retina of postmortem eyes with dry and neovascular age-related macular degeneration. Graefe’s archive for clinical and experimental ophthalmology = Albrecht von Graefes Archiv fur klinische und experimentelle Ophthalmologie. 2015;253(11):1941–5.

  29. Cherepanoff S, McMenamin P, Gillies MC, Kettle E, Sarks SH. Bruch’s membrane and choroidal macrophages in early and advanced age-related macular degeneration. Br J Ophthalmol. 2010;94(7):918–25.

    Article  CAS  PubMed  Google Scholar 

  30. McLeod DS, Bhutto I, Edwards MM, Silver RE, Seddon JM, Lutty GA. Distribution and quantification of Choroidal macrophages in human eyes with age-related Macular Degeneration. Invest Ophthalmol Vis Sci. 2016;57(14):5843–55.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Greferath U, Guymer RH, Vessey KA, Brassington K, Fletcher EL. Correlation of Histologic Features with in vivo imaging of reticular pseudodrusen. Ophthalmology. 2016;123(6):1320–31.

    Article  PubMed  Google Scholar 

  32. Hagbi-Levi S, Grunin M, Jaouni T, Tiosano L, Rinsky B, Elbaz-Hayoun S, et al. Proangiogenic characteristics of activated macrophages from patients with age-related macular degeneration. Neurobiol Aging. 2017;51:71–82.

    Article  CAS  PubMed  Google Scholar 

  33. Sennlaub F, Auvynet C, Calippe B, Lavalette S, Poupel L, Hu SJ, et al. CCR2(+) monocytes infiltrate atrophic lesions in age-related macular disease and mediate photoreceptor degeneration in experimental subretinal inflammation in Cx3cr1 deficient mice. EMBO Mol Med. 2013;5(11):1775–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kaarniranta K, Salminen A. Age-related macular degeneration: activation of innate immunity system via pattern recognition receptors. J Mol Med. 2009;87(2):117–23.

    Article  CAS  PubMed  Google Scholar 

  35. Winkler BS, Boulton ME, Gottsch JD, Sternberg P. Oxidative damage and age-related macular degeneration. Mol Vis. 1999;5:32.

    CAS  PubMed  Google Scholar 

  36. Ishibashi T, Murata T, Hangai M, Nagai R, Horiuchi S, Lopez P, et al. Advanced glycation end products in age-related macular degeneration. Archives Ophthalmol (Chicago Ill: 1960). 1998;116(12):1629–32.

    Article  CAS  Google Scholar 

  37. Golestaneh N, Chu Y, Xiao Y-Y, Stoleru GL, Theos AC. Dysfunctional autophagy in RPE, a contributing factor in age-related macular degeneration. Cell Death Dis. 2018;8(1):e2537–e.

    Article  Google Scholar 

  38. Kaneko H, Dridi S, Tarallo V, Gelfand BD, Fowler BJ, Cho WG, et al. DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration. Nature. 2011;471(7338):325–30.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Karunadharma PP, Nordgaard CL, Olsen TW, Ferrington DA. Mitochondrial DNA damage as a potential mechanism for age-related macular degeneration. Invest Ophthalmol Vis Sci. 2010;51(11):5470–9.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol. 2008;9(8):857–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cao X. Self-regulation and cross-regulation of pattern-recognition receptor signalling in health and disease. Nat Rev Immunol. 2016;16(1):35–50.

    Article  CAS  PubMed  Google Scholar 

  42. Liu-Bryan R. Synovium and the innate inflammatory network in osteoarthritis progression. Curr Rheumatol Rep. 2013;15(5):323.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Hanus J, Anderson C, Sarraf D, Ma J, Wang S. Retinal pigment epithelial cell necroptosis in response to sodium iodate. Cell Death Discovery. 2016;2(1):16054.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Feng L, Ju M, Lee KYV, Mackey A, Evangelista M, Iwata D, et al. A proinflammatory function of toll-like receptor 2 in the retinal pigment epithelium as a Novel Target for reducing Choroidal Neovascularization in Age-Related Macular Degeneration. Am J Pathol. 2017;187(10):2208–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Anderson DH, Talaga KC, Rivest AJ, Barron E, Hageman GS, Johnson LV. Characterization of beta amyloid assemblies in drusen: the deposits associated with aging and age-related macular degeneration. Exp Eye Res. 2004;78(2):243–56.

    Article  CAS  PubMed  Google Scholar 

  46. Wang K, Yao Y, Zhu X, Zhang K, Zhou F, Zhu L. Amyloid β induces NLRP3 inflammasome activation in retinal pigment epithelial cells via NADPH oxidase- and mitochondria-dependent ROS production. J Biochem Mol Toxicol. 2017;31(6).

  47. Gao J, Cui JZ, To E, Cao S, Matsubara JA. Evidence for the activation of pyroptotic and apoptotic pathways in RPE cells associated with NLRP3 inflammasome in the rodent eye. J Neuroinflamm. 2018;15(1):15.

    Article  Google Scholar 

  48. Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20(2):95–112.

    Article  CAS  PubMed  Google Scholar 

  49. Di Virgilio F, Dal Ben D, Sarti AC, Giuliani AL, Falzoni S. The P2X7 Receptor in Infection and Inflammation. Immunity. 2017;47(1):15–31.

    Article  PubMed  Google Scholar 

  50. Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol. 2014;5.

  51. O’Neill LA, Bowie AG. The family of five: TIR-domain-containing adaptors in toll-like receptor signalling. Nat Rev Immunol. 2007;7(5):353–64.

    Article  PubMed  Google Scholar 

  52. Gay NJ, Symmons MF, Gangloff M, Bryant CE. Assembly and localization of toll-like receptor signalling complexes. Nat Rev Immunol. 2014;14(8):546–58.

    Article  CAS  PubMed  Google Scholar 

  53. He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41(12):1012–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Swanson KV, Deng M, Ting JPY. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liu RT, Gao J, Cao S, Sandhu N, Cui JZ, Chou CL, et al. Inflammatory Mediators Induced by amyloid-Beta in the Retina and RPE in vivo: implications for Inflammasome activation in Age-Related Macular Degeneration. Investig Ophthalmol Vis Sci. 2013;54(3):2225–37.

