Next Article in Journal
Review on Microbially Influenced Concrete Corrosion
Next Article in Special Issue
Recent Advances in Diagnosis and Treatment Approaches in Fungal Keratitis: A Narrative Review
Previous Article in Journal
Early Fluvoxamine Reduces the Risk for Clinical Deterioration in Symptomatic Outpatients with COVID-19: A Real-World, Retrospective, before–after Analysis
Previous Article in Special Issue
Infectious Eye Diseases and Prevention Control
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Host–Pathogen Interplay: A Tale of Two Stories within the Cornea and Posterior Segment

by
Michael P. Dempsey
1 and
Christopher D. Conrady
1,2,*
1
Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
2
Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(8), 2074; https://doi.org/10.3390/microorganisms11082074
Submission received: 5 June 2023 / Revised: 10 August 2023 / Accepted: 10 August 2023 / Published: 12 August 2023
(This article belongs to the Special Issue Infectious Eye Diseases and Prevention Control)

Abstract

:
Ocular infectious diseases are an important cause of potentially preventable vision loss and blindness. In the following manuscript, we will review ocular immunology and the pathogenesis of herpesviruses and Pseudomonas aeruginosa infections of the cornea and posterior segment. We will highlight areas of future research and what is currently known to promote bench-to-bedside discoveries to improve clinical outcomes of these debilitating ocular diseases.

1. Introduction

The eye is a complex sensory organ directly exposed to the environment and responsible for converting light into electrical signals and eventually images interpreted by the brain. Local tissue changes associated with inflammation and infection can reduce eye function and visual acuity. In fact, an inflammatory event in one eye can affect the function of the clinically “normal” contralateral eye [1]. Complications from ocular infectious diseases are a significant cause of visual impairment, most notably with rates worldwide of 1.4% of total cases of blindness due to corneal opacification from Chlamydia trachomatis alone [2]. This results in estimates as high as 5.5 million people who are bilaterally blind from the disease [3]. Within the developed world, herpes simplex viruses are a common pathogen and the leading cause of corneal blindness [4]. As such, infectious diseases of the eye are a significant global burden, whether they be from corneal opacification from trachoma or herpetic keratitis or damage to other structures of the eye such as the retina from Toxoplasmosis gondii. Blindness from any cause is more common than previously thought and is estimated to cause a loss in productivity of over USD 400 billion worldwide [5,6] The socioeconomic and psychosocial burden of blindness to a patient, family, and community are substantial with underlying infectious etiologies being potentially treatable, if not completely preventable, common causes of severe vision loss. Unfortunately, our understanding of the host–pathogen interaction is limited within the eye, inhibiting advances in treatment and the prevention of these sight-threatening infections.
Bacteria and viruses represent the more common causes of ocular infections worldwide and can affect nearly any anatomical part of the eye (Table 1 and Table 2). The overall geographic distribution and common bacterial isolates from ocular infections have been extensively reviewed elsewhere, but Staphylococcus aureus, Coagulase negative Staphylococci, Streptococcus pneumoniae, and Pseudomonas aeruginosa are the leading isolates of bacterial infections of the eye and adnexa (Table 1) [7]. While not specifically evaluated in a large meta-analysis, adenoviruses and herpesviruses are the most routinely encountered viruses within the eye (Table 2) [8]. However, there can be geographic variation in the most common ocular pathogens and rare ocular presentations from unlikely organisms [9,10,11,12,13]. This is most clearly seen with the geographic distribution of the three clades of Toxoplasmosis gondii worldwide and the very rare occurrence of Trypanosome cruzii-associated retinitis [9,14,15,16]. Within this review, we aim to highlight the known host–pathogen responses by comparing bacterial and viral responses within the cornea and posterior segment and emphasize areas of potential future research to identify therapeutic targets to limit vision loss from these infectious diseases.

2. Basics of Ocular Immunology

The eye is a complex sensory organ with parts of it being directly exposed to the surrounding environment with this interface providing an external surface for the establishment of a local microbiome. As such, the host response to infection of the eye is complex as the organ must rid itself of invading environmental pathogens while minimizing immunologically driven pathology from the self or the nonpathogenic microbiota. The combination of the tear film contents (e.g., IgA, Lactoferrin, defensins, and growth factors) and the continuous washing by tears produced by the lacrimal gland serve to remove infectious agents, limit uncontrolled and unregulated inflammatory damage, provide a physical barrier to the surrounding environment, and promote the healing of the ocular surface to reduce the risk of ocular infection [17]. These antimicrobial compounds and barriers alone, however, are insufficient to protect the eye from pathogen invasion. Relatively similar barriers can be found within the retina and posterior segment [18].
Once physical barriers are breached on or within the eye, innate immune mechanisms serve to rapidly identify the pathogen. While the overall immunological response of the eye may slightly differ compared to that of other sites, innate immune sensors are expressed throughout the eye [19,20,21,22,23]. Much like those found in other tissues, Toll-like receptors (TLRs) and other innate sensors recognize conserved pathogen-associated molecular patterns and activate Myeloid differentiation primary response protein (MyD)88-dependent and -independent pathways following complex interactions with other inflammatory receptors [24,25]. NF-κB-related pathways and/or type I interferon (IFN) pathways are preferentially activated by these innate immune sensors [22,26]. Activation of these pathways leads to local innate immune responses including the production of chemokines to facilitate leukocyte trafficking and priming of the more pathogen-specific adaptive immune response [26]. The interplay between many of these pathways will be further highlighted below regarding specific pathogens within distinct areas of the eye.
To further complicate matters, certain parts of the eye are considered “immune privileged” and respond immunologically in ways not seen in other tissues and organs. This is due in part to blood–aqueous and blood–retinal barriers that prohibit the egress of macromolecules into the eye, limiting the exposure of ocular tissue, including local innate immune cells, to circulating pathogens and toxins [27]. Retinal pigment epithelial cells also upregulate T regulatory responses and suppress effector T cell activation [28,29]. This ocular anti-inflammatory milieu with an atypical response known as anterior chamber-associated immune deviation (ACAID) is a phenomenon within the eye in which antigens injected into the anterior chamber induce tolerance whereas the host develops a muted or abolished systemic response to the antigen. While the process is not completely understood, the generation of antigen-specific, antigen-presenting B cells within the spleen following antigen exposure intracamerally results in the development of T regulatory cells and interleukin (IL)-10 production in a relatively immunosuppressive microenvironment within the anterior chamber [30,31,32,33]. γδ T cells and IFN-γ production also appear to be important in this response as their loss does not allow ACAID to develop [34,35]. Subretinal exposure to antigens has been shown to induce a similar response to that induced by ACAID [36]. While different cells, cytokines, and chemokines have all been implicated, ACAID is still not completely understood and it is unclear if this is simply a mouse phenomenon or if it is seen in humans.
The hallmark of immune privilege and immune deviation within the eye is epitomized by the exceptionally high rates of success of corneal transplantation compared to those of other tissues and organs despite the lack of preoperative HLA typing [37]. The success of corneal transplantation appears to be related to the unique nature of the eye in which physical and cellular barriers of the cornea, ACAID-induced by T regulatory cells, and the immunosuppressive microenvironment of the anterior chamber reduce inflammatory reactions, promote immune privilege, and thereby reduce graft failure [37,38,39,40]. However, immune privilege and corneal transplant rejection can be overridden by severing corneal nerves and this rejection process appears to be driven by minor H alloantigens and the loss of regulatory T cells [41,42,43]. Ocular immune privilege is tightly controlled and can be overcome with relatively innocuous changes to eye anatomy.

3. The Cornea

3.1. Viral Keratitis

Keratitis poses a major global public health concern and can lead to a partial or complete loss of vision due to corneal opacification. Risk factors include specific pathogens (e.g., bacteria, fungi, amebae, and viruses) and forms of exposure (e.g., eye trauma, chemical exposure, ultraviolet exposure, and contact lenses). Viruses are a major cause of infectious blindness worldwide and herpes simplex virus (HSV) type-1 and -2 and varicella zoster virus (VZV) are the most commonly identified viruses [22]. As such, we will focus the viral section of this review on herpesviruses due to space constraints and their visual importance. These viruses are double-stranded DNA viruses that utilize an outer envelope consisting of 16 membrane proteins [44]. These glycoproteins such as gB, gD, gH, and gL represent 12 of the 16 membrane proteins and interact with host cell surface receptors to facilitate the penetration and infection of the cell through either endocytosis or membrane fusion [45,46,47]. Heparan sulfate, through its interaction with gB and/or gC, enables the attachment of the virus to host cells, followed by the binding of viral gD with heparan sulfate or nectin-1 to enable capsid release into the cytoplasm [48]. The capsid is then transported to the nucleus for viral genome replication using host cell DNA polymerases. The new virions produced are released from the cell to infect neighboring cells, and the virus spreads. Primary HSV-1 infection is thought to occur in children or young adults through direct contact with infected mucous membranes. Within the cornea, HSV replication occurs predominately within epithelial cells and results in the clinical development of dendrites and epithelial keratitis. In most cases, this is hypothesized to represent the reactivation of the virus from the trigeminal ganglia.
HSV-1 infection of any tissue, as with many other viral infections, evokes a strong inflammatory response involving polymorphonuclear leukocytes, macrophages, natural killer cells, and B- and T-cells [49]. While these different cell types are recruited to the site of infection, the following question remains: are they all necessary or is there significant redundancy within the immune response? Further, are all innate responses to all pathogens the same in all tissues? We have previously studied the host immune response to HSV-1 and -2 infection of the cornea. In mouse models of herpetic epithelial keratitis, there are specific immunological responses that occur following viral infection of the corneal epithelium that are similar to those seen in other epithelial tissues such as the rapid induction of antiviral type I IFNs and recruitment of infiltrating leukocytes [50,51]. However, there are distinct differences in the innate immune response to HSV-1 in the cornea compared to that in other commonly affected sites such as the brain. For example, TLR-3 is critical within the central nervous system. A loss of signals emanating from TLR-3 within microglia render mice and children more susceptible to herpetic encephalitis due to the loss of type I IFN production [52,53,54,55].
In the cornea, we found that the loss of TLR signaling had no effect on viral containment [51]. Rather, the innate response to HSV-1 is driven by a TLR-independent, IFN-inducible protein-16 (p204 mouse homolog)-dependent process to activate type I IFN production and inhibit viral spread through the activation of antiviral proteins such as protein kinase R and 2′,5′-oligoadenylate synthetase (Figure 1) [51,56,57,58]. Additionally, this tightly choregraphed response activates dendritic cells, specific chemokine production, and the recruitment of protective macrophages to the site of infection to further inhibit viral spread [59,60]. Resident plasmacytoid dendritic cells (pDCs), potent producers of type I IFNs, residing in the anterior corneal stroma increase in density following inflammatory changes of the cornea, and their selective depletion results in worse corneal pathology, a reduction in type I IFN production, and HSV-1 dissemination [61]. Antigen-presenting CD11c+ dendritic cells traffic to the draining lymph node following exposure to HSV-1 to prime adaptive immunity [62,63,64]. Following this first wave of infiltrating macrophages and the pDC response, type I IFNs induce natural killer (NK) cells and HSV-specific CD8+ T cells to undergo maturation and expansion and the leukocytes are then recruited to the site of infection [60,65,66,67,68,69]. These infiltrating cells further inhibit viral proliferation and spread [63]. γδ T cells are also emerging as important recruited leukocytes in HSV defense within the cornea [68]. It is unclear what, if any, other leukocyte populations contribute to viral immunity or pathology in the cornea during this early response. Once the virus has been controlled and establishes latency, typically in the trigeminal ganglia, HSV-specific CD8+ T cells are critical in inhibiting reactivation [70,71,72,73,74].
If the earliest steps in this response are inadequate or aberrant, chemokines become dysregulated, immune cell trafficking is altered, and the virus disseminates [56,62,65]. This highly regulated process is best epitomized by comparing wild-type and knock-out mice lacking a functional type I IFN receptor as these mice are unable to respond to type I IFNs. Within these knock out mice, we found that viral loads are significantly higher locally and systemically, the development of encephalitis is rapid, there is a loss of draining lymph node integrity, and there is aberrant chemokine production, specifically a loss of CCL2 and CXCL10 and an overproduction of CXCL1, within the first few days after infection with HSV-1 when compared to wild-type controls [51,53,56,59,62,65]. This resulted in a loss of infiltrating monocytes and the primary source of the antiviral compound nitric oxide [59,75,76,77,78]. The importance of macrophages early in HSV-1 infection of the cornea has been corroborated by others [79,80].
Late in the disease’s process, lymphangiogenesis is a common complication of herpetic infections of the cornea. Vessel ingrowth has been shown to be driven by the infected cell protein-4 of HSV-1 which binds directly to the vascular endothelial growth factor (VEGF)-A promoter to induce corneal neovascularization [81,82]. Further, an imbalance between the anti-neovascular soluble VEGF receptor-1 and VEGF develops during infection, promoting neovascularization [83]. Multiple immune cell types including CD4+ and CD8+ T cells and neutrophils produce VEGF subtypes and increase the risk of corneal graft failure [63,84,85]. The host immune response to the virus, specifically infiltrating antigen-specific and nonspecific CD4+ T cells that arrive within 7 days post-infection, has been implicated in the development of corneal opacification during stromal keratitis, another late but common complication of herpetic keratitis [86,87]. In further support of these findings, mice lacking T cells do not develop stromal lesions and the adoptive transfer of CD4+ T cells into these same mice restores lesion formation [88,89]. This is supported by the clinical use of topical corticosteroids during herpetic stromal keratitis to quell disease [90]. These topical corticosteroids likely inhibit the function of infiltrating neutrophils as well as other cell types that have been shown to damage the cornea by producing proinflammatory compounds including metalloproteinases and VEGF [91,92]. An intact complement pathway has been shown to be a potent inhibitor of these late complications due to the development of tolerance mediated by iC3b [93,94]. Thus, it is clear that there is a critical antiviral response and also a nonprotective, locally destructive response to HSV-1 infection within the cornea.

