Skip to main content

MINI REVIEW article

Front. Cell Dev. Biol., 28 February 2023
Sec. Molecular and Cellular Pathology
Volume 11 - 2023 | https://doi.org/10.3389/fcell.2023.1118524

Innate immune biology in age-related macular degeneration

  • 1Interdepartmental Neuroscience Program, Yale University, New Haven, CT, United States
  • 2Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, California
  • 3Choate Rosemary Hall, Wallingford, CT, United States
  • 4Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, United States
  • 5Department of Pathology, Yale University, New Haven, CT, United States

Age-related macular degeneration (AMD) is a neurodegenerative disease and a leading cause of irreversible vision loss in the developed world. While not classically described as an inflammatory disease, a growing body of evidence has implicated several components of the innate immune system in the pathophysiology of age-related macular degeneration. In particular, complement activation, microglial involvement, and blood-retinal-barrier disruption have been shown to play key roles in disease progression, and subsequent vision loss. This review discusses the role of the innate immune system in age-related macular degeneration as well as recent developments in single-cell transcriptomics that help advance the understanding and treatment of age-related macular degeneration. We also explore the several potential therapeutic targets for age-related macular degeneration in the context of innate immune activation.

Introduction

Age-related macular degeneration (AMD) is a neurodegenerative disorder characterized by the presence of drusen, extracellular deposits, followed by retinal pigment epithelium (RPE) and photoreceptor loss. AMD is rarely seen before age 55, after which prevalence increases with increasing age (Klein et al., 2010). Several classification systems exist for AMD, with disease taxonomy defined based on the frequency and size of macular drusen, the presence of pigmentary abnormalities, and the presence of geographic atrophy or neovascularization (Fleckenstein et al., 2021). AMD is commonly subcategorized into neovascular (wet) disease where choroidal vascular changes are present, and non-neovascular (dry) disease where such changes are absent. While only a minority (10%–15%) of patients will develop neovascular disease, wet AMD is responsible for a majority of cases of sudden vision loss in the disease (Jager et al., 2008).

AMD is a multifactorial disease, with a complex set of risk factors, like age, environment, and genetic susceptibility; the heritability of late stage AMD has been estimated to be up to 71%, higher than most complex age-related diseases (Fleckenstein et al., 2021). The strongest risk factor for AMD is age. Aging is a critical component of AMD pathology; during aging, injury, or inflammation, resident microglia migrate to the affected site. Microglial infiltration can trigger a response involving the infiltration of other myeloid-derived cells, including peripheral monocytes and monocyte-derived macrophages. CCL2 and CX2CL1 chemokines, for instance, are implicated in AMD-related subretinal microglial and macrophage infiltration (Raoul et al., 2010). Expression patterns of cytokine and cytokine receptors change as biological age and state progress, indicating these inflammatory mechanisms may be involved in AMD development.

Modifiable risk factors for AMD include smoking, diet, and physical activity. Smoking has been shown to increase the chance of developing AMD by two to four times, and cessation reduces AMD risk (Velilla et al., 2013). Consumption of foods with lutein, zeaxanthin, fish oil, and polyunsaturated fatty acids have been shown to decrease the chance of late stage AMD. These insights have led to the formulation of the AREDS vitamin supplementation regimen, a mainstay of therapy for non-neovascular AMD (AREDS2 Research Group et al., 2012).

While the precise pathogenesis of AMD remains unknown, several lines of evidence implicate drusen, the hallmark lesions of AMD. Several factors, including amyloid, complement, and 7-ketocholesterol, have been isolated from drusen and are hypothesized to contribute to AMD progression (Mullins et al., 2000; Rodriguez et al., 2014). The development of neovascular AMD in particular is strongly associated with the vascular endothelial growth factor (VEGF) family of cytokines and resultant signaling through Flt-1 (Nork et al., 2011). The identification of this link has been key to the development of a host of anti-VEGF therapies that have been used in the treatment of neovascular AMD (Solomon et al., 2019).

In the retina, glial cells support photoreceptor function by providing physical and metabolic support through a number of mechanisms such as transporting nutrients and clearing debris. The three broad types of glial cells in the retina are microglia, astrocytes and Müller cells (Reichenbach and Bringmann, 2019). Retinal microglia respond to retinal injury and mediating neuroinflammation, where retinal macroglia–astrocytes and Müller cells–maintain retinal homeostasis through regulation and transport of ions, glucose, and neurotransmitters. Microglia and macroglia involvement have both been implicated in AMD pathogenesis.

Methods exploring the underpinnings of AMD are rapidly developing. In recent years, scientists have been using single-cell RNA sequencing (scRNA-seq) to analyze the pathogenesis and etiology of diseases whose origins remain unclear. Due to the genetic complexity of AMD, it has been difficult to precisely identify the cell-types associated with AMD development. In 2019, researchers used scRNA-seq to develop the first single-cell transcriptomic atlas of the human retina, a full record of the transcriptomes of each cell type (Menon et al., 2019). Use of the transcriptomic atlas predicts Müller glia and astrocytes have a role in the pathogenesis of AMD (Menon et al., 2019).