    Article  Google Scholar 

  56. Pelegrin P. P2X7 receptor and the NLRP3 inflammasome: Partners in crime. Biochem Pharmacol. 2021;187:114385.

    Article  CAS  PubMed  Google Scholar 

  57. Downs KP, Nguyen H, Dorfleutner A, Stehlik C. An overview of the non-canonical inflammasome. Mol Aspects Med. 2020;76:100924.

    Article  CAS  PubMed  Google Scholar 

  58. Rodríguez-Martínez S, Cancino-Díaz ME, Jiménez-Zamudio L, García-Latorre E, Cancino-Díaz JC. TLRs and NODs mRNA expression pattern in healthy mouse eye. Br J Ophthalmol. 2005;89(7):904–10.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Takeda K, Akira S. Toll-like receptors in innate immunity. Int Immunol. 2005;17(1):1–14.

    Article  CAS  PubMed  Google Scholar 

  60. Chang JH, McCluskey P, Wakefield D. Expression of toll-like receptor 4 and its associated lipopolysaccharide receptor complex by resident antigen-presenting cells in the human uvea. Invest Ophthalmol Vis Sci. 2004;45(6):1871–8.

    Article  PubMed  Google Scholar 

  61. Kumar MV, Nagineni CN, Chin MS, Hooks JJ, Detrick B. Innate immunity in the retina: toll-like receptor (TLR) signaling in human retinal pigment epithelial cells. J Neuroimmunol. 2004;153(1–2):7–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kauppinen A, Niskanen H, Suuronen T, Kinnunen K, Salminen A, Kaarniranta K. Oxidative stress activates NLRP3 inflammasomes in ARPE-19 cells–implications for age-related macular degeneration (AMD). Immunol Lett. 2012;147(1–2):29–33.

    Article  CAS  PubMed  Google Scholar 

  63. Kumar A, Shamsuddin N. Retinal Muller Glia initiate Innate Response to Infectious Stimuli via Toll-Like receptor signaling. PLoS ONE. 2012;7(1):e29830.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  64. Lee MT, Hooper LC, Kump L, Hayashi K, Nussenblatt R, Hooks JJ, et al. Interferon-beta and adhesion molecules (E-selectin and s-intracellular adhesion molecule-1) are detected in sera from patients with retinal vasculitis and are induced in retinal vascular endothelial cells by toll-like receptor 3 signalling. Clin Exp Immunol. 2007;147(1):71–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Stewart EA, Wei R, Branch MJ, Sidney LE, Amoaku WM. Expression of toll-like receptors in human retinal and choroidal vascular endothelial cells. Exp Eye Res. 2015;138:114–23.

    Article  CAS  PubMed  Google Scholar 

  66. Noailles A, Kutsyr O, Maneu V, Ortuño-Lizarán I, Campello L, de Juan E, et al. The absence of toll-like receptor 4 mildly affects the structure and function in the Adult Mouse Retina. Front Cell Neurosci. 2019;13:59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Forrester JV, Xu H. Good news–bad news: the Yin and Yang of immune privilege in the eye. Front Immunol. 2012;3.

  68. da Silva EZ, Jamur MC, Oliver C. Mast cell function: a new vision of an old cell. J Histochem Cytochemistry: Official J Histochem Soc. 2014;62(10):698–738.

    Article  Google Scholar 

  69. Ferreira I, Liberal J, Martins JD, Silva A, Neves BM, Cruz MT. Inflammasome in dendritic cells immunobiology: implications to diseases and therapeutic strategies. Curr Drug Targets. 2017;18(9):1003–18.

    PubMed  Google Scholar 

  70. Chen W, Hu X, Zhao L, Li S, Lu H. Expression of toll-like receptor 4 in uvea-resident tissue macrophages during endotoxin-induced uveitis. Mol Vis. 2009;15:619–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Aluri J, Cooper MA, Schuettpelz LG. Toll-like receptor signaling in the establishment and function of the Immune System. Cells. 2021;10(6).

  72. Yu F-SX, Hazlett LD. Toll-like receptors and the Eye. Investig Ophthalmol Vis Sci. 2006;47(4):1255–63.

    Article  Google Scholar 

  73. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Volume 330. New York, NY: Science; 2010. pp. 841–5. 6005.

    Google Scholar 

  74. Dixon MA, Greferath U, Fletcher EL, Jobling AI. The contribution of Microglia to the development and maturation of the Visual System. Front Cell Neurosci. 2021;15.

  75. Todd L, Palazzo I, Suarez L, Liu X, Volkov L, Hoang TV, et al. Reactive microglia and IL1β/IL-1R1-signaling mediate neuroprotection in excitotoxin-damaged mouse retina. J Neuroinflamm. 2019;16(1):118.

    Article  Google Scholar 

  76. Kochan T, Singla A, Tosi J, Kumar A. Toll-like receptor 2 ligand pretreatment attenuates retinal microglial inflammatory response but enhances phagocytic activity toward Staphylococcus aureus. Infect Immun. 2012;80(6):2076–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Luo C, Yang X, Kain AD, Powell DW, Kuehn MH, Tezel G. Glaucomatous tissue stress and the regulation of immune response through glial toll-like receptor signaling. Invest Ophthalmol Vis Sci. 2010;51(11):5697–707.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Huang Z, Zhou T, Sun X, Zheng Y, Cheng B, Li M, et al. Necroptosis in microglia contributes to neuroinflammation and retinal degeneration through TLR4 activation. Cell Death Differ. 2018;25(1):180–9.

    Article  CAS  PubMed  Google Scholar 

  79. Halder SK, Matsunaga H, Ishii KJ, Akira S, Miyake K, Ueda H. Retinal cell type-specific prevention of ischemia-induced damages by LPS-TLR4 signaling through microglia. J Neurochem. 2013;126(2):243–60.

    Article  CAS  PubMed  Google Scholar 

  80. Ghosh F, Abdshill H, Arnér K, Voss U, Taylor L. Retinal neuroinflammatory induced neuronal degeneration - role of toll-like receptor-4 and relationship with gliosis. Exp Eye Res. 2018;169:99–110.