3.2. Bacterial Keratitis

The pathogenesis and immune reaction to herpetic keratitis contrasts that of bacterial infection of the cornea, or bacterial keratitis (BK). BK is a serious, vision-threatening eye infection that may lead to permanent vision loss due to corneal scarring, even among treated patients, or in rare cases, the development of endophthalmitis [95]. There is a risk of corneal perforation from extreme thinning and extension into surrounding tissues is possible. This ophthalmic infection requires prompt treatment to reduce the development of these ocular complications. Predisposing risk factors for BK include contact lens wear, previous topical steroid use, ocular surface disease (OSD), ocular trauma, previous keratitis, prior surgery, and ongoing corneal disease [96,97,98,99].
Pseudomonas aeruginosa is the most common causative agent in cases of BK associated with contact lens use [100]. Corneal infections with P. aeruginosa often have poor clinical outcomes and can result in long and costly treatments. Due to its ocular importance and representative preclinical models, we will focus our BK discussion on P. aeruginosa. Much like many other pathogens, P. aeruginosa requires a compromised ocular surface to penetrate and infect the cornea. Contact lens use increases the susceptibility of the cornea to infection due to several factors, mostly by causing a compromised ocular surface. These factors include a relative hypoxic state, ocular trauma, microtrauma to the corneal epithelium, herpetic keratitis, an immunocompromised state, and improper eye hygiene, which are related to or risks amplified by contact lens use [101]. Once P. aeruginosa is introduced and/or internalized by a compromised host epithelium, the pathogen is able to invade the cornea [102]. During the infection process, the pathogen exploits its large genome, encoding complex regulatory networks and a wide range of virulence factors, including motility and the secretion of various proteases and toxins [101]. There is increasing evidence that multidrug-resistant infections are associated with a phylogenetic subgroup of P. aeruginosa isolates carrying the gene encoding the potent cytotoxin exotoxin U, one of two mutually exclusive exotoxins secreted via the type III secretion system, and may be associated with worse clinical outcomes [103,104,105,106]. The bacterium has developed mechanisms to evade innate immune recognition [107]. Consequently, P. aeruginosa has been and will be an important and difficult cause of BK to treat within ophthalmology clinics due to emerging pathogen virulence factors and evasion mechanisms.
The host response to P. aeruginosa in the cornea is complex. In a mouse model of P. aeruginosa-associated keratitis, a loss of signals emanating from the MyD88 adaptor protein resulted in uncontrolled bacterial replication and spread; however, the loss of neutrophil recruitment resulted in a less severe corneal pathology in these same mice [108]. These findings emphasized the role of neutrophils in the cornea to contain the pathogen but at a cost of worsening local tissue destruction. This was supported with the knockdown of TLR-9 expression resulting in higher bacterial loads but fewer infiltrating polymorphonuclear leukocytes/neutrophils (PMNs) and corneal perforations, reduced inflammatory chemokine concentrations, and clearer corneas compared to controls [109]. Interestingly, the loss of TLR-4 in these infections reduced bacterial defense but also led to increased mRNA expression of proinflammatory cytokines, more infiltrating neutrophils, and higher rates of corneal perforation [110]. Thus, TLR-4 and -9 appear to have roles in bacterial containment but diverging roles in the development of a proinflammatory milieu. Pretreatment of the cornea with the TLR-5 agonist, flagellin, a known innate immune sensor of P. aeruginosa, through an interferon regulatory factor (IRF)-1-dependent pathway, reduces susceptibility to infection (Figure 2) [111,112]. Taken together, TLR activation within the cornea has protective and destructive implications within the cornea. It is unclear if each of these TLRs regulate specific responses or are redundant, activating similar downstream pathways.
Appropriate numbers of neutrophils appear to be an important leukocyte population in BK due to the production of several antibacterial compounds or responses from these cells [113]. Neutrophil extracellular trap formation (“dead zones”), activation of the transcription factor hypoxia inducible factor-1α, and the production of reactive oxygen species from PMNs inhibit P. aeruginosa spread beyond the outermost corneal layers [114,115,116]. PMNS are recruited to the cornea by macrophage inflammatory protein (MIP)-2, and the neutralization of this chemokine resulted in fewer infiltrating PMNs and less corneal damage but no change in bacterial counts (Figure 2) [117] An alarmin, high-mobility group box 1 (HMGB1), induces proinflammatory signals including the production of MIP-2 during these infections and can be downregulated by vasoactive intestinal peptide [118,119]. The production of MIP-2 to recruit is tightly regulated by IL-1 and the prolonged production of IL-1 results in PMN persistence, increased tissue destruction, and corneal perforation [117,120]. The regulation of MIP-2 by IL-1 has been extensively reviewed previously and their role in PMN infiltration during BK is critical in both pathogen clearance and the development of corneal pathology [113].
Innate immune activation and the recruitment of early infiltrating immune cells prime the adaptive response following BK. Activated CD4+ and CD8+ T cells are recruited into the cornea by MIP-1α by day 5 post-infection [121]. Unfortunately, a predominantly Th1 CD4+ T cell response in C57Bl/6J mice is associated with much higher rates of corneal perforation and higher bacterial counts than that seen with a Th2-driven response in BALB/c mice [121]. Further, activated T cells have been shown to enhance the recruitment of PMNs to the site of infection and PMN persistence [113,122]. Consequently, a Th1 response appears to be detrimental to the host defense response and local tissue integrity. The precise role of a Th2 response within the cornea to P. aeruginosa is not well-elucidated. However, exogenous IL-36α can overcome the Th1 signals in C57Bl/6J mice and drive a Th2 response, and conversely, IL-17 inhibits Th2 cytokine production [123,124]. Similarly to what is observed with viral infections of the cornea, an innate and adaptive immune response to bacterial infection is required to contain the pathogen but can cause local tissue pathology.

4. Infections of the Posterior Segment

4.1. Viral Retinitis

While rare, herpes viruses can affect the posterior segment as well. HSV-1, -2, and VZV are the most common causes of acute retinal necrosis (ARN), a rapidly progressive infection of the retina leading to severe vision loss and high rates of visual complications in children and adults [125,126,127]. There is broad upregulation of inflammatory markers within the eye in humans with the disease [128,129]. Mouse models of the disease have been created to simulate ARN with the most defined model, the von Szily model, one in which intracameral injection of HSV-1 into BALB/c mice results in anterior chamber inflammation in the injected eye and eventually the development of retinitis in the contralateral eye due to two waves of the virus invading the retina and a delayed hypersensitivity reaction [130,131,132]. With this model, there is broad upregulation of genes and inflammatory compounds within the posterior segment and neutrophils appear to inhibit the development of retinitis [133,134,135]. Tumor necrosis factor (TNF)-α, a proinflammatory cytokine and major regulator of inflammation, is upregulated, and its neutralization reduces retinal pathology [133,136]. Within areas of retinitis, infiltrating T cells, activated Muller cells, retinal pigment epithelial cells, PMNS, and macrophages can be found [133,137]. It is unclear what protective or pathology-causing role these local or infiltrating immune cells and inflammatory chemokines and cytokines have on the pathogenesis of ARN. However, the broad cellular infiltrate supports the broad upregulation of immune pathways and a robust local inflammatory milieu.
While we have learned much about both innate and adaptive immunity from the von Szily model, we felt there were limitations with the model in the study of innate immunity. These limitations include a lack of mouse immune knock-outs on a BALB/c background, the imprecise timing of retinal infection, and the variance from the typical clinical presentation (e.g., preceding inflammation in the contralateral eye is exceedingly rare) [138]. To directly infect the retina and create a novel mouse model more reminiscent of human disease to study innate immunity, our lab began utilizing an adapted mouse model of retinal detachments that had been previously used in rabbits [139,140]. Rather than injecting viscoelastic, our lab injects HSV-1 directly into the subretinal space of C57Bl/6J mice through a focal retinotomy, and after several days, the subretinal bleb resorbs and the mouse develops focal viral retinitis similar to that seen in humans [141]. We have found that much like in other tissues, type I IFNs and downstream antiviral responses are critical in containing the virus as mice lacking a functional type I IFN receptor have worse local tissue destruction, higher viral titers, and dissemination of the virus into the brain leading to mouse demise by day 5 post-infection, which is not seen in wild-type controls [51,56,62,141,142]. While the innate response within these mice and their extreme susceptibility to viral infection of the retina is not completely understood, there appear to be changes in the cell death pathways and chemokines produced compared to those in wild-type mice and controls [141]. While retinal infections with cytomegalovirus appear and progress very differently from how ARN does clinically, cell death pathways are also broadly upregulated and a loss of caspase-1, a mediator of programmed cell death, results in atypical retinal disease [143,144]. In other neuronal tissues, cell death pathways also play an important role in limiting herpetic encephalitis [145]. Within ARN, it is unclear what activation of these pathways is regulating, minimizing, or worsening in the disease’s process. We hypothesize that we will be able to identify potential therapeutic targets within cell death and/or innate immune pathways that can be targeted to reduce local tissue destruction while retaining viral control, improving clinical outcomes with our mouse model. Despite these two mouse models of disease, our understanding of innate and adaptive immunity to HSV-1 within the retina is limited, thus inhibiting advances in other ocular infectious and noninfectious diseases in which the innate immune system has been implicated or is required (e.g., diabetic retinopathy and age-related macular degeneration).