A 2021 study built upon these findings by focusing on the genes transcribed instead of the cells. This study used scRNA-seq to obtain the transcriptomes of 93,000 retinal cells; in addition to using the data to transcriptionally identify differences in retinal neurons, researchers also assessed the cell-type expression levels of each AMD risk gene (Lyu et al., 2021). Through this method, 23 risk genes were found to be cell-type-specific genes. For instance, CFH, a gene strongly associated with AMD, was determined to be specifically expressed in endothelium cells. This relationship implied that endothelial cells may play a role in the development of AMD. In addition, C3 and CFI, two other AMD genes, were found to be preferentially expressed; C3 by microglia and astrocytes, and CFI by astrocytes, endothelium, and Müller cells. Although the complete pathogenesis of AMD remains unclear, scRNA-seq allows scientists to better understand which cell types are involved in the disease’s development.

A growing body of research has shown that AMD pathogenesis is associated with neuroimmune interactions; most recently, neovascular AMD pathology was associated with COVID-19 infection severity and morbidity, providing just one example of the potential neuroimmune interactions involved in AMD (Yang et al., 2022). The blood-retina barrier protects the retina, making it an immune-privileged part of the eye; still, innate immune interactions may influence the progression of this neurodegenerative disorder. Immune privilege in the eye is characterized in part by Müller glia and astrocytes–in addition to endothelial cells, pericytes, and perivascular macrophages, among others–modulating the trafficking of cells in and out of the retina. However, the innate immune response–including macrophages, monocytes, neutrophils, and natural killer T cells–may serve to play a role in neurodegenerative diseases, including AMD (Novellino et al., 2020; Dhodapkar et al., 2022). Innate immune cells contribute to the maintenance of the blood-retina barrier, vascularization, and inflammation, and their disruption or dysfunction may have implications for the development of AMD. This article focuses on the role of these immune cells in AMD pathogenesis.

Macrophages

Macrophages are a diverse group of cells that serve several functions across tissue repair, immunity, and homeostasis (Wynn et al., 2013). In the central nervous system, macrophages can secrete proinflammatory factors; while microglia constitute the bulk of resident myeloid cells in the central nervous system, monocyte-derived macrophages may become activated depending on the surrounding milieu (Yang et al., 2020). Macrophages along the outer collagenous zones of Bruch’s membrane have been shown to be present in AMD lesions (Killingsworth et al., 1990). The phagocytic ability of cultured human RPE has been found to be decreased in AMD donors (Inana et al., 2018). Macrophages have been shown to become activated in several different ways. A widely used system classifies macrophage activation as either M1 (classic activation) or M2 (alternative activation); however, recently more activation states have been identified. M1 macrophages, specifically, are known to activate phagocytic and antitumor inflammatory responses (Liu et al., 2021). In neovascular AMD monocytes and in the neovascularization-choroidal rodent model, M1 macrophages had a proangiogenic effect; in both models, angiogenesis was mediated by TNF-α, whose inhibition led to a decrease of choroidal neovascularization (Hagbi-Levi et al., 2021). Perivascular macrophages, cells distinct from microglia, are present in the retina and are believed to contribute to the blood-retinal barrier maintenance during homeostasis. During photoreceptor degeneration, these cells may be recruited to sites of damage (Mendes-Jorge et al., 2009). Although the role of these peripheral macrophages in the perivascular choroidal space in AMD is not yet fully understood, alterations to their function may be associated with age-related changes in the choroidal vasculature (Yang et al., 2020).

In the murine model, macrophage ablation by CSF1R blockade leads to choroid vascular atrophy in addition to structural and angiogenic RPE dysfunction, suggesting that macrophages residing in the choroid may serve an important role in structural maintenance of the RPE and choroid vasculature (Yang et al., 2020). One signaling pathway that helps maintain retinal immune homeostasis is the interferon-beta signaling pathway, which in the murine model has been used to reduce microgliosis, macrophage response, and choroidal neovascularization (Lückoff et al., 2016). In cases of photoreceptor degeneration, monocyte infiltration can precede microglia infiltration during the early stages of the immune response (Karlen et al., 2018). This finding suggests that monocyte-derived macrophages along with microglia may play a role in the immune response in AMD (Karlen et al., 2018). In retinitis pigmentosa (Tebbe et al., 2020) monocyte-derived macrophages may contribute to cone death as their inhibition protects against cone degeneration in the rd10 mouse model of retinitis pigmentosa (Funatsu et al., 2022). Given their overall role in choroidal vascularization and RPE maintenance, targeting these cells may be a potential therapeutic approach for AMD, particularly as it relates to limiting angiogenesis and chronic inflammation. It may be possible to explore stem cell therapeutic potential to restore their phagocytic function as genetically-modified stem cells have previously been shown to promote turnover of perivascular macrophages in the macaque brain (Soulas et al., 2009).