    Article  CAS  PubMed  Google Scholar 

  81. Zhang Y, Xu Y, Sun Q, Xue S, Guan H, Ji M. Activation of P2X(7)R- NLRP3 pathway in Retinal microglia contribute to Retinal Ganglion Cells death in chronic ocular hypertension (COH). Exp Eye Res. 2019;188:107771.

    Article  CAS  PubMed  Google Scholar 

  82. Mai K, Chui JJY, Di Girolamo N, McCluskey PJ, Wakefield D. Role of toll-like receptors in human iris pigment epithelial cells and their response to pathogen-associated molecular patterns. J Inflamm. 2014;11(1):20.

    Article  Google Scholar 

  83. Lo S-M, Hwang Y-S, Liu C-L, Shen C-N, Hong W-H, Yang W-C et al. Inhibiting TLR7 expression in the retinal pigment epithelium suppresses experimental autoimmune uveitis. Front Immunol. 2022;12.

  84. Mulfaul K, Ozaki E, Fernando N, Brennan K, Chirco KR, Connolly E, et al. Toll-like receptor 2 facilitates oxidative damage-Induced Retinal Degeneration. Cell Rep. 2020;30(7):2209–24e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Singh PK, Guest JM, Kanwar M, Boss J, Gao N, Juzych MS, et al. Zika virus infects cells lining the blood-retinal barrier and causes chorioretinal atrophy in mouse eyes. JCI Insight. 2017;2(4):e92340.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Choi SJLK-H, Park SJ, Park HS, Kim J, Kim S-K. The expression pattern of toll-like receptor (TLR) and cytokine production to TLR agonists in human retinal pigment epithelial cells. J Bacteriol Virol. 2007;37(2):119–28.

    Article  CAS  Google Scholar 

  87. Terheyden L, Roider J, Klettner A. Basolateral activation with TLR agonists induces polarized cytokine release and reduces barrier function in RPE in vitro. Graefe’s Archive Clin Experimental Ophthalmol = Albrecht Von Graefes Archiv fur Klinische und Experimentelle Ophthalmologie. 2021;259(2):413–24.

    Article  CAS  Google Scholar 

  88. Elner VM, Elner SG, Bian ZM, Kindezelskii AL, Yoshida A, Petty HR. RPE CD14 immunohistochemical, genetic, and functional expression. Exp Eye Res. 2003;76(3):321–31.

    Article  CAS  PubMed  Google Scholar 

  89. Elner SG, Petty HR, Elner VM, Yoshida A, Bian ZM, Yang D et al. TLR4 mediates human retinal pigment epithelial endotoxin binding and cytokine expression. Trans Am Ophthalmol Soc. 2005;103:126 – 35; discussion 35 – 7.

  90. Kindzelskii AL, Elner VM, Elner SG, Yang D, Hughes BA, Petty HR. Toll-like receptor 4 (TLR4) of retinal pigment epithelial cells participates in transmembrane signaling in response to photoreceptor outer segments. J Gen Physiol. 2004;124(2):139–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Collin J, Hasoon MSR, Zerti D, Hammadi S, Dorgau B, Clarke L, et al. Single-cell RNA sequencing reveals transcriptional changes of human choroidal and retinal pigment epithelium cells during fetal development, in healthy adult and intermediate age-related macular degeneration. Hum Mol Genet. 2023;32(10):1698–710.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Voigt AP, Mulfaul K, Mullin NK, Flamme-Wiese MJ, Giacalone JC, Stone EM, et al. Single-cell transcriptomics of the human retinal pigment epithelium and choroid in health and macular degeneration. Proc Natl Acad Sci USA. 2019;116(48):24100–7.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  93. Kosmidou C, Efstathiou NE, Hoang MV, Notomi S, Konstantinou EK, Hirano M, et al. Issues with the specificity of Immunological reagents for NLRP3: implications for age-related Macular Degeneration. Sci Rep. 2018;8(1):461.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  94. Shi G, Chen S, Wandu WS, Ogbeifun O, Nugent LF, Maminishkis A, et al. Inflammasomes Induced by 7-Ketocholesterol and other Stimuli in RPE and in bone marrow-derived cells Differ markedly in their production of IL-1β and IL-18. Invest Ophthalmol Vis Sci. 2015;56(3):1658–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Piippo N, Korhonen E, Hytti M, Kinnunen K, Kaarniranta K, Kauppinen A. Oxidative stress is the principal contributor to Inflammasome activation in Retinal Pigment Epithelium cells with defunct proteasomes and Autophagy. Cell Physiol Biochemistry: Int J Experimental Cell Physiol Biochem Pharmacol. 2018;49(1):359–67.

    Article  CAS  Google Scholar 

  96. Piippo N, Korhonen E, Hytti M, Skottman H, Kinnunen K, Josifovska N, et al. Hsp90 inhibition as a means to inhibit activation of the NLRP3 inflammasome. Sci Rep. 2018;8(1):6720.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  97. Tarallo V, Hirano Y, Gelfand BD, Dridi S, Kerur N, Kim Y, et al. DICER1 loss and Alu RNA induce age-related macular degeneration via the NLRP3 inflammasome and MyD88. Cell. 2012;149(4):847–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Anderson OA, Finkelstein A, Shima DT. A2E induces IL-1ß production in retinal pigment epithelial cells via the NLRP3 inflammasome. PLoS ONE. 2013;8(6):e67263.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  99. Gelfand BD, Wright CB, Kim Y, Yasuma T, Yasuma R, Li S, et al. Iron Toxicity in the retina requires Alu RNA and the NLRP3 inflammasome. Cell Rep. 2015;11(11):1686–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Brandstetter C, Mohr LK, Latz E, Holz FG, Krohne TU. Light induces NLRP3 inflammasome activation in retinal pigment epithelial cells via lipofuscin-mediated photooxidative damage. J Mol Med. 2015;93(8):905–16.