4.2. Bacterial Endophthalmitis

While bacterial infections of the retina are rare, bacterial endophthalmitis is one of the most devastating complications of any intraocular procedure, and despite the current strategies for prevention, early identification, and aggressive treatment, substantial vision loss still frequently occurs [12,146,147,148,149,150]. Endophthalmitis can more rarely be associated with blood-borne infections (endogenous), or more commonly, following trauma or surgery to the eye (exogenous). Several bacterial organisms that cause endophthalmitis, such as Streptococcus pneumonaie, P. aeruginosa or other gram (-) organisms, have been associated with exceptionally poor visual outcomes and higher rates of evisceration or enucleation [12,151,152]. Fortunately, gram (-) organisms are rare and gram (+), coagulase-negative bacteria are the most commonly identified pathogens [148,153]. Due to the higher frequency of gram (+) organisms, we will focus this portion of the review on immunity to bacterial infections of the posterior segment from gram (+) organisms.
Once relatively high doses of Staphylococcus are introduced into the posterior segment via trauma or following an intraocular procedure, cell wall components such as peptidoglycan and lipoteichoic acid induce the production of proinflammatory chemokines and cytokines such as IL-6, TNF-α, CXCL-1, and MIP-2, while the bacterial toxins upregulate IL-1β [154,155]. Bacterial loads and inflammatory markers such as IL-1β, TNF-α, and CXCL1 peak within mice within 24 h of inoculation [155]. These inflammatory markers are also elevated in Staphylococcus epidermidis and Streptococcus pneumonia endophthalmitis very early on, leading to increased vascular permeability [154,155,156]. The early bacterial and inflammatory changes within the eye are associated with a 50% reduction in electroretinogram (ERG) amplitude in mice and the development of a local pathology within 12–24 h of infection that persists until at least 72 h but likely lasts much longer based on poor clinical outcomes [148,155,157]. Various innate immune sensors have been implicated in bacterial defense including TLR-dependent and -independent pathways within virtually all cell types within the retina and retinal pigment epithelium [20,21,158,159]. These sensors are upregulated following infection for unclear reasons but we hypothesize that they could accentuate immunopathology with their subsequent activation [155]. The NLRP3 inflammasome appears to regulate IL-1 production within PMNs and microglia, and the sensors’ dysfunction results in higher bacterial loads and worse retinal tissue damage [21,160]. Mice unable to produce CXCL1 have an attenuated and delayed development of intraocular inflammation and ERG changes despite the lack of differences in bacterial load compared to that of controls at multiple time points [157]. S. aureus produces several toxins that have been shown to induce host tissue changes including within the eye [161,162]. The neutralization of these toxins with nanospores improves retinal function (higher A- and B-wave retention) despite the similar bacterial loads of untreated but infected control mice [163]. Additionally, strains deficient in certain toxins have been shown to be associated with less retinal damage [162]. Taken together, these findings suggest that a higher bacterial burden, toxin production, and enhanced host inflammation are responsible for the development of retinal pathology. Thus, our understanding of immunity and neuropathology to bacterial endophthalmitis is in its infancy but may yield important clues and identify potential therapeutic targets, mitigate retinal pathology, and promote visual rehabilitation. What is readily apparent is that clinical treatments and interventions must be performed early on in the disease’s trajectory as pathological and ERG changes are seen within 12–24 h after infection in mice [155,157].

5. Conclusions: The Yin and Yang of Immunity in the Eye

The innate and adaptive immune responses within the eye are important inhibitors of pathogen dissemination. However, various local or infiltrating cell types have been implicated in the pathogen response but may also contribute to local tissue destruction. This has been partially abrogated clinically with the use of corticosteroids to inhibit inflammation and minimize the development of tissue pathology during many noninfectious and infectious diseases of the eye once appropriate antimicrobials have been initiated or the course is completed. This is best epitomized by the frequent need for corticosteroids in the treatment of Toxoplasma gondii chorioretinitis and herpetic stromal keratitis [90,164]. Systemic corticosteroids serve as broad, nonspecific immune suppressors but with many side effects. While downregulating an immune response is likely necessary to minimize long-term retinal pathology, there must be a clear identification of critical pathways in pathogen clearance and neuropathogenesis to create more specific immune modulators. To emphasize this point, an aggressive form of viral retinitis, progressive outer retinal necrosis, develops typically only in the immune-suppressed, so a broad downregulation of the immune system is not ideal. Targeted therapies will allow the clearance of the pathogen but minimize local tissue damage.
Lastly, responses within the cornea and posterior segment are similar in some respects but differ in others. Thus, findings in the cornea cannot be completely extrapolated to the posterior segment such as the retina. While this is not surprising due to the different embryological origins of ocular tissues, the diverse embryological origins, and differing responses of tissues within the eye potentially limit findings and advances from one segment to another. The inability to potentially extrapolate findings emphasizes the need for continued research in the ocular immunology, virology, and bacteriology of all segments of the eye.

Author Contributions

Conceptualization, M.P.D. and C.D.C.; writing—original draft preparation, M.P.D. and C.D.C.; writing—review and editing, M.P.D. and C.D.C.; funding acquisition, C.D.C. All authors have read and agreed to the published version of the manuscript.