Monocytes

Monocytes, as part of the immune system, can differentiate into macrophages or dendritic cells. Monocyte-derived macrophages, as aforementioned, appear to play a role in AMD, but the primary difference between these two cells is their localization; monocytes circulate in the bloodstream where macrophages infiltrate tissue. Peripheral monocyte counts, in particular, have been shown to be higher in AMD patients versus controls (Xue et al., 2021). In intermediate and advanced AMD, monocyte phagocytic function is shown to be reduced about 40% across several monocyte subtypes compared to controls (Gu et al., 2021). Gu et al. found that this effect could be ameliorated by application of glatiramer acetate, which may provide a therapeutic avenue for AMD (Gu et al., 2021). Glatiramer acetate is an immunomodulatory polypeptide that is used in the treatment of relapsing or remitting multiple sclerosis (Gran et al., 2000). Increased TNF-α is also associated with monocyte activation, as patients with higher prevalence of choroidal neovascularization demonstrated high levels of monocyte TNF-α (Cousins et al., 2004).

In atrophic, or dry, AMD, CCR2+ monocytes have been identified in the subretinal space (Sennlaub et al., 2013). In the mouse model, these monocytes were also shown to possibly contribute to photoreceptor degeneration. Furthermore, CD163+ monocytes and macrophages may have a role in AMD pathology and progression (Swayze et al., 2022). In late stages of AMD, these changes in monocyte function can potentially be modified by the P2X7 receptor, which is expressed in a variety of cells, especially in monocytes, and can function as a scavenger receptor mediating phagocytosis (Drysdale et al., 2022). The P2X7 receptor mediated membrane fluidity, which was reduced across several types of leukocytes, including monocytes, in advanced AMD. In mice, impaired cholesterol clearance by monocytes in the murine model triggers several events leading to the development of cholesterol-rich drusen deposits underneath the RPE, a hallmark of AMD (Ban et al., 2018). These drusen deposits may be associated with disease progression and may provide a potential therapeutic target for AMD.

Natural killer cells

Natural killer (NK) cells are a component of the innate immune system, affecting cytokine production and cytotoxicity, with antiviral and antitumoral effects by killing cells that downregulate the MHC Class I self-antigen presentation system (Vivier et al., 2008; Bern et al., 2019). In a genotyping study of AMD and control patients, the NK receptor AA haplotype was, in combination with the human leukocyte antigen (HLA)-Cw*0701 allele, associated with AMD (Goverdhan et al., 2008). In another study of gene expression profiles in AMD, there was a lower prevalence of resting NK cells in AMD versus control, but the C1S, ADM, and 1ER5L genes were shown to have a positive correlation with activation of NK cells, among others, along AMD progression (Zeng et al., 2021). Further experiments in mice models have shown that in vivo depletion of interferon-γ-secreting NK cells lead to reduced choroidal neovascularization (CNV) (Lee et al., 2014).

Neutrophils

Neutrophils are a type of leukocyte that drive both acute and chronic inflammation (Kolaczkowska and Kubes, 2013). As with monocytes, the P2X7 receptor mediates membrane fluidity, which is reduced in neutrophils and other leukocytes in advanced AMD (Drysdale et al., 2022). Furthermore, the neutrophil-to-leukocyte ratio is higher in patients with neovascular AMD (Niazi et al., 2019). Increased INFλ levels lead to LCN-2 upregulation and neutrophil activation; inhibition of this INFλ inflammatory signal, through AKT2 inhibition, leads to decreased LCN-2-mediated neutrophil infiltration, which may provide a novel therapeutic approach to early dry AMD (Ghosh et al., 2019).

Neutrophils are characterized by their strategy for pathogen eradication, which is known as a neutrophil extracellular trap (Kolaczkowska and Kubes, 2013). Neutrophil extracellular trap formation has previously been associated with retinal diseases (Ghosh et al., 2019; Wang et al., 2018). Aβ1-40 the main amyloid-beta component of the drusen characteristic of AMD may promote neutrophil extracellular trap formation in AMD through the Toll-like receptor 4 and neutrophil NADPH oxidase pathways (Chen et al., 2022). PAD4 inhibition in the mouse model led to reduced neutrophil extracellular trap formation, providing a potential therapeutic option for AMD (Chen et al., 2022). PAD4 inhibition is currently used to treat rheumatoid arthritis and lupus, among other autoimmune diseases. This finding suggests a possible role for neutrophils in AMD that needs to be explored further.

Dendritic, B, and T cells

Dendritic cells are innate immune cells that activate lymphocytes, in addition to capturing and processing antigens (Banchereau and Steinman, 1998). Early dendritic cell recruitment occurs during retinal injury (Lehmann et al., 2010). In a laser-induced CNV mouse model of AMD, both wild-type and dendritic-cell-deficient mice exhibited similar levels of CNV area, (Droho et al., 2021). Dendritic and B cells alike are yet to be strongly linked with AMD progression.