    Article  CAS  PubMed  Google Scholar 

  101. Schäfer N, Rasras A, Ormenisan DM, Amslinger S, Enzmann V, Jägle H, et al. Complement factor H-Related 3 enhanced inflammation and complement activation in human RPE cells. Front Immunol. 2021;12:769242.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Korhonen E, Piippo N, Hytti M, Hyttinen JMT, Kaarniranta K, Kauppinen A. Only IL-1β release is inflammasome-dependent upon ultraviolet B irradiation although IL-18 is also secreted. FASEB Journal: Official Publication Federation Am Soc Experimental Biology. 2020;34(5):6437–48.

    Article  CAS  Google Scholar 

  103. Alge CS, Hauck SM, Priglinger SG, Kampik A, Ueffing M. Differential protein profiling of primary versus immortalized human RPE cells identifies expression patterns associated with cytoskeletal remodeling and cell survival. J Proteome Res. 2006;5(4):862–78.

    Article  CAS  PubMed  Google Scholar 

  104. Kleinman ME, Yamada K, Takeda A, Chandrasekaran V, Nozaki M, Baffi JZ, et al. Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature. 2008;452(7187):591–7.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  105. Seidel A, Liu L, Jiang Y, Steinle JJ. Loss of TLR4 in endothelial cells but not Müller cells protects the diabetic retina. Exp Eye Res. 2021;206:108557.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Wang L, Wang J, Fang J, Zhou H, Liu X, Su SB. High glucose induces and activates toll-like receptor 4 in endothelial cells of diabetic retinopathy. Diabetol Metab Syndr. 2015;7:89.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Cioanca AV, McCluskey PJ, Eamegdool SS, Madigan MC. Human choroidal melanocytes express functional toll-like receptors (TLRs). Exp Eye Res. 2018;173:73–84.

    Article  CAS  PubMed  Google Scholar 

  108. Hu DN, Zhang R, Iacob CE, Yao S, Yang SF, Chan CC, et al. Toll-like receptor 2 and 6 agonist fibroblast-stimulating lipopeptide increases expression and secretion of CXCL1 and CXCL2 by uveal melanocytes. Exp Eye Res. 2022;216:108943.

    Article  CAS  PubMed  Google Scholar 

  109. Bringmann A, Wiedemann P. Müller glial cells in retinal disease. Ophthalmol J Int D’ophtalmologie Int J Ophthalmol Z fur Augenheilkunde. 2012;227(1):1–19.

    Article  Google Scholar 

  110. Lin X, Fang D, Zhou H, Su SB. The expression of toll-like receptors in murine Müller cells, the glial cells in retina. Neurol Sciences: Official J Italian Neurol Soc Italian Soc Clin Neurophysiol. 2013;34(8):1339–46.

    Article  Google Scholar 

  111. Jiang G, Sun D, Kaplan HJ, Shao H. Retinal astrocytes pretreated with NOD2 and TLR2 ligands activate uveitogenic T cells. PLoS ONE. 2012;7(7):e40510.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  112. Rivera JC, Sitaras N, Noueihed B, Hamel D, Madaan A, Zhou T et al. Microglia and interleukin-1β in ischemic retinopathy elicit microvascular degeneration through neuronal semaphorin-3A. Arteriosclerosis, thrombosis, and vascular biology. 2013;33(8):1881–91.

  113. Pronin A, Pham D, An W, Dvoriantchikova G, Reshetnikova G, Qiao J et al. Inflammasome Activation induces pyroptosis in the retina exposed to ocular hypertension Injury. Front Mol Neurosci. 2019;12.

  114. Qi Y, Zhao M, Bai Y, Huang L, Yu W, Bian Z, et al. Retinal ischemia/reperfusion injury is mediated by toll-like receptor 4 activation of NLRP3 inflammasomes. Invest Ophthalmol Vis Sci. 2014;55(9):5466–75.

    Article  CAS  PubMed  Google Scholar 

  115. Sauter MM, Kolb AW, Brandt CR. Toll-like receptors 4, 5, 6 and 7 are constitutively expressed in non-human primate retinal neurons. J Neuroimmunol. 2018;322:26–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Singh PK, Kumar A. Retinal photoreceptor expresses toll-like receptors (TLRs) and elicits innate responses following TLR Ligand and bacterial challenge. PLoS ONE. 2015;10(3):e0119541.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Gao ML, Wu KC, Deng WL, Lei XL, Xiang L, Zhou GH, et al. Toll-like receptor 3 activation initiates photoreceptor cell death in vivo and in Vitro. Invest Ophthalmol Vis Sci. 2017;58(2):801–11.

    Article  CAS  PubMed  Google Scholar 

  118. Yi H, Patel AK, Sodhi CP, Hackam DJ, Hackam AS. Novel role for the innate immune receptor toll-like receptor 4 (TLR4) in the regulation of the wnt signaling pathway and photoreceptor apoptosis. PLoS ONE. 2012;7(5):e36560.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  119. Viringipurampeer IA, Metcalfe AL, Bashar AE, Sivak O, Yanai A, Mohammadi Z, et al. NLRP3 inflammasome activation drives bystander cone photoreceptor cell death in a P23H rhodopsin model of retinal degeneration. Hum Mol Genet. 2016;25(8):1501–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Albalawi F, Lu W, Beckel JM, Lim JC, McCaughey SA, Mitchell CH. The P2X7 Receptor Primes IL-1β and the NLRP3 Inflammasome in Astrocytes Exposed to Mechanical Strain. Front Cell Neurosci. 2017;11:227.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Tseng WA, Thein T, Kinnunen K, Lashkari K, Gregory MS, D’Amore PA, et al. NLRP3 inflammasome activation in retinal pigment epithelial cells by lysosomal destabilization: implications for age-related macular degeneration. Invest Ophthalmol Vis Sci. 2013;54(1):110–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Jager RD, Mieler WF, Miller JW. Age-related Macular Degeneration. N Engl J Med. 2008;358(24):2606–17.

    Article  CAS  PubMed  Google Scholar 

  123. Güven M, Batar B, Mutlu T, Bostancı M, Mete M, Aras C, et al. Toll-like receptors 2 and 4 polymorphisms in age-related Macular Degeneration. Curr Eye Res. 2016;41(6):856–61.