Funding

CDC was supported by the Knights Templar Eye Foundation career starter grant, the IDeA Clinical and Translational Research Early Career Investigator Program, startup funds provided by the University of Nebraska Medical Center, and the National Eye Institute for a K08 career development award (EY034892) to support ongoing work.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Azarmina, M.; Soheilian, M.; Ahmadieh, H.; Azarmina, H. Electroretinogram changes in the sound eye of subjects with unilateral necrotizing herpetic retinitis. J. Ophthalmic Vis. Res. 2014, 9, 195–203. [Google Scholar] [PubMed]
  2. Burton, M.J.; Mabey, D.C.W. The Global Burden of Trachoma: A Review. PLoS Neglected Trop. Dis. 2009, 3, e460. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, E.; Kong, X.; Wolle, M.; Gasquet, N.; Ssekasanvu, J.; Mariotti, S.P.; Bourne, R.; Taylor, H.; Resnikoff, S.; West, S. Global trends in blindness and vision impairment due to corneal opacity 1984–2020: A meta-analysis. Ophthalmology 2023, 2022, 8828358. [Google Scholar] [CrossRef]
  4. Liesegang, T.J.; Melton, L.J., III; Daly, P.J.; Ilstrup, D.M. Epidemiology of Ocular Herpes Simplex: Incidence in Rochester, Minn, 1950 Through 1982. Arch. Ophthalmol. 1989, 107, 1155–1159. [Google Scholar] [CrossRef] [PubMed]
  5. Marques, A.P.; Ramke, J.; Cairns, J.; Butt, T.; Zhang, J.H.; Muirhead, D.; Jones, I.; Tong, B.A.M.A.; Swenor, B.K.; Faal, H.; et al. Global economic productivity losses from vision impairment and blindness. eClinicalMedicine 2021, 35, 100852. [Google Scholar] [CrossRef]
  6. Flaxman, A.D.; Wittenborn, J.S.; Robalik, T.; Gulia, R.; Gerzoff, R.B.; Lundeen, E.A.; Saaddine, J.; Rein, D.B. Vision and Eye Health Surveillance System study group Prevalence of Visual Acuity Loss or Blindness in the US: A Bayesian Meta-analysis. JAMA Ophthalmol. 2021, 139, 717–723. [Google Scholar] [CrossRef]
  7. Teweldemedhin, M.; Gebreyesus, H.; Atsbaha, A.H.; Asgedom, S.W.; Saravanan, M. Bacterial profile of ocular infections: A systematic review. BMC Ophthalmol. 2017, 17, 212. [Google Scholar] [CrossRef] [Green Version]
  8. Petrillo, F.; Petrillo, A.; Sasso, F.P.; Schettino, A.; Maione, A.; Galdiero, M. Viral Infection and Antiviral Treatments in Ocular Pathologies. Microorganisms 2022, 10, 2224. [Google Scholar] [CrossRef]
  9. Conrady, C.D.; Hanson, K.E.; Mehra, S.; Carey, A.; Larochelle, M.; Shakoor, A. The First Case of Trypanosoma cruzi-Associated Retinitis in an Immunocompromised Host Diagnosed With Pan-Organism Polymerase Chain Reaction. Clin. Infect. Dis. 2018, 67, 141–143. [Google Scholar] [CrossRef]
  10. Fashina, T.; Huang, Y.; Thomas, J.; Conrady, C.D.; Yeh, S. Ophthalmic Features and Implications of Poxviruses: Lessons from Clinical and Basic Research. Microorganisms 2022, 10, 2487. [Google Scholar] [CrossRef]
  11. Conrady, C.D.; Faia, L.J.; Gregg, K.S.; Rao, R.C. Coronavirus-19-Associated Retinopathy. Ocul. Immunol. Inflamm. 2021, 29, 675–676. [Google Scholar] [CrossRef] [PubMed]
  12. Conrady, C.D.; Demirci, H.; Wubben, T.J. Retinal Whitening After Lung Transplant for Cystic Fibrosis. JAMA Ophthalmol. 2020, 138, 994–995. [Google Scholar] [CrossRef] [PubMed]
  13. Conrady, C.D.; Feist, R.M.; Crum, A. Shingles as the underlying cause of orbital myositis in an adolescent: A case report. Am. J. Ophthalmol. Case Rep. 2017, 5, 111–113. [Google Scholar] [CrossRef] [PubMed]
  14. Pappas, G.; Roussos, N.; Falagas, M.E. Toxoplasmosis snapshots: Global status of Toxoplasma gondii seroprevalence and implications for pregnancy and congenital toxoplasmosis. Int. J. Parasitol. 2009, 39, 1385–1394. [Google Scholar] [CrossRef]
  15. Desai, R.J.; Solomon, D.H.; Shadick, N.; Iannaccone, C.; Kim, S.C. Identification of smoking using Medicare data--A validation study of claims-based algorithms. Pharmacoepidemiol. Drug Saf. 2016, 25, 472–475. [Google Scholar] [CrossRef] [Green Version]
  16. Khan, A.; Jordan, C.; Muccioli, C.; Vallochi, A.L.; Rizzo, L.V.; Belfort, R., Jr.; Vitor, R.W.A.; Silveira, C.; Sibley, L.D. Genetic divergence of Toxoplasma gondii strains associated with ocular toxoplasmosis, Brazil. Emerg. Infect. Dis. 2006, 12, 942–949. [Google Scholar] [CrossRef]
  17. Conrady, C.D.; Joos, Z.P.; Patel, B.C.K. Review: The Lacrimal Gland and Its Role in Dry Eye. J. Ophthalmol. 2016, 2016, 7542929. [Google Scholar]
  18. Du, Y.; Yan, B. Ocular immune privilege and retinal pigment epithelial cells. J. Leukoc. Biol. 2023, 113, 288–304. [Google Scholar] [CrossRef]
  19. Jin, X.; Qin, Q.; Chen, W.; Qu, J. Expression of Toll-Like Receptors in the Healthy and Herpes Simplex Virus-Infected Cornea. Cornea 2007, 26, 847–852. [Google Scholar] [CrossRef]
  20. Kumar, M.V.; Nagineni, C.N.; Chin, M.S.; Hooks, J.J.; Detrick, B. Innate immunity in the retina: Toll-like receptor (TLR) signaling in human retinal pigment epithelial cells. J. Neuroimmunol. 2004, 153, 7–15. [Google Scholar] [CrossRef]
  21. Singh, P.K.; Kumar, A. Retinal Photoreceptor Expresses Toll-like Receptors (TLRs) and Elicits Innate Responses Following TLR Ligand and Bacterial Challenge. PLoS ONE 2015, 10, e0119541. [Google Scholar] [CrossRef] [Green Version]
  22. Bryant-Hudson, K.; Conrady, C.D.; Carr, D.J.J. Type I interferon and lymphangiogenesis in the HSV-1 infected cornea—Are they beneficial to the host? Prog. Retin. Eye Res. 2013, 36, 281–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Song, P.I.; Abraham, T.A.; Park, Y.; Zivony, A.S.; Harten, B.; Edelhauser, H.F.; Ward, S.L.; Armstrong, C.A.; Ansel, J.C. The Expression of Functional LPS Receptor Proteins CD14 And Toll-like Receptor 4 in Human Corneal Cells. Investig. Ophthalmol. Vis. Sci. 2001, 42, 2867–2877. [Google Scholar]
  24. Huang, X.; Hazlett, L.D. Analysis of Pseudomonas aeruginosa Corneal Infection Using an Oligonucleotide Microarray. Investig. Ophthalmol. Vis. Sci. 2003, 44, 3409–3416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yu, F.-S.X.; Hazlett, L.D. Toll-like Receptors and the Eye. Investig. Ophthalmol. Vis. Sci. 2006, 47, 1255–1263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Conrady, C.D.; Drevets, D.A.; Carr, D.J.J. Herpes simplex type I (HSV-1) infection of the nervous system: Is an immune response a good thing? J. Neuroimmunol. 2010, 220, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Enzmann, V.; Row, B.W.; Yamauchi, Y.; Kheirandish, L.; Gozal, D.; Kaplan, H.J.; McCall, M.A. Behavioral and anatomical abnormalities in a sodium iodate-induced model of retinal pigment epithelium degeneration. Exp. Eye Res. 2006, 82, 441–448. [Google Scholar] [CrossRef]
  28. Horie, S.; Sugita, S.; Futagami, Y.; Yamada, Y.; Mochizuki, M. Human retinal pigment epithelium-induced CD4+CD25+ regulatory T cells suppress activation of intraocular effector T cells. Clin. Immunol. 2010, 136, 83–95. [Google Scholar] [CrossRef]
  29. Sugita, S.; Horie, S.; Nakamura, O.; Futagami, Y.; Takase, H.; Keino, H.; Aburatani, H.; Katunuma, N.; Ishidoh, K.; Yamamoto, Y.; et al. Retinal Pigment Epithelium-Derived CTLA-2α Induces TGFβ-Producing T Regulatory Cells12. J. Immunol. 2008, 181, 7525–7536. [Google Scholar] [CrossRef] [Green Version]
  30. Ashour, H.M.; Niederkorn, J.Y. Peripheral Tolerance Via the Anterior Chamber of the Eye: Role of B Cells in MHC Class I and II Antigen Presentation1. J. Immunol. 2006, 176, 5950–5957. [Google Scholar] [CrossRef] [Green Version]
  31. Skelsey, M.E.; Mayhew, E.; Niederkorn, J.Y. Splenic B Cells Act as Antigen Presenting Cells for the Induction of Anterior Chamber-Associated Immune Deviation. Investig. Ophthalmol. Vis. Sci. 2003, 44, 5242–5251. [Google Scholar] [CrossRef] [Green Version]
  32. Skelsey, M.E.; Mayhew, E.; Niederkorn, J.Y. CD25+, interleukin-10-producing CD4+ T cells are required for suppressor cell production and immune privilege in the anterior chamber of the eye. Immunology 2003, 110, 18–29. [Google Scholar] [CrossRef]
  33. Streilein, J.W. Ocular immune privilege: Therapeutic opportunities from an experiment of nature. Nat. Rev. Immunol. 2003, 3, 879–889. [Google Scholar] [CrossRef]
  34. Skelsey, M.E.; Mellon, J.; Niederkorn, J.Y. γδ T Cells Are Needed for Ocular Immune Privilege and Corneal Graft Survival1. J. Immunol. 2001, 166, 4327–4333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Paunicka, K.; Chen, P.W.; Niederkorn, J.Y. Role of IFN-γ in the establishment of anterior chamber-associated immune deviation (ACAID)-induced CD8+ T regulatory cells. J. Leukoc. Biol. 2012, 91, 475–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Wenkel, H.; Streilein, J.W. Analysis of immune deviation elicited by antigens injected into the subretinal space. Investig. Ophthalmol. Vis. Sci. 1998, 39, 1823–1834. [Google Scholar]
  37. Hori, J.; Yamaguchi, T.; Keino, H.; Hamrah, P.; Maruyama, K. Immune privilege in corneal transplantation. Prog. Retin. Eye Res. 2019, 72, 100758. [Google Scholar] [CrossRef] [PubMed]
  38. Niederkorn, J.Y. Corneal Transplantation and Immune Privilege. Int. Rev. Immunol. 2013, 32, 57–67. [Google Scholar] [CrossRef] [Green Version]