T cells are lymphocytes that can be activated by dendritic cells. Chemokine profiles of CD4+ and CD8+ patients differ between AMD and control conditions (Choi et al., 2022). Furthermore, in AMD versus control gene expression analysis, the AMD condition exhibited lower resting CD4+ memory T cells (Zeng et al., 2021). T cells may become activated in response to oxidative stress (Cruz-Guilloty et al., 2014). T-helper (Th) 1 T cells were found at lower frequency in patients with neovascular AMD (Singh et al., 2017). CD56+ CD28- T cells in the peripheral blood are higher in AMD versus control patients, suggesting a possible function of T cells in AMD (Faber et al., 2013).

CNV lesions in the murine model triggered T cell immune response as well, with an increase in IL-17-producing γδT-cells through C5a (Coughlin et al., 2016). In CD4+ T cells, C5a stimulates IL-17 production, which may imply a role for C5a in AMD and T lymphocyte activation (Liu et al., 2011). Natural killer T (NKT) cells make up a small percentage of innate immune cells. NKT cells are implicated in CNV, and in Cd1d-restricted invariant and NKT-deficient mice, there are lower levels of CNV and VEGF, indicating the potential role of NKT cells in AMD (Hijioka et al., 2008).

Possible therapeutic targets include targeting the interferon-beta signaling pathway or the AKT2 signaling pathway (Lückoff et al., 2016; Ghosh et al., 2019). Targeting proteins such as PAD4 or C5a may produce similar results. Recent clinical trials for geographic atrophy have shown success at targeting the complement system (Coughlin et al., 2016; Chen et al., 2022). Therapeutic targets implicated in other neurodegenerative diseases such as glatiramer acetate, which is primarily used in MS has shown some efficacy in reversing AMD monocyte pathology in vitro (Gu et al., 2021). One limitation of potential therapies is their use in the murine model, which lacks an anatomical macula (Pennesi et al., 2012).

Conclusion

Progress in understanding the peripheral immune response in AMD has been made and future research on human retinas with AMD using single-cell approaches will further help the field. Furthermore, the development of non-human primate models of AMD or human retinal organoids derived from human pluripotent stem cells from patients with AMD may facilitate the translational development therapeutic interventions for AMD targeting immune dysregulation.

Author contributions

KA, RD, and BH all contributed to the generation of this topic and literature review. KA wrote all sections except for some sections of the Abstract, Introduction, and Conclusion. RD and BH wrote Abstract, Introduction, and Conclusion, in addition to substantially editing KA’s sections. DH wrote a section of the Introduction.

Funding

BH receives research funding from NIH R01-EY034234, the Thome Memorial Foundation, the Doris Duke Charitable Foundation, the H. Eric Cushing Foundation, the Nancy Lurie Marks Family Foundation, the C. J. L. Charitable Foundation, the Reynold and Michiko Spector Award in Neuroscience, Hoffmann-La Roche Pharmaceuticals, and as the William R. Orthwein Jr. Scholar.

Conflict of interest

BH is on the scientific advisory board of Carmine Therapeutics.

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

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Arnon, R., and Rina, A. (2004). Mechanism of action of glatiramer acetate in multiple sclerosis and its potential for the development of new applications. Proc. Natl. Acad. Sci. U. S. A. 101, 14593–14598. –98. doi:10.1073/pnas.0404887101

PubMed Abstract | CrossRef Full Text | Google Scholar

AREDS2 Research Group, Chew, E. Y., Clemons, T., SanGiovanni, J. P., Danis, R., Domalpally, A., McBee, W., et al. (2012). The age-related eye disease study 2 (AREDS2): Study design and baseline characteristics (AREDS2 report number 1). Ophthalmology 119 (11), 2282–2289. doi:10.1016/j.ophtha.2012.05.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Ban, N., Lee, T. J., Abdoulaye, S., Choudhary, M., Lekwuwa, M., Dong, Z., et al. (2018). Impaired monocyte cholesterol clearance initiates age-related retinal degeneration and vision loss. JCI Insight 3 (17), e120824. doi:10.1172/jci.insight.120824

PubMed Abstract | CrossRef Full Text | Google Scholar

Banchereau, J., and Steinman, R. M. (1998). Dendritic cells and the control of immunity. Nature 392 (6673), 245–252. doi:10.1038/32588

PubMed Abstract | CrossRef Full Text | Google Scholar

Bern, M. D., Parikh, B. A., Liping, Y., Beckman, D. L., Poursine-Laurent, J., Wayne, M. Y., et al. (2019). Inducible down-regulation of MHC Class I results in natural killer cell tolerance. J. Exp. Med. 216 (1), 99–116. doi:10.1084/jem.20181076

PubMed Abstract | CrossRef Full Text | Google Scholar

Brauning, A., Michael, R., Zhu, G., Fulton, E., Alexandra, S., and Sharma, A. (2022). Aging of the immune system: Focus on natural killer cells phenotype and functions. Cells 11 (6), 1017. doi:10.3390/cells11061017

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Zhao, L., Ding, X., Wen, Y., Wang, L., Shu, Q., et al. (2022). Aβ1-40 oligomers trigger neutrophil extracellular trap formation through TLR4-and NADPH oxidase-dependent pathways in age-related macular degeneration. Oxidative Med. Cell. Longev. 2022, 6489923. doi:10.1155/2022/6489923