    Article  PubMed  Google Scholar 

  124. Yang Z, Stratton C, Francis PJ, Kleinman ME, Tan PL, Gibbs D, et al. Toll-like receptor 3 and geographic atrophy in age-related macular degeneration. N Engl J Med. 2008;359(14):1456–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Edwards AO, Chen D, Fridley BL, James KM, Wu Y, Abecasis G, et al. Toll-like receptor polymorphisms and age-related Macular Degeneration. Investig Ophthalmol Vis Sci. 2008;49(4):1652–9.

    Article  Google Scholar 

  126. Bochud PY, Hawn TR, Aderem A. Cutting edge: a toll-like receptor 2 polymorphism that is associated with lepromatous leprosy is unable to mediate mycobacterial signaling. J Immunol (Baltimore Md: 1950). 2003;170(7):3451–4.

    Article  CAS  Google Scholar 

  127. Zhou P, Fan L, Yu KD, Zhao MW, Li XX. Toll-like receptor 3 C1234T may protect against geographic atrophy through decreased dsRNA binding capacity. FASEB Journal: Official Publication Federation Am Soc Experimental Biology. 2011;25(10):3489–95.

    Article  CAS  Google Scholar 

  128. Sharma NK, Sharma K, Gupta A, Prabhakar S, Singh R, Gupta PK et al. Does toll-like receptor-3 (TLR-3) have any role in Indian AMD phenotype? Molecular and cellular biochemistry. 2014;393(1–2):1–8.

  129. Cheng Y, Li MW, Li HP, Zeng WT, Zhou P, Huang LZ, et al. Toll-like receptor 3 polymorphism is not associated with neovascular age-related macular degeneration and polypoidal choroidal vasculopathy in the Chinese. Genet Mol Research: GMR. 2014;13(1):302–9.

    Article  CAS  PubMed  Google Scholar 

  130. Sng CC, Cackett PD, Yeo IY, Thalamuthu A, Venkatraman A, Venkataraman D, et al. Toll-like receptor 3 polymorphism rs3775291 is not associated with choroidal neovascularization or polypoidal choroidal vasculopathy in Chinese subjects. Ophthalmic Res. 2011;45(4):191–6.

    Article  CAS  PubMed  Google Scholar 

  131. Zareparsi S, Buraczynska M, Branham KE, Shah S, Eng D, Li M, et al. Toll-like receptor 4 variant D299G is associated with susceptibility to age-related macular degeneration. Hum Mol Genet. 2005;14(11):1449–55.

    Article  CAS  PubMed  Google Scholar 

  132. Sarli A, Skalidakis I, Velissari A, Koutsandrea C, Stefaniotou M, Petersen MB et al. Investigation of associations of ARMS2, CD14, and TLR4 gene polymorphisms with wet age-related macular degeneration in a Greek population. Clinical ophthalmology (Auckland, NZ). 2017;11:1347–58.

  133. Cho Y, Wang JJ, Chew EY, Ferris FL III, Mitchell P, Chan C-C, et al. Toll-like receptor polymorphisms and age-related Macular Degeneration: replication in three case–control samples. Investig Ophthalmol Vis Sci. 2009;50(12):5614–8.

    Article  Google Scholar 

  134. Despriet DD, Bergen AA, Merriam JE, Zernant J, Barile GR, Smith RT, et al. Comprehensive analysis of the candidate genes CCL2, CCR2, and TLR4 in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2008;49(1):364–71.

    Article  PubMed  Google Scholar 

  135. Kaur I, Hussain A, Hussain N, Das T, Pathangay A, Mathai A, et al. Analysis of CFH, TLR4, and APOE polymorphism in India suggests the Tyr402His variant of CFH to be a global marker for age-related macular degeneration. Invest Ophthalmol Vis Sci. 2006;47(9):3729–35.

    Article  PubMed  Google Scholar 

  136. Roshanipour N, Shahriyari E, Ghaffari Laleh M, Vahedi L, Mirjand Gerami S, Khamaneh A. Associations of TLR4 and IL-8 genes polymorphisms with age-related macular degeneration (AMD): a systematic review and meta-analysis. Ophthalmic Genet. 2021;42(6):641–9.

    Article  CAS  PubMed  Google Scholar 

  137. Vacchelli E, Galluzzi L, Rousseau V, Rigoni A, Tesniere A, Delahaye N, et al. Loss-of-function alleles of P2RX7 and TLR4 fail to affect the response to chemotherapy in non-small cell lung cancer. Oncoimmunology. 2012;1(3):271–8.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Asea A, Rehli M, Kabingu E, Boch JA, Bare O, Auron PE, et al. Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem. 2002;277(17):15028–34.

    Article  CAS  PubMed  Google Scholar 

  139. Park JS, Gamboni-Robertson F, He Q, Svetkauskaite D, Kim JY, Strassheim D, et al. High mobility group box 1 protein interacts with multiple toll-like receptors. Am J Physiol Cell Physiol. 2006;290(3):C917–24.

    Article  CAS  PubMed  Google Scholar 

  140. Midwood K, Sacre S, Piccinini AM, Inglis J, Trebaul A, Chan E, et al. Tenascin-C is an endogenous activator of toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease. Nat Med. 2009;15(7):774–80.

    Article  CAS  PubMed  Google Scholar 

  141. Termeer C, Benedix F, Sleeman J, Fieber C, Voith U, Ahrens T, et al. Oligosaccharides of Hyaluronan activate dendritic cells via toll-like receptor 4. J Exp Med. 2002;195(1):99–111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Johnson GB, Brunn GJ, Kodaira Y, Platt JL. Receptor-mediated monitoring of tissue well-being via detection of soluble heparan sulfate by Toll-like receptor 4. Journal of immunology (Baltimore, Md: 1950). 2002;168(10):5233-9.

  143. Jiang K, Fairless E, Kanda A, Gotoh N, Cogliati T, Li T, et al. Divergent effects of HSP70 overexpression in photoreceptors during inherited retinal degeneration. Invest Ophthalmol Vis Sci. 2020;61(12):25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Arimura N, Ki-i Y, Hashiguchi T, Kawahara K, Biswas KK, Nakamura M, et al. Intraocular expression and release of high-mobility group box 1 protein in retinal detachment. Lab Invest. 2009;89(3):278–89.