  39. Cunnusamy, K.; Chen, P.W.; Niederkorn, J.Y. IL-17A–Dependent CD4+CD25+ Regulatory T Cells Promote Immune Privilege of Corneal Allografts. J. Immunol. 2011, 186, 6737–6745. [Google Scholar] [CrossRef] [Green Version]
  40. Toda, M.; Ueno, M.; Yamada, J.; Hiraga, A.; Asada, K.; Hamuro, J.; Sotozono, C.; Kinoshita, S. Quiescent innate and adaptive immune responses maintain the long-term integrity of corneal endothelium reconstituted through allogeneic cell injection therapy. Sci. Rep. 2022, 12, 18072. [Google Scholar] [CrossRef]
  41. Neelam, S.; Niederkorn, J.Y. Corneal Nerve Ablation Abolishes Ocular Immune Privilege by Downregulating CD103 on T Regulatory Cells. Investig. Ophthalmol. Vis. Sci. 2020, 61, 25. [Google Scholar] [CrossRef]
  42. Paunicka, K.J.; Mellon, J.; Robertson, D.; Petroll, M.; Brown, J.R.; Niederkorn, J.Y. Severing Corneal Nerves in One Eye Induces Sympathetic Loss of Immune Privilege and Promotes Rejection of Future Corneal Allografts Placed in Either Eye. Am. J. Transplant. 2015, 15, 1490–1501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Haskova, Z.; Sproule, T.J.; Roopenian, D.C.; Ksander, B.R. An immunodominant minor histocompatibility alloantigen that initiates corneal allograft rejection1. Transplantation 2003, 75, 1368–1374. [Google Scholar] [CrossRef] [PubMed]
  44. Mettenleiter, T.C. Budding events in herpesvirus morphogenesis. Virus Res. 2004, 106, 167–180. [Google Scholar] [CrossRef]
  45. Akhtar, J.; Shukla, D. Viral entry mechanisms: Cellular and viral mediators of herpes simplex virus entry. FEBS J. 2009, 276, 7228–7236. [Google Scholar] [CrossRef] [PubMed]
  46. Nicola, A.V.; Hou, J.; Major, E.O.; Straus, S.E. Herpes Simplex Virus Type 1 Enters Human Epidermal Keratinocytes, but Not Neurons, via a pH-Dependent Endocytic Pathway. J. Virol. 2005, 79, 7609–7616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Turner, A.; Bruun, B.; Minson, T.; Browne, H. Browne Helena Glycoproteins gB, gD, and gHgL of Herpes Simplex Virus Type 1 Are Necessary and Sufficient To Mediate Membrane Fusion in a Cos Cell Transfection System. J. Virol. 1998, 72, 873–875. [Google Scholar] [CrossRef]
  48. Dai, X.; Zhang, X.; Ostrikov, K.; Abrahamyan, L. Host receptors: The key to establishing cells with broad viral tropism for vaccine production. Crit. Rev. Microbiol. 2020, 46, 147–168. [Google Scholar] [CrossRef] [Green Version]
  49. Greenan, E.; Gallagher, S.; Khalil, R.; Murphy, C.C.; Ní Gabhann-Dromgoole, J. Advancing Our Understanding of Corneal Herpes Simplex Virus-1 Immune Evasion Mechanisms and Future Therapeutics. Viruses 2021, 13, 1856. [Google Scholar] [CrossRef]
  50. Zheng, M.; Conrady, C.D.; Ward, J.M.; Bryant-Hudson, K.M.; Carr, D.J.J. Comparison of the host immune response to herpes simplex virus 1 (HSV-1) and HSV-2 at two different mucosal sites. J. Virol. 2012, 86, 7454–7458. [Google Scholar] [CrossRef] [Green Version]
  51. Conrady, C.D.; Zheng, M.; Fitzgerald, K.A.; Liu, C.; Carr, D.J.J. Resistance to HSV-1 Infection in the Epithelium Resides with the Novel Innate Sensor, IFI-16. Mucosal Immunol 2012, 5, 173–183. [Google Scholar] [CrossRef] [Green Version]
  52. Zhang, S.-Y.; Jouanguy, E.; Ugolini, S.; Smahi, A.; Elain, G.; Romero, P.; Segal, D.; Sancho-Shimizu, V.; Lorenzo, L.; Puel, A.; et al. TLR3 Deficiency in Patients with Herpes Simplex Encephalitis. Science 2007, 317, 1522–1527. [Google Scholar] [CrossRef] [PubMed]
  53. Conrady, C.D.; Zheng, M.; van Rooijen, N.; Drevets, D.A.; Royer, D.; Alleman, A.; Carr, D.J.J. Microglia and a functional type I IFN pathway are required to counter HSV-1-driven brain lateral ventricle enlargement and encephalitis. J. Immunol. 2013, 190, 2807–2817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Lafaille, F.G.; Pessach, I.M.; Zhang, S.-Y.; Ciancanelli, M.J.; Herman, M.; Abhyankar, A.; Ying, S.-W.; Keros, S.; Goldstein, P.A.; Mostoslavsky, G.; et al. Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 2012, 491, 769–773. [Google Scholar] [CrossRef] [Green Version]
  55. Casrouge, A.; Zhang, S.-Y.; Eidenschenk, C.; Jouanguy, E.; Puel, A.; Yang, K.; Alcais, A.; Picard, C.; Mahfoufi, N.; Nicolas, N.; et al. Herpes Simplex Virus Encephalitis in Human UNC-93B Deficiency. Science 2006, 314, 308–312. [Google Scholar] [CrossRef] [PubMed]
  56. Conrady, C.D.; Jones, H.; Zheng, M.; Carr, D.J.J. A Functional Type I Interferon Pathway Drives Resistance to Cornea Herpes Simplex Virus Type 1 Infection by Recruitment of Leukocytes. J. Biomed Res. 2011, 25, 111–119. [Google Scholar] [CrossRef]
  57. Al-khatib, K.; Williams, B.R.G.; Silverman, R.H.; Halford, W.; Carr, D.J.J. The murine double-stranded RNA-dependent protein kinase PKR and the murine 2′,5′-oligoadenylate synthetase-dependent RNase L are required for IFN-β-mediated resistance against herpes simplex virus type 1 in primary trigeminal ganglion culture. Virology 2003, 313, 126–135. [Google Scholar] [CrossRef] [Green Version]
  58. Al-khatib, K.; Williams, B.R.G.; Silverman, R.H.; Halford, W.; Carr, D.J.J. Distinctive Roles for 2′,5′-Oligoadenylate Synthetases and Double-Stranded RNA-Dependent Protein Kinase R in the In Vivo Antiviral Effect of an Adenoviral Vector Expressing Murine IFN-β1. J. Immunol. 2004, 172, 5638–5647. [Google Scholar] [CrossRef] [Green Version]
  59. Conrady, C.D.; Zheng, M.; Mandal, N.A.; van Rooijen, N.; Carr, D.J.J. IFN-α-driven CCL2 production recruits inflammatory monocytes to infection site in mice. Mucosal Immunol 2013, 6, 45–55. [Google Scholar] [CrossRef] [Green Version]
  60. Frank, G.M.; Buela, K.-A.G.; Maker, D.M.; Harvey, S.A.K.; Hendricks, R.L. Early Responding Dendritic Cells Direct the Local NK Response To Control Herpes Simplex Virus 1 Infection within the Cornea. J. Immunol. 2012, 188, 1350–1359. [Google Scholar] [CrossRef] [Green Version]
  61. Jamali, A.; Hu, K.; Sendra, V.G.; Blanco, T.; Lopez, M.J.; Ortiz, G.; Qazi, Y.; Zheng, L.; Turhan, A.; Harris, D.L.; et al. Characterization of Resident Corneal Plasmacytoid Dendritic Cells and Their Pivotal Role in Herpes Simplex Keratitis. Cell Rep. 2020, 32, 108099. [Google Scholar] [CrossRef] [PubMed]
  62. Conrady, C.D.; Thapa, M.; Wuest, T.; Carr, D.J.J. Loss of mandibular lymph node integrity is associated with an increase in sensitivity to HSV-1 infection in CD118-deficient mice. J. Immunol. 2009, 182, 3678–3687. [Google Scholar] [CrossRef] [Green Version]
  63. Conrady, C.D.; Zheng, M.; Stone, D.U.; Carr, D.J.J. CD8+ T cells suppress viral replication in the cornea but contribute to VEGF-C-induced lymphatic vessel genesis. J. Immunol. 2012, 189, 425–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bryant-Hudson, K.M.; Carr, D.J.J. PD-L1-expressing dendritic cells contribute to viral resistance during acute HSV-1 infection. Clin. Dev. Immunol. 2012, 2012, 924619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Royer, D.J.; Conrady, C.D.; Carr, D.J.J. Herpesvirus-Associated Lymphadenitis Distorts Fibroblastic Reticular Cell Microarchitecture and Attenuates CD8 T Cell Responses to Neurotropic Infection in Mice Lacking the STING-IFNα/β Defense Pathways. J. Immunol. 2016, 197, 2338–2352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lee, A.J.; Chen, B.; Chew, M.V.; Barra, N.G.; Shenouda, M.M.; Nham, T.; van Rooijen, N.; Jordana, M.; Mossman, K.L.; Schreiber, R.D.; et al. Inflammatory monocytes require type I interferon receptor signaling to activate NK cells via IL-18 during a mucosal viral infection. J. Exp. Med. 2017, 214, 1153–1167. [Google Scholar] [CrossRef] [Green Version]
  67. Madera, S.; Rapp, M.; Firth, M.A.; Beilke, J.N.; Lanier, L.L.; Sun, J.C. Type I IFN promotes NK cell expansion during viral infection by protecting NK cells against fratricide. J. Exp. Med. 2016, 213, 225–233. [Google Scholar] [CrossRef]
  68. Fitzpatrick, S.; Lausch, R.; Barrington, R.A. CCR6-Positive γδ T Cells Provide Protection Against Intracorneal HSV-1 Infection. Investig. Ophthalmol. Vis. Sci. 2019, 60, 3952–3962. [Google Scholar] [CrossRef] [Green Version]
  69. Ghiasi, H.; Cai, S.; Perng, G.-C.; Nesburn, A.B.; Wechsler, S.L. The role of natural killer cells in protection of mice against death and corneal scarring following ocular HSV-1 infection. Antivir. Res. 2000, 45, 33–45. [Google Scholar] [CrossRef]
  70. Orr, M.T.; Mathis, M.A.; Lagunoff, M.; Sacks, J.A.; Wilson, C.B. CD8 T Cell Control of HSV Reactivation from Latency Is Abrogated by Viral Inhibition of MHC Class I. Cell Host Microbe 2007, 2, 172–180. [Google Scholar] [CrossRef] [Green Version]
  71. Yu, W.; Geng, S.; Suo, Y.; Wei, X.; Cai, Q.; Wu, B.; Zhou, X.; Shi, Y.; Wang, B. Critical Role of Regulatory T Cells in the Latency and Stress-Induced Reactivation of HSV-1. Cell Rep. 2018, 25, 2379–2389.e3. [Google Scholar] [CrossRef] [Green Version]
  72. Knickelbein, J.E.; Khanna, K.M.; Yee, M.B.; Baty, C.J.; Kinchington, P.R.; Hendricks, R.L. Noncytotoxic Lytic Granule-Mediated CD8+ T Cell Inhibition of HSV-1 Reactivation from Neuronal Latency. Science 2008, 322, 268–271. [Google Scholar] [CrossRef] [Green Version]