CrossRef Full Text | Google Scholar

Choi, Y. J., Lim, D., Byeon, S. H., Shin, E.-C., and Chung, H. (2022). Chemokine receptor profiles of T cells in patients with age-related macular degeneration. Yonsei Med. J. 63 (4), 357–364. doi:10.3349/ymj.2022.63.4.357

PubMed Abstract | CrossRef Full Text | Google Scholar

Coughlin, B., Schnabolk, G., Joseph, K., Raikwar, H., Kannan, K., Johnson, K., et al. (2016). Connecting the innate and adaptive immune responses in mouse choroidal neovascularization via the anaphylatoxin C5a and γδt-cells. Sci. Rep. 6, 23794. doi:10.1038/srep23794

PubMed Abstract | CrossRef Full Text | Google Scholar

Cousins, S. W., Espinosa-Heidmann, D. G., and Csaky, K. G. (2004). Monocyte activation in patients with age-related macular degeneration: A biomarker of risk for choroidal neovascularization? Archives Ophthalmol. 122 (7), 1013–1018. doi:10.1001/archopht.122.7.1013

PubMed Abstract | CrossRef Full Text | Google Scholar

Cruz-Guilloty, F., Saeed, A. M., Duffort, S., Cano, M., Ebrahimi, K. B., Ballmick, A., et al. (2014). T cells and macrophages responding to oxidative damage cooperate in pathogenesis of a mouse model of age-related macular degeneration. PloS One 9 (2), e88201. doi:10.1371/journal.pone.0088201

PubMed Abstract | CrossRef Full Text | Google Scholar

Dhodapkar, R. M., Martell, D., and Hafler, B. P. (2022). Glial-mediated neuroinflammatory mechanisms in age-related macular degeneration. Seminars Immunopathol. 44 (5), 673–683. doi:10.1007/s00281-022-00939-3

CrossRef Full Text | Google Scholar

Droho, S., Perlman, H., and Lavine, J. A. (2021). Dendritic cells play No significant role in the laser-induced choroidal neovascularization model. Sci. Rep. 11 (1), 17254. doi:10.1038/s41598-021-96704-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Drysdale, C., Park, K., Vessey, K. A., Huang, X., Caruso, E., Li, Y., et al. (2022). P2X7-Mediated alteration of membrane fluidity is associated with the late stages of age-related macular degeneration. Purinergic Signal. 18, 469–479. doi:10.1007/s11302-022-09894-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Faber, C., Singh, A., Falk, M. K., Juel, H. B., Sørensen, T. L., and Nissen, M. H. (2013). Age-related macular degeneration is associated with increased proportion of CD56(+) T cells in peripheral blood. Ophthalmology 120 (11), 2310–2316. doi:10.1016/j.ophtha.2013.04.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Fleckenstein, M., Keenan, T. D. L., Guymer, R. H., Chakravarthy, U., Schmitz-Valckenberg, S., Klaver, C. C., et al. (2021). Age-related macular degeneration. Nat. Rev. Dis. Prim. 7 (1), 31. doi:10.1038/s41572-021-00265-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Funatsu, J., Murakami, Y., Shimokawa, S., Nakatake, S., Fujiwara, K., Okita, A., et al. (2022). Circulating inflammatory monocytes oppose microglia and contribute to cone cell death in retinitis pigmentosa. PNAS Nexus 1 (1), pgac003. doi:10.1093/pnasnexus/pgac003

PubMed Abstract | CrossRef Full Text | Google Scholar

Gawecki, M., Doroszkiewicz, M., and Rydzewski, J.(2010). Age related macular degeneration and presence of posterior vitreous detachment. Klin. Ocz. 112 (7-9), 210–212.

PubMed Abstract | Google Scholar

Ghosh, S., Padmanabhan, A., Vaidya, T., Watson, A. M., Bhutto, I. A., Hose, S., et al. (2019). Neutrophils homing into the retina trigger pathology in early age-related macular degeneration. Commun. Biol. 2, 348. doi:10.1038/s42003-019-0588-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Goverdhan, S. V., Khakoo, S. I., Gaston, H., Chen, X., and Lotery, A. J. (2008). Age-related macular degeneration is associated with the HLA-cw*0701 genotype and the natural killer cell receptor AA haplotype. Investigative Ophthalmol. Vis. Sci. 49 (11), 5077–5082. doi:10.1167/iovs.08-1837

PubMed Abstract | CrossRef Full Text | Google Scholar

Gran, B., Tranquill, L. R., Chen, M., Bielekova, B., Zhou, W., Dhib-Jalbut, S., et al. (2000). Mechanisms of immunomodulation by glatiramer acetate. Neurology 55 (11), 1704–1714. doi:10.1212/wnl.55.11.1704

PubMed Abstract | CrossRef Full Text | Google Scholar

Gu, B. J., Huang, X., Avula, P. K., Caruso, E., Drysdale, C., Vessey, K. A., et al. (2021). Deficits in monocyte function in age related macular degeneration: A novel systemic change associated with the disease. Front. Med. 8, 634177. doi:10.3389/fmed.2021.634177

CrossRef Full Text | Google Scholar

Hagbi-Levi, S., Tiosano, L., Rinsky, B., Levinger, N., Elbaz-Hayoun, S., Carmi, S., et al. (2021). Anti-tumor necrosis factor alpha reduces the proangiogenic effects of activated macrophages derived from patients with age-related macular degeneration. Mol. Vis. 27, 622–631.

PubMed Abstract | Google Scholar

Hijioka, K., Sonoda, K-H., Tsutsumi-Miyahara, C., Fujimoto, T., Oshima, Y., Taniguchi, M., et al. (2008). Investigation of the role of CD1d-restricted invariant NKT cells in experimental choroidal neovascularization. Biochem. Biophysical Res. Commun. 374 (1), 38–43. doi:10.1016/j.bbrc.2008.06.080

PubMed Abstract | CrossRef Full Text | Google Scholar

Inana, G., Murat, C., An, W., Yao, X., Harris, I. R., Cao, J., et al. (2018). RPE phagocytic function declines in age-related macular degeneration and is rescued by human umbilical tissue derived cells. J. Transl. Med. 16 (1), 63. doi:10.1186/s12967-018-1434-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Jager, R. D., Mieler, W. F., and Miller., J. W. (2008). Age-related macular degeneration. N. Engl. J. Med. 358 (24), 2606–2617. doi:10.1056/NEJMra0801537

PubMed Abstract | CrossRef Full Text | Google Scholar

Karlen, S. J., Miller, E. B., Wang, X., Emily, S., Levine, R., Zawadzki, J., et al. (2018). Monocyte infiltration rather than microglia proliferation dominates the early immune response to rapid photoreceptor degeneration. J. Neuroinflammation 15 (1), 344. doi:10.1186/s12974-018-1365-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Kataoka, K., Nishiguchi, K. M., Kaneko, H., van Rooijen, N., Shu, K., and Terasaki, H. (2011). The roles of vitreal macrophages and circulating leukocytes in retinal neovascularization. Investigative Ophthalmol. Vis. Sci. 52 (3), 1431–1438. doi:10.1167/iovs.10-5798

PubMed Abstract | CrossRef Full Text | Google Scholar

Killingsworth, M. C., Sarks, J. P., and Sarks, S. H. (1990). Macrophages related to Bruch’s membrane in age-related macular degeneration. Eye 4, 613–621. doi:10.1038/eye.1990.86

PubMed Abstract | CrossRef Full Text | Google Scholar

Klein, R., Cruickshanks, K. J., Nash, S. D., Krantz, E. M., Nieto, F. J., Huang, G. H., et al. (2010). The prevalence of age-related macular degeneration and associated risk factors. Arch. Ophthalmol. Jun 128 (6), 750–758. doi:10.1001/archophthalmol.2010.92

PubMed Abstract | CrossRef Full Text | Google Scholar

Kolaczkowska, E., and Kubes, P. (2013). Neutrophil recruitment and function in Health and inflammation. Nat. Rev. Immunol. 13 (3), 159–175. doi:10.1038/nri3399

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H., Schlereth, S. L., Park, E. Y., Emami-Naeini, P., Chauhan, S. K., Dana, R., et al. (2014). A novel pro-angiogenic function for interferon-γ-secreting natural killer cells. Investigative Ophthalmol. Vis. Sci. 55 (5), 2885–2892. doi:10.1167/iovs.14-14093

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, K. S., Lin, S., Copland, D. A., Dick, A. D., and Liu, J. (2021). Cellular senescence in the aging retina and developments of senotherapies for age-related macular degeneration. J. Neuroinflammation 18 (1), 32. doi:10.1186/s12974-021-02088-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Lehmann, U., Heuss, N. D., McPherson, S. W., Roehrich, H., and Gregerson, D. S. (2010). Dendritic cells are early responders to retinal injury. Neurobiol. Dis. 40 (1), 177–184. doi:10.1016/j.nbd.2010.05.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, B., Lai, W., Meyerle, C., Tuo, J., Nida Sen, H., Li, Z., et al. (2011). Complement component C5a promotes expression of IL-22 and IL-17 from human T cells and its implication in age-related macular degeneration. J. Transl. Med. 9, 1–12. doi:10.1186/1479-5876-9-111

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Geng, X., Hou, J., and Wu, G. (2021). New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 21 (1), 389. doi:10.1186/s12935-021-02089-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Lückoff, A., Albert, C., Scholz, R., Prinz, M., Ulrich, K., and Langmann, T. (2016). Interferon-beta signaling in retinal mononuclear phagocytes attenuates pathological neovascularization. EMBO Mol. Med. 8 (6), 670–678. doi:10.15252/emmm.201505994

PubMed Abstract | CrossRef Full Text | Google Scholar

Lyu, Y., Zauhar, R., Dana, N., Strang, C. E., Hu, J., Wang, K., et al. (2021). 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. 11 (1), 15612. doi:10.1038/s41598-021-95122-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Magnus, H. S., and Sørensen, T. L. (2017). Circulating monocytes and B-lymphocytes in neovascular age-related macular degeneration. Clin. Ophthalmol. 11, 179–184. doi:10.2147/OPTH.S121332

PubMed Abstract | CrossRef Full Text | Google Scholar

Mendes-Jorge, L., Ramos, D., Luppo, M., Llombart, C., Alexandre-Pires, G., Nacher, V., et al. (2009). Scavenger function of resident autofluorescent perivascular macrophages and their contribution to the maintenance of the blood-retinal barrier. Investigative Ophthalmol. Vis. Sci. 50 (12), 5997–6005. doi:10.1167/iovs.09-3515

PubMed Abstract | CrossRef Full Text | Google Scholar

Mullins, R. F., Russell, S. R., Anderson, D. H., and Hageman, G. S. (2000). Drusen associated with aging and age-related macular degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit disease. FASEB J. 14 (7), 835–846. doi:10.1096/fasebj.14.7.835

PubMed Abstract | CrossRef Full Text | Google Scholar

Niazi, S., Krogh Nielsen, M., Sørensen, T. L., and Subhi, Y.(2019). Neutrophil-to-Lymphocyte ratio in age-related macular degeneration: A systematic review and meta-analysis. Acta Ophthalmol. 97 (6), 558–566. doi:10.1111/aos.14072

PubMed Abstract | CrossRef Full Text | Google Scholar

Nork, T. M., Dubielzig, R. R., Christian, B. J., Miller, P. E., Miller, J. M., Cao, J., et al. (2011). Prevention of experimental choroidal neovascularization and resolution of active lesions by VEGF trap in nonhuman primates. Arch. Ophthalmol. Aug 129 (8), 1042–1052. doi:10.1001/archophthalmol.2011.210

PubMed Abstract | CrossRef Full Text | Google Scholar

Novellino, F., Saccà, V., Donato, A., Zaffino, P., Francesca Spadea, M., Vismara, M., et al. (2020). Innate immunity: A common denominator between neurodegenerative and neuropsychiatric diseases. Int. J. Mol. Sci. 21 (3), 1115. doi:10.3390/ijms21031115

PubMed Abstract | CrossRef Full Text | Google Scholar

Pennesi, M. E., Neuringer, M., and Courtney, R. J. (2012). Animal models of age related macular degeneration. Mol. Aspects Med. 33 (4), 487–509. doi:10.1016/j.mam.2012.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Rajesh, A., Droho, S., and Lavine, J. A. (2022). Macrophages in close proximity to the vitreoretinal interface are potential biomarkers of inflammation during retinal vascular disease. J. Neuroinflammation 19 (1), 203. doi:10.1186/s12974-022-02562-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Raoul, W., Auvynet, C., Camelo, S., Guillonneau, X., Feumi, C., Combadiere, C., et al. (2010). CCL2/CCR2 and CX3CL1/CX3CR1 chemokine axes and their possible involvement in age-related macular degeneration. J. Neuroinflammation 7, 87. doi:10.1186/1742-2094-7-87

PubMed Abstract | CrossRef Full Text | Google Scholar

Reichenbach, A., and Bringmann, A. (2019). Glia of the human retina. Glia 68 (4), 768–796. doi:10.1002/glia.23727

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez, I. R., Clark, M. E., Lee, J. W., and Curcio, C. A. (2014). 7-ketocholesterol accumulates in ocular tissues as a consequence of aging and is present in high levels in drusen. Exp. Eye Res. Nov. 128, 151–155. doi:10.1016/j.exer.2014.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Roubeix, C., Dominguez, E., Raoul, W., Guillonneau, X., Paques, M., Sahel, J-A., et al. (2019). Mo-derived perivascular macrophage recruitment protects against endothelial cell death in retinal vein occlusion. J. Neuroinflammation 16 (1), 157. doi:10.1186/s12974-019-1547-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Sennlaub, F., Auvynet, C., Bertrand, C., Lavalette, S., Poupel, L., Hu, S. J., et al. (2013). 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. 5 (11), 1775–1793. doi:10.1002/emmm.201302692

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, A., Subhi, Y., Krogh Nielsen, M., Falk, M. K., Hyldig Matzen, S. M., Finn, S., et al. (2017). Systemic frequencies of T helper 1 and T helper 17 cells in patients with age-related macular degeneration: A case-control study. Sci. Rep. 7 (1), 605. doi:10.1038/s41598-017-00741-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Solomon, S. D., Lindsley, K., Vedula, S. S., Krzystolik, M. G., and Hawkins, B. S. (2019). Anti-vascular endothelial growth factor for neovascular age-related macular degeneration. Cochrane Database Syst. Rev. 3 (3), CD005139. doi:10.1002/14651858.CD005139.pub4

PubMed Abstract | CrossRef Full Text | Google Scholar

Soulas, C., Donahue, R. E., Dunbar, C. E., Persons, D. A., Alvarez, X., and Williams, K. C. (2009). Genetically modified CD34+ hematopoietic stem cells contribute to turnover of brain perivascular macrophages in long-term repopulated primates. Am. J. Pathology 174 (5), 1808–1817. doi:10.2353/ajpath.2009.081010

PubMed Abstract | CrossRef Full Text | Google Scholar

Swayze, A., Kurzlechner, L., Sexton, M., Proia, A. D., Degan, S., and Lad, E. M. (2022). Monocyte biomarkers of early-intermediate age-related macular degeneration. Investigative Ophthalmol. Vis. Sci. 63 (7), 362–F0193.

Google Scholar

Tebbe, L., Kakakhel, M., Makia, M. S., Al-Ubaidi, M. R., and Naash, M. I. (2020). The interplay between peripherin 2 complex formation and degenerative retinal diseases. Cells 9 (3), 784. doi:10.3390/cells9030784

PubMed Abstract | CrossRef Full Text | Google Scholar

Trial, J., Cieslik, K. A., Haudek, S. B., Duerrschmid, C., and Entman, M. L. (2013). Th1/M1 conversion to th2/m2 responses in models of inflammation lacking cell death stimulates maturation of monocyte precursors to fibroblasts. Front. Immunol. 4, 287. doi:10.3389/fimmu.2013.00287

PubMed Abstract | CrossRef Full Text | Google Scholar

Velilla, S., García-Medina, J. J., García-Layana, A., Dolz-Marco, R., Pons-Vázquez, S., Pinazo-Durán, M. D., et al. (2013). Smoking and age-related macular degeneration: Review and update. J. Ophthalmol. 2013, 895147. Epub PMID: 24368940; PMCID: PMC3866712. doi:10.1155/2013/895147

PubMed Abstract | CrossRef Full Text | Google Scholar

Vivier, E., Elena, T., Baratin, M., Walzer, T., and Ugolini, S. (2008). Functions of natural killer cells. Nat. Immunol. 9 (5), 503–510. doi:10.1038/ni1582

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J. M., Janice, X., Ong, P. L. N., Fawzi, A. A., and Lavine, J. A. (2022). Macrophage-like cells are still detectable on the retinal surface after posterior vitreous detachment. Sci. Rep. 12 (1), 12864. doi:10.1038/s41598-022-17229-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Zhou, X., Yin, Y., Mai, Y., Wang, D., and Zhang, X. (2018). Hyperglycemia induces neutrophil extracellular traps formation through an NADPH oxidase-dependent pathway in diabetic retinopathy. Front. Immunol. 9, 3076. doi:10.3389/fimmu.2018.03076

PubMed Abstract | CrossRef Full Text | Google Scholar

Wynn, T. A., Chawla, A., and Pollard, J. W. (2013). Macrophage biology in development, homeostasis and disease. Nature 496 (7446), 445–455. doi:10.1038/nature12034

PubMed Abstract | CrossRef Full Text | Google Scholar

Xue, , Cui, J., Gao, L. Q., Zhang, C., Dou, H. L., Chen, D. N., et al. (2021). Peripheral monocyte count and age-related macular degeneration. The tongren Health care study. Am. J. Ophthalmol. 227, 143–153. doi:10.1016/j.ajo.2021.03.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, J. M., SungMoon, Y., JooLee, Y., Agalliu, D., DongYon, K., and Lee, S. W. (2022). COVID-19 morbidity and severity in patients with age-related macular degeneration: A Korean nationwide cohort study. Am. J. Ophthalmol. 239, 159–169. doi:10.1016/j.ajo.2021.05.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Q., Wang, G., and Zhang, F. (2020). Role of peripheral immune cells-mediated inflammation on the process of neurodegenerative diseases. Front. Immunol. 11, 582825. doi:10.3389/fimmu.2020.582825

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, X., Zhao, L., MariaCampos, M., Abu-Asab, M., Ortolan, D., Nathan, H., et al. (2020). CSF1R blockade induces macrophage ablation and results in mouse choroidal vascular atrophy and RPE disorganization. eLife 9, e55564. doi:10.7554/eLife.55564

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, Y., Yin, X., Chen, C., and Xing, Y. (2021). Identification of diagnostic biomarkers and their correlation with immune infiltration in age-related macular degeneration. Diagn. (Basel, Switz. 11 (6), 1079. doi:10.3390/diagnostics11061079

CrossRef Full Text | Google Scholar

Keywords: neuroinflammation, age-related macular degeneration (AMD), neurodegeneration, neuroimmune biology, innate immunity

Citation: Ascunce K, Dhodapkar RM, Huang D and Hafler BP (2023) Innate immune biology in age-related macular degeneration. Front. Cell Dev. Biol. 11:1118524. doi: 10.3389/fcell.2023.1118524

Received: 07 December 2022; Accepted: 06 February 2023;
Published: 28 February 2023.

Edited by:

Yiguo Qiu, Dana-Farber Cancer Institute and Harvard Medical School, United States

Reviewed by:

Wenxin Ma, National Eye Institute (NIH), United States
Chang He, Zhongshan Ophthalmic Center, Sun Yat-sen University, China

Copyright © 2023 Ascunce, Dhodapkar, Huang and Hafler. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Brian P. Hafler, brian.hafler@yale.edu

Download