    Article  CAS  PubMed  Google Scholar 

  145. Kobayashi Y, Yoshida S, Zhou Y, Nakama T, Ishikawa K, Kubo Y, et al. Tenascin-C secreted by transdifferentiated retinal pigment epithelial cells promotes choroidal neovascularization via integrin αV. Lab Invest. 2016;96(11):1178–88.

    Article  CAS  PubMed  Google Scholar 

  146. Kelly U, Yu L, Kumar P, Ding JD, Jiang H, Hageman GS et al. Heparan sulfate, including that in Bruch’s membrane, inhibits the complement alternative pathway: implications for age-related macular degeneration. Journal of immunology (Baltimore, Md: 1950). 2010;185(9):5486-94.

  147. Keenan TD, Clark SJ, Unwin RD, Ridge LA, Day AJ, Bishop PN. Mapping the differential distribution of proteoglycan core proteins in the adult human retina, choroid, and sclera. Invest Ophthalmol Vis Sci. 2012;53(12):7528–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Yoshida H, Matsushita T, Kimura E, Fujita Y, Keany R, Ikeda T, et al. Systemic expression of Alu RNA in patients with geographic atrophy secondary to age-related macular degeneration. PLoS ONE. 2019;14(8):e0220887.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Zhu Y, Liang L, Qian D, Yu H, Yang P, Lei B, et al. Increase in peripheral blood mononuclear cell toll-like receptor 2/3 expression and reactivity to their ligands in a cohort of patients with wet age-related macular degeneration. Mol Vis. 2013;19:1826–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Maloney SC, Antecka E, Orellana ME, Fernandes BF, Odashiro AN, Eghtedari M, et al. Choroidal neovascular membranes express toll-like receptor 3. Ophthalmic Res. 2010;44(4):237–41.

    Article  CAS  PubMed  Google Scholar 

  151. Sánchez-Cruz A, Méndez AC, Lizasoain I, de la Villa P, de la Rosa EJ, Hernández-Sánchez C. Tlr2 Gene Deletion Delays Retinal Degeneration in Two Genetically Distinct Mouse Models of Retinitis Pigmentosa. International journal of molecular sciences. 2021;22(15).

  152. Syeda S, Patel AK, Lee T, Hackam AS. Reduced photoreceptor death and improved retinal function during retinal degeneration in mice lacking innate immunity adaptor protein MyD88. Exp Neurol. 2015;267:1–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Will-Orrego A, Qiu Y, Fassbender ES, Shen S, Aranda J, Kotagiri N, et al. Amount of mononuclear Phagocyte Infiltrate does not predict area of experimental Choroidal Neovascularization (CNV). J Ocular Pharmacol Therapeutics: Official J Association Ocular Pharmacol Ther. 2018;34(7):489–99.

    Article  CAS  Google Scholar 

  154. Fujimoto T, Sonoda KH, Hijioka K, Sato K, Takeda A, Hasegawa E, et al. Choroidal neovascularization enhanced by Chlamydia pneumoniae via toll-like receptor 2 in the retinal pigment epithelium. Invest Ophthalmol Vis Sci. 2010;51(9):4694–702.

    Article  PubMed  Google Scholar 

  155. Sun Y, Ni Y, Kong N, Huang C. TLR2 signaling contributes to the angiogenesis of oxygen-induced retinopathy. Exp Eye Res. 2021;210:108716.

    Article  CAS  PubMed  Google Scholar 

  156. Guimaraes TAC, Georgiou M, Bainbridge JWB, Michaelides M. Gene therapy for neovascular age-related macular degeneration: rationale, clinical trials and future directions. Br J Ophthalmol. 2021;105(2):151–7.

    Article  PubMed  Google Scholar 

  157. Chakraborty C, Sharma AR, Sharma G, Doss CGP, Lee SS. Therapeutic miRNA and siRNA: moving from Bench to Clinic as Next Generation Medicine. Mol Therapy Nucleic Acids. 2017;8:132–43.

    Article  CAS  PubMed  Google Scholar 

  158. Ashikari M, Tokoro M, Itaya M, Nozaki M, Ogura Y. Suppression of laser-induced choroidal neovascularization by nontargeted siRNA. Invest Ophthalmol Vis Sci. 2010;51(7):3820–4.

    Article  PubMed  Google Scholar 

  159. Shiose S, Chen Y, Okano K, Roy S, Kohno H, Tang J, et al. Toll-like receptor 3 is required for development of retinopathy caused by impaired all-trans-retinal clearance in mice. J Biol Chem. 2011;286(17):15543–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Grote K, Schütt H, Schieffer B. Toll-like receptors in angiogenesis. TheScientificWorldJournal. 2011;11:981–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Ma B, Dohle E, Li M, Kirkpatrick CJ. TLR4 stimulation by LPS enhances angiogenesis in a co-culture system consisting of primary human osteoblasts and outgrowth endothelial cells. J Tissue Eng Regen Med. 2017;11(6):1779–91.

    Article  CAS  PubMed  Google Scholar 

  162. Xia S, Menden HL, Korfhagen TR, Kume T, Sampath V. Endothelial immune activation programmes cell-fate decisions and angiogenesis by inducing angiogenesis regulator DLL4 through TLR4-ERK-FOXC2 signalling. J Physiol. 2018;596(8):1397–417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. He C, Sun Y, Ren X, Lin Q, Hu X, Huang X, et al. Angiogenesis mediated by toll-like receptor 4 in ischemic neural tissue. Arterioscler Thromb Vasc Biol. 2013;33(2):330–8.

    Article  CAS  PubMed  Google Scholar 

  164. Lin Q, Yang XP, Fang D, Ren X, Zhou H, Fang J, et al. High-mobility group box-1 mediates toll-like receptor 4-dependent angiogenesis. Arterioscler Thromb Vasc Biol. 2011;31(5):1024–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Wu Q, Chen Z, Wu C, Zhang L, Wu Y, Liu X, et al. MD2 inhibits Choroidal Neovascularization via antagonizing TLR4/MD2 mediated signaling pathway. Curr Eye Res. 2023;48(5):474–84.