  73. Prakash, S.; Roy, S.; Srivastava, R.; Coulon, P.-G.; Dhanushkodi, N.R.; Vahed, H.; Jankeel, A.; Geertsema, R.; Amezquita, C.; Nguyen, L.; et al. Unique molecular signatures of antiviral memory CD8+ T cells associated with asymptomatic recurrent ocular herpes. Sci. Rep. 2020, 10, 13843. [Google Scholar] [CrossRef] [PubMed]
  74. St. Leger, A.J.; Jeon, S.; Hendricks, R.L. Broadening the Repertoire of Functional Herpes Simplex Virus Type 1–Specific CD8+ T Cells Reduces Viral Reactivation from Latency in Sensory Ganglia. J. Immunol. 2013, 191, 2258–2265. [Google Scholar] [CrossRef] [Green Version]
  75. Croen, K.D. Evidence for antiviral effect of nitric oxide. Inhibition of herpes simplex virus type 1 replication. J. Clin. Investig. 1993, 91, 2446–2452. [Google Scholar] [CrossRef] [PubMed]
  76. Pertile, T.L.; Karaca, K.; Sharma, J.M.; Walser, M.M. An Antiviral Effect of Nitric Oxide: Inhibition of Reovirus Replication. Avian Dis. 1996, 40, 342–348. [Google Scholar] [CrossRef]
  77. Mehta, D.R.; Ashkar, A.A.; Mossman, K.L. The Nitric Oxide Pathway Provides Innate Antiviral Protection in Conjunction with the Type I Interferon Pathway in Fibroblasts. PLoS ONE 2012, 7, e31688. [Google Scholar] [CrossRef] [Green Version]
  78. Karupiah, G.; Xie, Q.; Buller, R.M.L.; Nathan, C.; Duarte, C.; MacMicking, J.D. Inhibition of Viral Replication by Interferon-γ-Induced Nitric Oxide Synthase. Science 1993, 261, 1445–1448. [Google Scholar] [CrossRef]
  79. Cheng, H.; Tumpey, T.M.; Staats, H.F.; van Rooijen, N.; Oakes, J.E.; Lausch, R.N. Role of Macrophages in Restricting Herpes Simplex Virus Type 1 Growth after Ocular Infection. Investig. Ophthalmol. Vis. Sci. 2000, 41, 1402–1409. [Google Scholar]
  80. Jaggi, U.; Matundan, H.H.; Yu, J.; Hirose, S.; Mueller, M.; Wormley, F.L., Jr.; Ghiasi, H. Essential role of M1 macrophages in blocking cytokine storm and pathology associated with murine HSV-1 infection. PLOS Pathog. 2021, 17, e1009999. [Google Scholar] [CrossRef] [PubMed]
  81. Wuest, T.R.; Carr, D.J.J. VEGF-A expression by HSV-1–infected cells drives corneal lymphangiogenesis. J. Exp. Med. 2009, 207, 101–115. [Google Scholar] [CrossRef] [PubMed]
  82. Wuest, T.; Zheng, M.; Efstathiou, S.; Halford, W.P.; Carr, D.J.J. The Herpes Simplex Virus-1 Transactivator Infected Cell Protein-4 Drives VEGF-A Dependent Neovascularization. PLoS Pathog. 2011, 7, e1002278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Suryawanshi, A.; Mulik, S.; Sharma, S.; Reddy, P.B.J.; Sehrawat, S.; Rouse, B.T. Ocular Neovascularization Caused by Herpes Simplex Virus Type 1 Infection Results from Breakdown of Binding between Vascular Endothelial Growth Factor A and Its Soluble Receptor. J. Immunol. 2011, 186, 3653–3665. [Google Scholar] [CrossRef] [Green Version]
  84. Yun, H.; Yee, M.B.; Lathrop, K.L.; Kinchington, P.R.; Hendricks, R.L.; St. Leger, A.J. Production of the Cytokine VEGF-A by CD4+ T and Myeloid Cells Disrupts the Corneal Nerve Landscape and Promotes Herpes Stromal Keratitis. Immunity 2020, 53, 1050–1062.e5. [Google Scholar] [CrossRef]
  85. Kuffova, L.; Knickelbein, J.E.; Yu, T.; Medina, C.; Amescua, G.; Rowe, A.M.; Hendricks, R.L.; Forrester, J.V. High-Risk Corneal Graft Rejection in the Setting of Previous Corneal Herpes Simplex Virus (HSV)-1 Infection. Investig. Ophthalmol. Vis. Sci. 2016, 57, 1578–1587. [Google Scholar] [CrossRef] [Green Version]
  86. Biswas, P.S.; Rouse, B.T. Early events in HSV keratitis—Setting the stage for a blinding disease. Microbes Infect. 2005, 7, 799–810. [Google Scholar] [CrossRef] [PubMed]
  87. Giménez, F.; Suryawanshi, A.; Rouse, B.T. Pathogenesis of herpes stromal keratitis—A focus on corneal neovascularization. Prog. Retin. Eye Res. 2013, 33, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Mercadal, C.M.; Bouley, D.M.; DeStephano, D.; Rouse, B.T. Herpetic stromal keratitis in the reconstituted scid mouse model. J. Virol. 1993, 67, 3404–3408. [Google Scholar] [CrossRef]
  89. Russell, R.G.; Nasisse, M.P.; Larsen, H.S.; Rouse, B.T. Role of T-lymphocytes in the pathogenesis of herpetic stromal keratitis. Investig. Ophthalmol. Vis. Sci. 1984, 25, 938–944. [Google Scholar]
  90. Wilhelmus, K.R.; Gee, L.; Hauck, W.W.; Kurinij, N.; Dawson, C.R.; Jones, D.B.; Barron, B.A.; Kaufman, H.E.; Sugar, J.; Hyndiuk, R.A.; et al. Herpetic Eye Disease Study: A Controlled Trial of Topical Corticosteroids for Herpes Simplex Stromal Keratitis. Ophthalmology 1994, 101, 1883–1896. [Google Scholar] [CrossRef]
  91. Royer, D.J.; Zheng, M.; Conrady, C.D.; Carr, D.J.J. Granulocytes in Ocular HSV-1 Infection: Opposing Roles of Mast Cells and Neutrophils. Investig. Ophthalmol. Vis. Sci. 2015, 56, 3763–3775. [Google Scholar] [CrossRef] [Green Version]
  92. Zheng, M.; Deshpande, S.; Lee, S.; Ferrara, N.; Rouse, B.T. Contribution of Vascular Endothelial Growth Factor in the Neovascularization Process during the Pathogenesis of Herpetic Stromal Keratitis. J. Virol. 2001, 75, 9828–9835. [Google Scholar] [CrossRef] [Green Version]
  93. Sohn, J.-H.; Bora, P.S.; Suk, H.-J.; Molina, H.; Kaplan, H.J.; Bora, N.S. Tolerance is dependent on complement C3 fragment iC3b binding to antigen-presenting cells. Nat. Med. 2003, 9, 206–212. [Google Scholar] [CrossRef] [PubMed]
  94. Filiberti, A.; Gmyrek, G.B.; Berube, A.N.; Royer, D.J.; Carr, D.J.J. An intact complement system dampens cornea inflammation during acute primary HSV-1 infection. Sci. Rep. 2021, 11, 10247. [Google Scholar] [CrossRef] [PubMed]
  95. Henry, C.R.; Flynn, H.W.J.; Miller, D.; Forster, R.K.; Alfonso, E.C. Infectious keratitis progressing to endophthalmitis: A 15-year study of microbiology, associated factors, and clinical outcomes. Ophthalmology 2012, 119, 2443–2449. [Google Scholar] [CrossRef] [Green Version]
  96. Durand, M.L.; Barshak, M.B.; Chodosh, J. Infectious Keratitis in 2021. JAMA 2021, 326, 1319–1320. [Google Scholar] [CrossRef]
  97. Khoo, P.; Cabrera-Aguas, M.P.; Nguyen, V.; Lahra, M.M.; Watson, S.L. Microbial keratitis in Sydney, Australia: Risk factors, patient outcomes, and seasonal variation. Graefes Arch. Clin. Exp. Ophthalmol. 2020, 258, 1745–1755. [Google Scholar] [CrossRef]
  98. Das, S.; Jhanji, V. Infections of the Cornea and Conjunctiva; Springer: Berlin/Heidelberg, Germany, 2021; ISBN 9811588112. [Google Scholar]
  99. Lin, A.; Rhee, M.K.; Akpek, E.K.; Amescua, G.; Farid, M.; Garcia-Ferrer, F.J.; Varu, D.M.; Musch, D.C.; Dunn, S.P.; Mah, F.S. Bacterial Keratitis Preferred Practice Pattern®. Ophthalmology 2019, 126, P1–P55. [Google Scholar] [CrossRef] [Green Version]
  100. Ting, D.S.J.; Cairns, J.; Gopal, B.P.; Ho, C.S.; Krstic, L.; Elsahn, A.; Lister, M.; Said, D.G.; Dua, H.S. Risk Factors, Clinical Outcomes, and Prognostic Factors of Bacterial Keratitis: The Nottingham Infectious Keratitis Study. Front. Med. 2021, 8, 715118. [Google Scholar] [CrossRef]
  101. Eby, A.; Hazlett, L. Pseudomonas Keratitis, a Review of Where We’ve Been and What Lies Ahead. J. Microb. Biochem. Technol. 2015, 8, 9–13. [Google Scholar]
  102. Esen, M.; Grassmé, H.; Riethmüller, J.; Riehle, A.; Fassbender, K.; Gulbins, E. Gulbins Erich Invasion of Human Epithelial Cells by Pseudomonas aeruginosa Involves Src-Like Tyrosine Kinases p60Src and p59Fyn. Infect. Immun. 2001, 69, 281–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hilliam, Y.; Kaye, S.; Winstanley, C. Pseudomonas aeruginosa and microbial keratitis. J. Med. Microbiol. 2020, 69, 3–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Finck-Barbançon, V.; Goranson, J.; Zhu, L.; Sawa, T.; Wiener-Kronish, J.P.; Fleiszig, S.M.J.; Wu, C.; Mende-Mueller, L.; Frank, D.W. ExoU expression by Pseudomonas aeruginosa correlates with acute cytotoxicity and epithelial injury. Mol. Microbiol. 1997, 25, 547–557. [Google Scholar] [CrossRef]
  105. Murugan, N.; Malathi, J.; Umashankar, V.; Madhavan, H.N. Resistome and pathogenomics of multidrug resistant (MDR) Pseudomonas aeruginosa VRFPA03, VRFPA05 recovered from alkaline chemical keratitis and post-operative endophthalmitis patient. Gene 2016, 578, 105–111. [Google Scholar] [CrossRef] [PubMed]
  106. Borkar, D.S.; Acharya, N.R.; Leong, C.; Lalitha, P.; Srinivasan, M.; Oldenburg, C.E.; Cevallos, V.; Lietman, T.M.; Evans, D.J.; Fleiszig, S.M.J. Cytotoxic clinical isolates of Pseudomonas aeruginosaidentified during the Steroids for Corneal Ulcers Trial show elevated resistance to fluoroquinolones. BMC Ophthalmol. 2014, 14, 54. [Google Scholar] [CrossRef] [Green Version]
  107. Bardoel, B.W.; van der Ent, S.; Pel, M.J.C.; Tommassen, J.; Pieterse, C.M.J.; van Kessel, K.P.M.; van Strijp, J.A.G. Pseudomonas Evades Immune Recognition of Flagellin in Both Mammals and Plants. PLoS Pathog. 2011, 7, e1002206. [Google Scholar] [CrossRef] [Green Version]
  108. Zaidi, T.S.; Zaidi, T.; Pier, G.B. Role of Neutrophils, MyD88-Mediated Neutrophil Recruitment, and Complement in Antibody-Mediated Defense against Pseudomonas aeruginosa Keratitis. Investig. Ophthalmol. Vis. Sci. 2010, 51, 2085–2093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Huang, X.; Barrett, R.P.; McClellan, S.A.; Hazlett, L.D. Silencing Toll-like Receptor-9 in Pseudomonas aeruginosa Keratitis. Investig. Ophthalmol. Vis. Sci. 2005, 46, 4209–4216. [Google Scholar] [CrossRef] [Green Version]