    Article  CAS  PubMed  Google Scholar 

  166. Hata M, Andriessen E, Hata M, Diaz-Marin R, Fournier F, Crespo-Garcia S, et al. Past history of obesity triggers persistent epigenetic changes in innate immunity and exacerbates neuroinflammation. Sci (New York NY). 2023;379(6627):45–62.

    Article  ADS  CAS  Google Scholar 

  167. Chan C-C, Shen D, Wang Y, Chu X, Abu-Asab M, Tuo J. Inflammasomes in human eyes with AMD and mouse retinas with focal retinal degeneration. Investig Ophthalmol Vis Sci. 2013;54(15):315.

    Google Scholar 

  168. Doyle SL, Campbell M, Ozaki E, Salomon RG, Mori A, Kenna PF, et al. NLRP3 has a protective role in age-related macular degeneration through the induction of IL-18 by drusen components. Nat Med. 2012;18(5):791–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Narendran S, Pereira F, Yerramothu P, Apicella I, Wang S-b, Ambati K, et al. Nucleoside reverse transcriptase inhibitors and Kamuvudines inhibit amyloid-β induced retinal pigmented epithelium degeneration. Signal Transduct Target Therapy. 2021;6(1):149.

    Article  CAS  Google Scholar 

  170. Wakx A, Dutot M, Massicot F, Mascarelli F, Limb GA, Rat P. Amyloid β Peptide Induces Apoptosis Through P2X7 Cell Death Receptor in Retinal Cells: Modulation by Marine Omega-3 Fatty Acid DHA and EPA. Appl Biochem Biotechnol. 2016;178(2):368–81.

    Article  CAS  PubMed  Google Scholar 

  171. Zhao M, Bai Y, Xie W, Shi X, Li F, Yang F, et al. Interleukin-1β level is increased in vitreous of patients with neovascular age-related Macular Degeneration (nAMD) and Polypoidal Choroidal Vasculopathy (PCV). PLoS ONE. 2015;10(5):e0125150.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Nassar K, Grisanti S, Elfar E, Lüke J, Lüke M, Grisanti S. Serum cytokines as biomarkers for age-related macular degeneration. Graefe’s Archive Clin Experimental Ophthalmol. 2015;253(5):699–704.

    Article  CAS  Google Scholar 

  173. Yildirim Z, Ucgun NI, Yildirim F, Sepici-Dincel A. Choroidal Neovascular Membrane in Age-Related Macular Degeneration is Associated with increased Interleukin-6. Int J Gerontol. 2012;6(2):101–4.

    Article  Google Scholar 

  174. Krogh Nielsen M, Subhi Y, Molbech CR, Falk MK, Nissen MH, Sørensen TL. Systemic levels of Interleukin-6 correlate with Progression Rate of Geographic Atrophy secondary to age-related Macular Degeneration. Invest Ophthalmol Vis Sci. 2019;60(1):202–8.

    Article  PubMed  Google Scholar 

  175. Lyu Y, Zauhar R, Dana N, Strang CE, Hu J, Wang K, et al. Implication of specific retinal cell-type involvement and gene expression changes in AMD progression using integrative analysis of single-cell and bulk RNA-seq profiling. Sci Rep. 2021;11(1):15612.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  176. Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20(13).

  177. Abais JM, Xia M, Zhang Y, Boini KM, Li PL. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid Redox Signal. 2015;22(13):1111–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Beatty S, Koh H, Phil M, Henson D, Boulton M. The role of oxidative stress in the pathogenesis of age-related macular degeneration. Surv Ophthalmol. 2000;45(2):115–34.

    Article  CAS  PubMed  Google Scholar 

  179. Liao Y, Zhang H, He D, Wang Y, Cai B, Chen J, et al. Retinal pigment epithelium cell death is Associated with NLRP3 inflammasome activation by all-trans Retinal. Invest Ophthalmol Vis Sci. 2019;60(8):3034–45.

    Article  CAS  PubMed  Google Scholar 

  180. Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, et al. NLRP3 inflammasome is expressed and functional in mouse brain Microglia but not in astrocytes. PLoS ONE. 2015;10(6):e0130624.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Gong T, Yang Y, Jin T, Jiang W, Zhou R. Orchestration of NLRP3 inflammasome activation by Ion Fluxes. Trends Immunol. 2018;39(5):393–406.

    Article  CAS  PubMed  Google Scholar 

  182. Beckel JM, Argall AJ, Lim JC, Xia J, Lu W, Coffey EE, et al. Mechanosensitive release of adenosine 5’-triphosphate through pannexin channels and mechanosensitive upregulation of pannexin channels in optic nerve head astrocytes: a mechanism for purinergic involvement in chronic strain. Glia. 2014;62(9):1486–501.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Suzuki T, Hide I, Ido K, Kohsaka S, Inoue K, Nakata Y. Production and release of neuroprotective tumor necrosis factor by P2X7 receptor-activated microglia. J Neuroscience: Official J Soc Neurosci. 2004;24(1):1–7.

    Article  CAS  Google Scholar 

  184. He Y, Taylor N, Fourgeaud L, Bhattacharya A. The role of microglial P2X7: modulation of cell death and cytokine release. J Neuroinflamm. 2017;14(1):135.

    Article  Google Scholar 

  185. Shieh CH, Heinrich A, Serchov T, van Calker D, Biber K. P2X7-dependent, but differentially regulated release of IL-6, CCL2, and TNF-α in cultured mouse microglia. Glia. 2014;62(4):592–607.

    Article  PubMed  Google Scholar 

  186. Kataoka A, Tozaki-Saitoh H, Koga Y, Tsuda M, Inoue K. Activation of P2X7 receptors induces CCL3 production in microglial cells through transcription factor NFAT. J Neurochem. 2009;108(1):115–25.

    Article  CAS  PubMed  Google Scholar 

  187. Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–41.

    Article  ADS  CAS  PubMed  Google Scholar 

  188. Jessop F, Hamilton RF Jr., Rhoderick JF, Fletcher P, Holian A. Phagolysosome acidification is required for silica and engineered nanoparticle-induced lysosome membrane permeabilization and resultant NLRP3 inflammasome activity. Toxicol Appl Pharmcol. 2017;318:58–68.

    Article  CAS  Google Scholar 

  189. Tsutsumi C, Sonoda KH, Egashira K, Qiao H, Hisatomi T, Nakao S, et al. The critical role of ocular-infiltrating macrophages in the development of choroidal neovascularization. J Leukoc Biol. 2003;74(1):25–32.

    Article  CAS  PubMed  Google Scholar 

  190. Kleinman ME, Kaneko H, Cho WG, Dridi S, Fowler BJ, Blandford AD, et al. Short-interfering RNAs induce retinal degeneration via TLR3 and IRF3. Mol Therapy: J Am Soc Gene Therapy. 2012;20(1):101–8.

    Article  CAS  Google Scholar 

  191. Orgel LE, Crick FH. Selfish DNA: the ultimate parasite. Nature. 1980;284(5757):604–7.

    Article  ADS  CAS  PubMed  Google Scholar 

  192. Deininger P. Alu elements: know the SINEs. Genome Biol. 2011;12(12):236.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Kerur N, Hirano Y, Tarallo V, Fowler BJ, Bastos-Carvalho A, Yasuma T, et al. TLR-independent and P2X7-dependent signaling mediate Alu RNA-induced NLRP3 inflammasome activation in geographic atrophy. Invest Ophthalmol Vis Sci. 2013;54(12):7395–401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Kerur N, Fukuda S, Banerjee D, Kim Y, Fu D, Apicella I, et al. cGAS drives noncanonical-inflammasome activation in age-related macular degeneration. Nat Med. 2018;24(1):50–61.

    Article  CAS  PubMed  Google Scholar 

  195. Li J, Zhang F, Bian W, Chen Y, Liu J, Liu Z, et al. cGAS inhibition alleviates Alu RNA-induced immune responses and cytotoxicity in retinal pigmented epithelium. Cell Bioscience. 2022;12(1):116.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  196. Evavold CL, Ruan J, Tan Y, Xia S, Wu H, Kagan JC. The pore-forming protein gasdermin D regulates Interleukin-1 secretion from living macrophages. Immunity. 2018;48(1):35–44e6.

    Article  CAS  PubMed  Google Scholar 

  197. Decout A, Katz JD, Venkatraman S, Ablasser A. The cGAS–STING pathway as a therapeutic target in inflammatory diseases. Nat Rev Immunol. 2021;21(9):548–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Jarrett SG, Lin H, Godley BF, Boulton ME. Mitochondrial DNA damage and its potential role in retinal degeneration. Prog Retin Eye Res. 2008;27(6):596–607.

    Article  CAS  PubMed  Google Scholar 

  199. Gupta U, Ghosh S, Wallace CT, Shang P, Xin Y, Nair AP, et al. Increased LCN2 (lipocalin 2) in the RPE decreases autophagy and activates inflammasome-ferroptosis processes in a mouse model of dry AMD. Autophagy. 2023;19(1):92–111.

    Article  CAS  PubMed  Google Scholar 

  200. Sekar R, Wooff Y, Cioanca AV, Kurera M, Ngo C, Man SM, et al. Impairing gasdermin D-mediated pyroptosis is protective against retinal degeneration. J Neuroinflamm. 2023;20(1):239.

    Article  CAS  Google Scholar 

  201. Hirano Y, Yasuma T, Mizutani T, Fowler BJ, Tarallo V, Yasuma R, et al. IL-18 is not therapeutic for neovascular age-related macular degeneration. Nat Med. 2014;20(12):1372–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Doyle SL, Adamson P, López FJ, Humphries P, Campbell M. Reply to IL-18 is not therapeutic for neovascular age-related macular degeneration. Nat Med. 2014;20(12):1376–7.

    Article  CAS  PubMed  Google Scholar 

  203. Malsy J, Alvarado AC, Lamontagne JO, Strittmatter K, Marneros AG. Distinct effects of complement and of NLRP3- and non-NLRP3 inflammasomes for choroidal neovascularization. eLife. 2020;9:e60194.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Zhang W, Ma Y, Zhang Y, Yang J, He G, Chen S. Photo-oxidative Blue-Light Stimulation in Retinal Pigment Epithelium cells promotes Exosome Secretion and increases the activity of the NLRP3 inflammasome. Curr Eye Res. 2019;44(1):67–75.

    Article  CAS  PubMed  Google Scholar 

  205. Liu J, Copland DA, Theodoropoulou S, Chiu HA, Barba MD, Mak KW, et al. Impairing autophagy in retinal pigment epithelium leads to inflammasome activation and enhanced macrophage-mediated angiogenesis. Sci Rep. 2016;6:20639.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  206. Fowler BJ, Gelfand BD, Kim Y, Kerur N, Tarallo V, Hirano Y, et al. Nucleoside reverse transcriptase inhibitors possess intrinsic anti-inflammatory activity. Sci (New York NY). 2014;346(6212):1000–3.

    Article  ADS  CAS  Google Scholar 

  207. Garces K, Carmy T, Illiano P, Brambilla R, Hackam AS. Increased neuroprotective microglia and photoreceptor survival in the Retina from a peptide inhibitor of myeloid differentiation factor 88 (MyD88). J Mol Neurosci. 2020;70(6):968–80.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The illustrations were created using BioRender.com.

Funding

AB from NHMRC (#GNT1141587), ELF and KV from NHMRC (APP#1138253; #2011200).

Author information

Authors and Affiliations

Authors

Contributions

AB wrote the initial draft and created the figures. ELF wrote part of the initial draft and edited the final draft. KV edited final draft. All authors read and approved final manuscript.

Corresponding author

Correspondence to Erica L. Fletcher.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

A.B. is an employee of F. Hoffmann-La Roche Ltd.

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

Brandli, A., Vessey, K.A. & Fletcher, E.L. The contribution of pattern recognition receptor signalling in the development of age related macular degeneration: the role of toll-like-receptors and the NLRP3-inflammasome. J Neuroinflammation 21, 64 (2024). https://0-doi-org.brum.beds.ac.uk/10.1186/s12974-024-03055-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12974-024-03055-1

Keywords