  110. Huang, X.; Du, W.; McClellan, S.A.; Barrett, R.P.; Hazlett, L.D. TLR4 Is Required for Host Resistance in Pseudomonas aeruginosa Keratitis. Investig. Ophthalmol. Vis. Sci. 2006, 47, 4910–4916. [Google Scholar] [CrossRef] [Green Version]
  111. Yoon, G.S.; Dong, C.; Gao, N.; Kumar, A.; Standiford, T.J.; Yu, F.-S.X. Interferon Regulatory Factor-1 in Flagellin-Induced Reprogramming: Potential Protective Role of CXCL10 in Cornea Innate Defense Against Pseudomonas aeruginosa Infection. Investig. Ophthalmol. Vis. Sci. 2013, 54, 7510–7521. [Google Scholar] [CrossRef] [Green Version]
  112. Zhang, J.; Xu, K.; Ambati, B.; Yu, F.-S.X. Toll-like Receptor 5-Mediated Corneal Epithelial Inflammatory Responses to Pseudomonas aeruginosa Flagellin. Investig. Ophthalmol. Vis. Sci. 2003, 44, 4247–4254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Hazlett, L.D. Corneal response to Pseudomonas aeruginosa infection. Prog. Retin. Eye Res. 2004, 23, 1–30. [Google Scholar] [CrossRef] [PubMed]
  114. Thanabalasuriar, A.; Scott, B.N.V.; Peiseler, M.; Willson, M.E.; Zeng, Z.; Warrener, P.; Keller, A.E.; Surewaard, B.G.J.; Dozier, E.A.; Korhonen, J.T.; et al. Neutrophil Extracellular Traps Confine Pseudomonas aeruginosa Ocular Biofilms and Restrict Brain Invasion. Cell Host Microbe 2019, 25, 526–536.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Vareechon, C.; Zmina, S.E.; Karmakar, M.; Pearlman, E.; Rietsch, A. Pseudomonas aeruginosa Effector ExoS Inhibits ROS Production in Human Neutrophils. Cell Host Microbe 2017, 21, 611–618.e5. [Google Scholar] [CrossRef] [Green Version]
  116. Berger, E.A.; McClellan, S.A.; Vistisen, K.S.; Hazlett, L.D. HIF-1α Is Essential for Effective PMN Bacterial Killing, Antimicrobial Peptide Production and Apoptosis in Pseudomonas aeruginosa Keratitis. PLoS Pathog. 2013, 9, e1003457. [Google Scholar] [CrossRef]
  117. Kernacki, K.A.; Barrett, R.P.; Hobden, J.A.; Hazlett, L.D. Macrophage Inflammatory Protein-2 Is a Mediator of Polymorphonuclear Neutrophil Influx in Ocular Bacterial Infection1. J. Immunol. 2000, 164, 1037–1045. [Google Scholar] [CrossRef] [Green Version]
  118. McClellan, S.; Jiang, X.; Barrett, R.; Hazlett, L.D. High-Mobility Group Box 1: A Novel Target for Treatment of Pseudomonas aeruginosa Keratitis. J. Immunol. 2015, 194, 1776–1787. [Google Scholar] [CrossRef] [Green Version]
  119. Szliter, E.A.; Lighvani, S.; Barrett, R.P.; Hazlett, L.D. Vasoactive Intestinal Peptide Balances Pro- and Anti-Inflammatory Cytokines in the Pseudomonas aeruginosa-Infected Cornea and Protects against Corneal Perforation1. J. Immunol. 2007, 178, 1105–1114. [Google Scholar] [CrossRef] [Green Version]
  120. Rudner, X.L.; Kernacki, K.A.; Barrett, R.P.; Hazlett, L.D. Prolonged Elevation of IL-1 in Pseudomonas aeruginosa Ocular Infection Regulates Macrophage-Inflammatory Protein-2 Production, Polymorphonuclear Neutrophil Persistence, and Corneal Perforation1. J. Immunol. 2000, 164, 6576–6582. [Google Scholar] [CrossRef]
  121. Hazlett, L.D.; McClellan, S.; Kwon, B.; Barrett, R. Increased Severity of Pseudomonas aeruginosa Corneal Infection in Strains of Mice Designated as Th1 versus Th2 Responsive. Investig. Ophthalmol. Vis. Sci. 2000, 41, 805–810. [Google Scholar]
  122. Gao, J.-L.; Wynn, T.A.; Chang, Y.; Lee, E.J.; Broxmeyer, H.E.; Cooper, S.; Tiffany, H.L.; Westphal, H.; Kwon-Chung, J.; Murphy, P.M. Impaired Host Defense, Hematopoiesis, Granulomatous Inflammation and Type 1–Type 2 Cytokine Balance in Mice Lacking CC Chemokine Receptor 1. J. Exp. Med. 1997, 185, 1959–1968. [Google Scholar] [CrossRef] [PubMed]
  123. Me, R.; Gao, N.; Zhang, Y.; Lee, P.S.Y.; Wang, J.; Liu, T.; Standiford, T.J.; Mi, Q.-S.; Yu, F.-S.X. IL-36α Enhances Host Defense against Pseudomonas aeruginosa Keratitis in C57BL/6 Mouse Corneas. J. Immunol. 2021, 207, 2868–2877. [Google Scholar] [CrossRef] [PubMed]
  124. Me, R.; Gao, N.; Dai, C.; Yu, F.X. IL-17 Promotes Pseudomonas aeruginosa Keratitis in C57BL/6 Mouse Corneas. J. Immunol. 2020, 204, 169–179. [Google Scholar] [CrossRef]
  125. Kopplin, L.J.; Thomas, A.S.; Cramer, S.; Kim, Y.H.; Yeh, S.; Lauer, A.K.; Flaxel, C.J. Long-Term Surgical Outcomes of Retinal Detachment Associated With Acute Retinal Necrosis. Ophthalmic Surg. Lasers Imaging Retin. 2016, 47, 660–664. [Google Scholar] [CrossRef]
  126. Silva, R.A.; Berrocal, A.M.; Moshfeghi, D.M.; Blumenkranz, M.S.; Sanislo, S.; Davis, J.L. Herpes simplex virus type 2 mediated acute retinal necrosis in a pediatric population: Case series and review. Graefe’s Arch. Clin. Exp. Ophthalmol. 2013, 251, 559–566. [Google Scholar] [CrossRef] [PubMed]
  127. Schoenberger, S.D.; Kim, S.J.; Thorne, J.E.; Mruthyunjaya, P.; Yeh, S.; Bakri, S.J.; Ehlers, J.P. Diagnosis and Treatment of Acute Retinal Necrosis: A Report by the American Academy of Ophthalmology. Ophthalmology 2017, 124, 382–392. [Google Scholar] [CrossRef] [Green Version]
  128. Fukunaga, H.; Kaburaki, T.; Shirahama, S.; Tanaka, R.; Murata, H.; Sato, T.; Takeuchi, M.; Tozawa, H.; Urade, Y.; Katsura, M.; et al. Analysis of inflammatory mediators in the vitreous humor of eyes with pan-uveitis according to aetiological classification. Sci. Rep. 2020, 10, 2783. [Google Scholar] [CrossRef] [Green Version]
  129. de Visser, L.; de Boer, J.H.; Rijkers, G.T.; Wiertz, K.; van den Ham, H.J.; de Boer, R.; van Loon, A.M.; Rothova, A.; de Groot-Mijnes, D.F. Cytokines and Chemokines Involved in Acute Retinal Necrosis. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2139–2151. [Google Scholar] [CrossRef] [Green Version]
  130. Von Szily, A. Experimental endogenous transmission of infection from bulbus to bulbus. Klin Monatsbl Augenheilkd 1924, 75, 593–602. [Google Scholar]
  131. Atherton, S.S.; Streilein, J.W. Two waves of virus following anterior chamber inoculation of HSV-1. Investig. Ophthalmol. Vis. Sci. 1987, 28, 571–579. [Google Scholar] [CrossRef]
  132. Kezuka, T.; Sakai, J.; Usui, N.; Streilein, J.W.; Usui, M. Evidence for Antigen-Specific Immune Deviation in Patients With Acute Retinal Necrosis. Arch. Ophthalmol. 2001, 119, 1044–1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zheng, M.; Atherton, S.S. Cytokine Profiles and Inflammatory Cells during HSV-1–Induced Acute Retinal Necrosis. Investig. Ophthalmol. Vis. Sci. 2005, 46, 1356–1363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zheng, M.; Qian, H.; Joshi, R.M.; Nechtman, J.; Atherton, S.S. DNA microarray analysis of the uninoculated eye following anterior chamber inoculation of HSV-1. Ocul. Immunol. Inflamm. 2003, 11, 187–195. [Google Scholar] [CrossRef]
  135. Zheng, M.; Fields, M.A.; Liu, Y.; Cathcart, H.; Richter, E.; Atherton, S.S. Neutrophils Protect the Retina of the Injected Eye from Infection after Anterior Chamber Inoculation of HSV-1 in BALB/c Mice. Investig. Ophthalmol. Vis. Sci. 2008, 49, 4018–4025. [Google Scholar] [CrossRef] [PubMed]
  136. Grajewski, R.S.; Li, J.; Wasmuth, S.; Hennig, M.; Bauer, D.; Heiligenhaus, A. Intravitreal treatment with antisense oligonucleotides targeting tumor necrosis factor-α in murine herpes simplex virus type 1 retinitis. Graefe’s Arch. Clin. Exp. Ophthalmol. 2012, 250, 231–238. [Google Scholar] [CrossRef]
  137. Azumi, A.; Atherton, S.S. T cells in the uninjected eye after anterior chamber inoculation of herpes simplex virus type 1. Investig. Ophthalmol. Vis. Sci. 1998, 39, 78–83. [Google Scholar]
  138. Ng, C.C.; Chen, J.J.; Agarwal, A.; Cunningham, E.T. Clinical course of von Szily reaction: Case report and comprehensive review of the literature. Am. J. Ophthalmol. Case Rep. 2020, 20, 100927. [Google Scholar] [CrossRef]
  139. Ross, B.X.; Jia, L.; Kong, D.; Wang, T.; Hager, H.M.; Abcouwer, S.F.; Zacks, D.N. Conditional Knock out of High-Mobility Group Box 1 (HMGB1) in Rods Reduces Autophagy Activation after Retinal Detachment. Cells 2021, 10, 2010. [Google Scholar] [CrossRef]
  140. Freeman, W.; Schneiderman, T.; Wiley, C.; Listhaus, A.; Svendsen, P.; Munguia, D.; Bergeron-Lynn, G. An animal model of focal, subacute, viral retinitis. Retina 1993, 13, 214–221. [Google Scholar] [CrossRef]
  141. Fan, S.; Yoo, J.H.; Park, G.; Yeh, S.; Conrady, C.D. Type I Interferon Signaling Is Critical During the Innate Immune Response to HSV-1 Retinal Infection. Investig. Ophthalmol. Vis. Sci. 2022, 63, 28. [Google Scholar] [CrossRef]
  142. Conrady, C.D.; Halford, W.P.; Carr, D.J.J. Loss of the type I interferon pathway increases vulnerability of mice to genital herpes simplex virus 2 infection. J. Virol. 2011, 85, 1625–1633. [Google Scholar] [CrossRef] [Green Version]
  143. Chien, H.; Dix, R.D. Evidence For Multiple Cell Death Pathways during Development of Experimental Cytomegalovirus Retinitis in Mice with Retrovirus-Induced Immunosuppression: Apoptosis, Necroptosis, and Pyroptosis. J. Virol. 2012, 86, 10961–10978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Carter, J.J.; Nemeno, J.G.E.; Oh, J.J.; Houghton, J.E.; Dix, R.D. Atypical cytomegalovirus retinal disease in pyroptosis-deficient mice with murine acquired immunodeficiency syndrome. Exp. Eye Res. 2021, 209, 108651. [Google Scholar] [CrossRef] [PubMed]
  145. Guo, H.; Koehler, H.S.; Mocarski, E.S.; Dix, R.D. RIPK3 and caspase 8 collaborate to limit herpes simplex encephalitis. PLoS Pathog. 2022, 18, e1010857. [Google Scholar] [CrossRef]
  146. Brundrett, A.; Conrady, C.D.; Shakoor, A.; Lin, A. Current Strategies for Prevention and Treatment of Postoperative Endophthalmitis. Curr. Ophthalmol. Rep. 2018, 6, 105–114. [Google Scholar] [CrossRef]
  147. Conrady, C.D.; Feistmann, J.A.; Roller, A.B.; Boldt, H.C.; Shakoor, A. Hemorrhagic vasculitis and retinopathy heralding as an early sign of bacterial endophthalmitis after intravitreal injection. Retin. Cases Brief Rep. 2017, 13, 329–332. [Google Scholar] [CrossRef]
  148. Conrady, C.D.; Feist, R.M., Jr.; Vitale, A.T.; Shakoor, A. Long-term visual outcomes of endophthalmitis and the role of systemic steroids in addition to intravitreal dexamethasone. BMC Ophthalmol. 2020, 20, 181. [Google Scholar] [CrossRef] [PubMed]
  149. Ho, I.-V.; Fernandez-Sanz, G.; Levasseur, S.; Ting, E.; Liew, G.; Playfair, J.; Downie, J.; Gorbatov, M.; Hunyor, A.P.; Chang, A.A. Early Pars Plana Vitrectomy for Treatment of Acute Infective Endophthalmitis. Asia-Pac. J. Ophthalmol. 2019, 8, 3–7. [Google Scholar]
  150. Combey de Lambert, A.; Campolmi, N.; Cornut, P.-L.; Aptel, F.; Creuzot-Garcher, C.; Chiquet, C. French Institutional Endophthalmitis Study Group Baseline factors predictive of visual prognosis in acute postoperative bacterial endophthalmitis in patients undergoing cataract surgery. JAMA Ophthalmol. 2013, 131, 1159–1166. [Google Scholar] [CrossRef] [Green Version]
  151. Lin, J.; Huang, S.; Liu, M.; Lin, L.; Gu, J.; Duan, F. Endophthalmitis Caused by Pseudomonas aeruginosa: Clinical Characteristics, Outcomes, and Antibiotics Sensitivities. J. Ophthalmol. 2022, 2022, 1265556. [Google Scholar] [CrossRef]
  152. Staropoli, P.C.; Flynn, H.W., Jr.; Miller, D.; Persad, P.J.; Vanner, E.A. Endophthalmitis Caused by Streptococcus:Clinical Outcomes and Antimicrobial Susceptibilities 2014–2019. Ophthalmic Surg. Lasers Imaging Retin. 2021, 52, 182–189. [Google Scholar] [CrossRef] [PubMed]
  153. Endophthalmitis Vitrectomy Study Group. A randomized trial of immediate vitrectomy and of intravenous antibiotics for the treatment of postoperative bacterial endophthalmitis. Arch. Ophthalmol. 1995, 113, 1479–1496. [Google Scholar] [CrossRef]
  154. Kumar, A.; Kumar, A. Role of Staphylococcus aureus Virulence Factors in Inducing Inflammation and Vascular Permeability in a Mouse Model of Bacterial Endophthalmitis. PLoS ONE 2015, 10, e0128423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Das, S.; Singh, S.; Kumar, A. Bacterial Burden Declines But Neutrophil Infiltration and Ocular Tissue Damage Persist in Experimental Staphylococcus epidermidis Endophthalmitis. Front. Cell. Infect. Microbiol. 2021, 11, 780648. [Google Scholar] [CrossRef] [PubMed]
  156. Soon, M.Y.; Allen, P.J.; Dawkins, R.C.H. Cytokine Expression in Staphylococcal and Streptococcal Endophthalmitis. Biomed. Hub 2022, 7, 88–98. [Google Scholar] [CrossRef]
  157. Coburn, P.S.; Parrott, A.C.; Miller, F.C.; LaGrow, A.L.; Mursalin, M.H.; Callegan, M.C. The Role of C-X-C Chemokines in Staphylococcus aureus Endophthalmitis. Investig. Ophthalmol. Vis. Sci. 2023, 64, 10. [Google Scholar] [CrossRef]
  158. Tsioti, I.; Steiner, B.L.; Escher, P.; Zinkernagel, M.S.; Benz, P.M.; Kokona, D. Endothelial Toll-like receptor 4 is required for microglia activation in the murine retina after systemic lipopolysaccharide exposure. J. Neuroinflammation 2023, 20, 25. [Google Scholar] [CrossRef]
  159. 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, 2076–2088. [Google Scholar] [CrossRef] [Green Version]
  160. Kumar, A.; Singh, P.K.; Ahmed, Z.; Singh, S.; Kumar, A. Kumar Ashok Essential Role of NLRP3 Inflammasome in Mediating IL-1β Production and the Pathobiology of Staphylococcus aureus Endophthalmitis. Infect. Immun. 2022, 90, e00103–e00122. [Google Scholar] [CrossRef]
  161. Grumann, D.; Nübel, U.; Bröker, B.M. Staphylococcus aureus toxins—Their functions and genetics. Infect. Genet. Evol. 2014, 21, 583–592. [Google Scholar] [CrossRef] [Green Version]
  162. Callegan, M.C.; Engelbert, M.; Parke, D.W.; Jett, B.D.; Gilmore, M.S. Bacterial Endophthalmitis: Epidemiology, Therapeutics, and Bacterium-Host Interactions. Clin. Microbiol. Rev. 2002, 15, 111–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Coburn, P.S.; Miller, F.C.; LaGrow, A.L.; Land, C.; Mursalin, H.; Livingston, E.; Amayem, O.; Chen, Y.; Gao, W.; Zhang, L.; et al. Disarming Pore-Forming Toxins with Biomimetic Nanosponges in Intraocular Infections. mSphere 2019, 4, e00262-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Baharivand, N.; Mahdavifard, A.; Fouladi, R.F. Intravitreal clindamycin plus dexamethasone versus classic oral therapy in toxoplasmic retinochoroiditis: A prospective randomized clinical trial. Int. Ophthalmol. 2013, 33, 39–46. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Innate immune response to HSV-1 infection of the cornea. M, macrophages.
Figure 1. Innate immune response to HSV-1 infection of the cornea. M, macrophages.
Microorganisms 11 02074 g001
Figure 2. Innate immune response to bacterial infection of the cornea. P, neutrophils.
Figure 2. Innate immune response to bacterial infection of the cornea. P, neutrophils.
Microorganisms 11 02074 g002
Table 1. Ocular infections and their common causative bacteria.
Table 1. Ocular infections and their common causative bacteria.
Sites of Ocular Infections and Associated Bacterial Microbes
Site of Infection Associated Bacteria
Ocular SurfaceKeratitisGram-PositiveStaphylococcus spp., Enterococcus faecalis, Corynebacterium spp., and Bacillus spp.
Gram-NegativePseudomonas aeruginosa, Escherichia coli, K. pneumoniae, Acinetobacter, Serratia marcescens, Serratia liquefaciens, Aeromonas spp., Fusobacterium spp., Enterobacter spp., Proteus mirabilis, P Pasteurella multocida, Morexella catarrhalis, and Haemophilus influenzae
ConjunctivitisGram-PositiveStaphylococcus spp., Streptoccus spp., E. faecalis, Corynebacterium spp., and Bacillus spp.
Gram-NegativeNeisseria gonorrhoeae, Chlamydia trachomatis, Escherichia coli, P. aeruginosa, Enterobacter spp. (i.e., E. cloacae and E. aerogenes), Citrobacter koseri, Proteus spp., Moraxella spp., Morexella catarrhalis, and Haemophilus influenzae
Episcleritis/ScleritisGram-PositiveStaphylococcus spp., Streptococcus spp., Nocardia
Gram-NegativePseudomonas aeruginosa, Klebsiella spp., and Mycobacterium tuberculosis
IntraocularUveitisGram-Positive* Nocardia brasiliensis
Gram-NegativeMycobaterium tuberculosis, * Rickettsia spp., * Francisella tularensis, * Bartonella henselae, * Yersinia enterocolitica, * Pasteurella multocida, and * Chlamydia trachomatis
EndophthalmitisGram-PositiveStaphylococcus spp., Streptoccus spp., Corynebacterium spp., Bacillus spp., and * Clostridium spp.
Gram-NegativePseudomonas aeruginosa, Escherichia coli, Propiolactone spp., Serratia spp., Klebsiella pneumoniae, Enterobacter spp., Acinetobacter spp., Morexella catarrhailis, and Haemophilus spp.
Ocular AdnexaDacryocystisGram-PositiveStaphylococcus spp., Streptococcus spp., and Corynebacterium spp.
Gram-NegativePseudomonas spp., Enterobacter spp., Klebsella. pneumoniae, Haemophilus influenzae, Escherichia coli, * Acinetobacter iwoifi, * Haemophilus parainfluenzae, and * Haemophilus aegypticus
BlepharitisGram-PositiveStaphylococcus aureus, Staphylococcus spp., Enterococcus faecalis, and Corynebacterium spp.
Gram-NegativeHaemophilus influenzae
Rarely Observed *.
Table 2. Ocular infections and their common causative viruses, fungi, arthropods, and protozoan species.
Table 2. Ocular infections and their common causative viruses, fungi, arthropods, and protozoan species.
Sites of Ocular Infections and Associated Microbes
Site of InfectionInfection CategoryAssociated Pathogens
Ocular SurfaceKeratitisVirusesHSV, VSV, and adenovirus
FungiCandida spp., and Aspergillus spp.
ProtozoaAcanthamoeba spp.
ConjunctivitisVirusesHSV, and adenoviruses
Episcleritis/ScleritisVirusesVZV
IntraocularAnterior UveitisVirusesHSV, VZV, and CMV
Posterior UveitisVirusesHSV, VZV, and CMV
EndophthalmitisProtozoaToxoplasma gondii
Viruses
FungiCandida spp., Aspergillus spp., and Fusarium spp.
Ocular AdnexaBlepharitisMitesDemodex folliculorum
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dempsey, M.P.; Conrady, C.D. The Host–Pathogen Interplay: A Tale of Two Stories within the Cornea and Posterior Segment. Microorganisms 2023, 11, 2074. https://doi.org/10.3390/microorganisms11082074

AMA Style

Dempsey MP, Conrady CD. The Host–Pathogen Interplay: A Tale of Two Stories within the Cornea and Posterior Segment. Microorganisms. 2023; 11(8):2074. https://doi.org/10.3390/microorganisms11082074

Chicago/Turabian Style

Dempsey, Michael P., and Christopher D. Conrady. 2023. "The Host–Pathogen Interplay: A Tale of Two Stories within the Cornea and Posterior Segment" Microorganisms 11, no. 8: 2074. https://doi.org/10.3390/microorganisms11082074

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop