Skip to main content

REVIEW article

Front. Immunol., 19 March 2019
Sec. Viral Immunology
This article is part of the Research Topic Immunity and Immunopathogenesis to Herpesviruses View all 10 articles

The Role of T Cells in Herpes Stromal Keratitis

\r\nNaveen K. RajasagiNaveen K. RajasagiBarry T. Rouse*Barry T. Rouse*
  • Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, United States

The blinding inflammatory lesion stromal keratitis (SK), which occurs in some patients in response to ocular herpes simplex virus (HSV) infection, represents mainly an immune cell mediated inflammatory response to the virus infection. The principal orchestrators of the immunopathological lesions are T cells although additional events participate that include the extent of recruitment of non-lymphoid cells, the extent of neoangiogenesis, and the extent of damage to nerve function. This review focuses on evidence that the balance of the functional subsets of T cells has a major impact on lesion severity and duration. Accordingly, if proinflammatory Th1 and Th17 CD4 T cells, and perhaps in some cases CD8 T cells, predominate lesions occur earlier and are more severe. Lesions are diminished when cells with regulatory function predominate. Moreover, when regulatory cells acquire the property to produce Amphiregulin this may facilitate lesion resolution. An objective to controlling lesions is to learn how to manipulate the balance of T cells to favor the representation and function of regulatory T cells and their products over proinflammatory cells. In this review we emphasize how exploiting the differential metabolic requirements of immune cells could be a valuable approach to control SK.

Background

Herpes simplex virus (HSV) type 1 is a major human pathogen worldwide. It is estimated that around 67% of people worldwide (under age 50) are infected with HSV-1 (1). HSV-1 establishes a lifelong, latent infection for which no effective vaccine is currently available (2). Primary infection with HSV-1 is usually mild or subclinical and most individuals remain asymptomatic (3). However, HSV-1 infection can cause several complications in humans. Among these, corneal infection can lead to blinding immunopathological lesions in the eye referred to as herpes stromal keratitis (SK) (4, 5). Epidemiology studies outside of the United States have estimated incidence rates of HSV eye disease range from ~4 to 13 new cases per 100,000 per year. A previous study from Rochester, Minnesota, estimated an incidence of 8.4 new cases per 100,000 and 20.7 total episodes per 100,000 people per year. Extrapolating these data to the US population census in 2000, the study predicted an estimated incidence of ~24,000 new cases and 58,000 total episodes per year (6). Moreover, a study published in 2014, estimated an incidence of 6.8 new cases/100,000 in Northern California (7). Thus, herpes keratitis represents a clinically relevant syndrome and the SK form is a frequent cause of vision damage.

Primary ocular infection most likely occurs by the direct infection of the eye with HSV-1. Upon infection, the virus replicates in the corneal epithelial cells and can causes epithelial lesions. These primary lesions can last up to 2 weeks and usually resolve with minimal damage and the virus is efficiently cleared by the immune system (8). However, one of the consequences of HSV ocular infection is the establishment of latency in the trigeminal ganglia (TG) (9). Some of the HSV virions can enter the sensory nerve endings which innervate the infected cells and traffic via retrograde transport mechanisms to the sensory ganglia where the virus can persist in a latent stage (10). Sometimes the latent virus reactivates by disturbances caused by environmental or physiological stress and the reactivated HSV replicates in the TG. The virus can then travel by anterograde axonal transport to the peripheral tissues and cause recurrent lesions either in the corneal or orofacial tissues often resulting in clinical consequences (11). In humans, recurrent virus infections of the cornea are usually confined to the epithelial layer, but in some individuals such frequent recurrent infections could affect the deeper corneal stroma leading to an immunopathological disease referred to as herpes stromal keratitis (SK). This chronic inflammatory response in the corneal stroma is mediated by both innate and adaptive immune cells in response to virus infection and can lead to progressive corneal scarring and vision loss. The local corneal epithelial lesions and virus infections are usually treated using antivirals such as acyclovir, but SK lesions are often treated with a combination of an antiviral and a corticosteroid (12).

Most of our current understanding of the pathogenesis of SK in humans comes from studies done animal models (5, 13). HSV-1 corneal infection in mice is the most widely used animal model to study SK as it offers several advantages and the inflammatory lesions in the corneal stroma mimic SK lesions observed in humans (14). However, one limitation of the mice model is that it is mainly a primary infection model, but not a reactivation model of disease as mostly occurs in humans. The immune response to HSV-1 ocular infection occurs in a bi-phasic manner and involves both innate and adaptive components of the immune system (8). During the pre-clinical or acute phase, the first wave of immune cells mainly consisting of neutrophils, natural killer cells, and macrophages enter into the corneal stroma and help to clear the replicating virus (5). In the later clinical or chronic phase of the disease, CD4 T cells start to appear in the cornea around day 6–7 post-infection, a stage when virus is usually already cleared from the cornea (8). The CD4 T cells are considered to be the primary orchestrators of SK lesions as they facilitate the influx of the second wave of neutrophils (15). The massive cellular infiltration especially neutrophils coupled with the inflammatory mediators secreted by the immune cells are primarily responsible for the swelling and destruction of the cornea (16, 17).

Role of Th1, Th17, and CD8 T Cells in SK Lesions

Stromal keratitis (SK) is an immunopathological disease orchestrated by T cells (14). This view is supported by findings which show that mice depleted of T cells are less susceptible to HSV-1 induced corneal stromal disease. In both humans and mice, there is a predominance of CD4 T cells in the ocular tissues during SK and their functional activities are often associated with the tissue damage in the corneal stroma. In mice, CD4 T cells appear in the corneas around day 6 post-ocular infection with HSV-1 and their numbers continue to increase during the latter stage of SK development. Among the CD4 T cell population, there is a preferential accumulation of CD4 T helper (Th1) subset in the eye (18). Th1 cells express the transcription factor, T-bet, and produce various immune-modulatory mediators which play a role in SK lesion expression. The Th1 cells secrete the cytokines IFN-γ and IL-2 which are capable of inducing corneal inflammation and neovascularization (19, 20). In addition, these cytokines also modulate chemokine factors, and in doing so could facilitate the massive influx of neutrophils and macrophages into the cornea during the latter phase of SK development (21, 22). Another CD4 subset which gained recent prominence in inflammation and autoimmunity are the Th17 cells (23). These cells express the transcription factor ROR-γt and produce cytokines such as IL-17, IL-21, and IL-22. They preferentially produce IL-17 which is a potent inducer of additional pro-inflammatory cytokines, chemokines, and metalloproteinases (24, 25). Th17 cells accumulate in the HSV infected cornea during the later stages of SK pathogenesis and help sustain and expand the disease (26, 27). Moreover, HSV-1 ocular infection of IL-17R knock-out mice or neutralization of IL-17 using monoclonal antibodies delayed disease progression and reduced the severity of HSK (26). Importantly, IL-17 was expressed in corneas of patients with SK (28). In addition, the human corneal fibroblasts constitutively express the IL-17R. The data from these studies suggest that IL-17 strongly induces the production of key inflammatory mediators such as IL-6, IL-8, and matrix mettalloproteinase-1 in the human corneal fibroblast cultures (28). Thus, Th17 cells through the production of IL-17 modulate the levels of chemotactic factors such as CXCL-1 and IL-8 and influence the migration of neutrophils into the inflamed corneal tissues (26).

Although, CD4 T cells are considered to be the chief perpetuators of SK, the data presented in some experimental models implicate CD8 T cells in the pathogenesis of SK. The outcome depends to a large extent on the virus strain used for the studies. Some studies found that ocular infection of mice with the HSV-1 RE strain mainly induces SK mediated by CD4 T cells, whereas infection of the same stain of mice with HSV-1 KOS show SK which is dependent on CD8 T cells (29). In mice infected with a recombinant stain of HSV-1 (HSV-gK), the corneal scaring and the corneal disease were mainly mediated by CD8 T cells (30, 31). Results from these studies suggest that gK strongly induces CD8 T cell responses leading to exacerbation of SK lesions. Of note, the recombinant HSV-gK strain used in these studies contains three copies of glycoprotein K (gk) (a protein essential for virus replication) compared to one copy in the wild type HSV-1 McKrae strain (30). The HSV-1 mutant strains which lack gK were found to be defective in infectivity and failed to establish latency in the neurons in mouse models which suggests that gK expression is crucial for virus replication (32). Thus, the respective roles of different CD4 and CD8 subsets in SK is not clear and remains an unresolved issue. Additionally, some evidence shows that CD8 T cells mainly play more of a protective role (33). Observations in both mice and humans show that HSV-1 specific CD8 T cells are selectively retained in the TG and might help control HSV reactivation (3436). These tissue resident CD8 T cells appear to use IFN-γ and non-cytolytic mechanisms to block virus reactivation in the TG (37, 38).

Role of Regulatory T Cells (Treg) in SK Pathogenesis

A beneficial subset of CD4 T cells in SK are regulatory T cells (Treg) (39, 40). Treg express the master transcription factor, Foxp3 which controls their development, and function (41). Treg are either produced as a functionally mature T cell sub population in the thymus (natural Treg) or are induced in the periphery from naive CD4 T cells (induced Treg). Treg mainly function to maintain tolerance to self-antigens and prevent autoimmune diseases (42). They also constrain excessive immune responses to non-self-antigens or infectious agents and help to maintain peripheral tolerance and immune homeostasis (41). Treg use several mechanisms to suppress aberrant immune responses and these include immunomodulatory cytokines (IL-10, TGF-β, IL-35) or contact dependent suppression (granzyme/perforin) (41, 43, 44). In addition, Tregs also exert their function on effector T cells through inhibitory molecules such as CTLA-4. Treg also condition dendritic cells to secrete indoleamine 2,3-dioxygenase, a molecule which suppresses the activation of effector T cells (44).

During microbial infections, a major function of Treg is to control the excessive inflammatory responses to prevent collateral tissue damage and limit injury to the host. In HSV-1 ocular infection, Treg were shown to be crucial to control HSV induced corneal immunopathology. SK lesions were more severe if mice were depleted of Treg before infection using monoclonal antibody treatment, whereas adoptive transfer of in vitro converted Treg suppressed HSK severity (45, 46). Furthermore, findings using the depletion of regulatory T cells (DEREG) transgenic mice showed that lesions became more severe even when depletion was begun in the later phases (clinical/chronic phase) of the disease (47). The DEREG mice carry the diphtheria toxin receptor-enhanced green fluorescent protein (DTR-eGFP) transgene under the control of an additional Foxp3 promoter, which facilitates specific depletion of Treg by application of diphtheria toxin at any chosen point of time (48). Thus, measures to expand the representation of Treg by the administration of various reagents have been useful in reducing the severity of SK lesions in the mouse model. One such approach used was galectin-9 which induces apoptosis of pathogenic CD4 Th1 cells and increases the representation of the anti-inflammatory Treg population (49). In addition, a combination treatment using a tumor necrosis factor receptor superfamily member 25 (TNFRSF25) agonist antibody which expands Treg numbers along with galectin-9 was particularly effective in diminishing HSV-1 induced corneal immunopathology (50). Other approaches that were successful in expanding Treg population and reducing SK lesions included the use of IL-2/anti-IL-2 mAb complexes and the fungal metabolite drug, fingolimod hydrochloride (FTY720) (51, 52). In addition, phosphorylated FTY720 also targets sphingosine-1-phosphate receptor and perhaps diminishes inflammation by modulating lymphocyte trafficking (53).

Although increasing the representation of Treg in lesions is a valuable approach to minimize lesion severity, it has become evident that the Treg population is functionally heterogeneous. Accordingly, some functions are more valuable to achieve control than others. For example, our group recently observed that a function of Treg valuable for resolving SK lesions is their ability to produce amphiregulin (AMP) (54). This molecule acts to facilitate tissue repair by binding to the epidermal growth factor receptor expressed mainly on epithelial cells and stem cells and its binding can result in the activation of downstream signaling kinases resulting in growth, proliferation, and migration of cells (55). Treg that produce AMP are relatively infrequent in the early stages of SK, but their representation is most evident in later stages. The change of Treg function to become AMP producers appears to be driven by the cytokines IL-12 and IL-18. In fact, exposure of AMP negative Treg cells in vitro to these cytokines can induce them to become AMP producers. In addition, if animals were treated in vivo with a plasmid which expresses IL-18, this led to the reduced expression of SK lesions, an effect that correlated with a higher frequency of Treg that were AMP producers (54). Finding practical approaches to induce cells in SK to become AMP producers could represent a useful approach to therapy, an issue that merits further investigation.

Plasticity of Regulatory T Cell Populations

Some recent observations suggest that Treg might become unstable in certain highly inflammatory environments and lose their regulatory activity (56). Under such conditions, Treg that downregulate Foxp3 expression might even take up an effector phenotype and start producing pro-inflammatory cytokines such as IFN-γ and IL-17 Treg, a phenomenon commonly referred to as plasticity (5759). In recent times, plasticity in T cells has been a matter of debate as it has biological implications especially in therapeutic regimens which use Treg (60, 61). Factors which influence Treg stability are as yet not clear and remains an active area of research. Although multiple mechanism might be involved in the stability and plasticity of Treg, most evidence indicates that Treg stability and Foxp3 expression is controlled by epigenetic mechanisms, namely DNA methylation in the non-coding region (CNS2) of the Foxp3 gene locus, also known as Treg-specific demethylation region (TSDR) (62). Any changes or modifications in the DNA methylation status in the TSDR region tend to have an effect on Foxp3 expression and stability of Treg populations (63). Most Treg populations are generally resistant to destabilization and reprogramming and maintain their transcriptional expression of regulatory genes and functional phenotype (61). Some of the Tregs generated in vitro or in vivo which have incomplete demethylation status in the cytosine-phospho-guanine (CpG) sites in the TSDR region are more prone to instability when exposed to cytokine milieu containing IL-6, IL-12, IL-21, or IL-23 (57, 64). The Bluestone group, using Foxp3-Cre reporter mice in an Experimental autoimmune encephalomyelitis (EAE) model observed that some of the Treg cells downregulated Foxp3 expression and these were referred to as exFoxp3 cells (59). Such exFoxp3 cells isolated from the CNS at the peak of the response produced IFN- γ when stimulated with cognate antigen (59). Our group using fate mapping mice showed that Treg plasticity can occur in HSV-1-induced inflammatory environment and such Treg may contribute to SK lesion severity by secreting the proinflammatory cytokine IFN-γ (65). In particular, Treg cells showing low expression of the IL-2R (CD25) could exhibit instability, in part due to the exposure to the pro-inflammatory cytokine IL-12 in the cornea (65). In such circumstances, drugs such as azacytidine, retinoic acid, and vitamin C which maintain demethylation of the TSDR region of Foxp3, can be helpful in promoting the stability and improving the functionality of Treg especially under chronic inflammatory conditions (65). In fact, in a recent study, Treg generated in vitro in the presence of Azacytidine expressed a fully demethylated TSDR and these cells displayed enhanced suppressive activity (66). Moreover, administration of 5-Azacytidine reduced the incidence of SK lesions in mice infected ocularly with HSV-1 (66).

Manipulating Metabolism to Constrain SK Lesions

In the previous section, we have argued that the clinical expression of SK is affected by the representation of different participants in lesions. When the T cell participants were dominated by Treg, lesions will be less severe and may even resolve. Hence, a potentially valuable approach to therapy is to use maneuvers that can shift the balance of events away from dominance by proinflammatory components. This therapeutic challenge is also faced by those working with other in other chronic inflammatory diseases, especially autoimmune diseases (AID). In the AID field, some are considering using approaches such as adoptive cell transfer to enrich the population of Treg (67). However, such an approach, which is most effective when the Treg are antigen specific, would likely fail to adequately gain access to the eye. Other approaches include administering reagents that expand the Treg population as we discussed previously. A potentially more useful therapeutic option would be to exploit the accumulating knowledge that cells involved in immune function may differ in the major metabolic pathways they use to provide them with energy and other events that maintain of their various functions (68, 69). For example, proinflammatory and Treg cells use different pathways to provide energy with the former mainly use extracellular glucose and Treg rely on fatty acid oxidation (68). Rathmell's group reported that effector T cells (both CD4 and CD8) express high levels of the glucose transporter Glut1 and utilize the mammalian target of rapamycin (mTOR) pathway to increase glycolysis to support their function (70). In contrast, Treg primarily use AMP-activated protein kinase and rely upon lipid oxidation for their energy. The activated AMPK pathway in Treg acts to inhibit mTOR by suppressing mTOR signaling and promotes mitochondrial oxidative metabolism rather than glycolysis and is considered to be anti-inflammatory (70). In our own studies, we have begun to exploit the differences by which proinflammatory and Treg cells derive their energy needs. We have shown that if glucose utilization is inhibited, as can be achieved by the use of 2 deoxy glucose administration from the initial time of lesion development, that lesions are significantly reduced (71). The outcome occurred because the activity of proinflammatory cells such as Th1 and Th17 cells were inhibited, but Treg were unaffected. Thus, the representation of the two populations changed with Treg becoming enriched (71). Findings from another group demonstrated the importance of hypoxia associated glycolytic molecules in SK pathogenesis (72). Besides glycolytic metabolism, T effectors, and Treg also show differences in amino acid metabolism. Amino acids, particularly glutamine, plays a key role in fueling effector T cell differentiation, whereas Treg are less dependent on amino acids for their energy (68). In addition, microbial metabolites such as short chain fatty acids or diets rich in vitamin A promote Treg differentiation and function in the gut (73, 74). Additional metabolic differences are also under investigation such as the differential use of lipid oxidation and synthesis pathways. Thus, manipulating metabolic pathways to influence inflammatory lesions is in the early stages of investigation but the approach has great potential and could be more affordable than many of the alternatives. However, the strategy will need considerable scrutiny especially if used for long term therapy. Indeed, our own studies have already documented some untoward consequences when glucose metabolism is compromised during the time when virus is actively replicating.

Contribution of Corneal Nerve Damage to SK Pathology

Following corneal infection, HSV-1 replicates in the epithelial cells and gains access to the sensory nerve endings which drain the corneal tissues and can travel up (retrograde) to the TG where the virus establishes latency. The virus travels back (anterograde) from the TG to the cornea through the sensory nerves after reactivation. HSV-1 corneal infection can result in destruction of corneal nerve endings resulting in loss of corneal sensitivity (75). Such loss of corneal sensation and nerve function is one of the hall marks of SK in humans and is commonly referred to as neurotrophic keratopathy (76). Evidence from recent studies in mice have shown that sympathetic nerves innervate the cornea and replace the sensory nerve endings lost after HSV-1 corneal infection (75). These sympathetic nerves enhance the infiltration of immune cells resulting in severe corneal inflammation and pathology. A surgical procedure called superior cervical ganglionectomy (SCGx) that removes sympathetic nerves from the cornea helped to alleviate SK severity. Of note, after the SCGx procedure, the sensory nerves reinnervated the cornea resulting in the restoration of corneal sensitivity (75). The exact mechanisms involved in sympathetic corneal innervation are not known and this aspect requires further examination. It is likely that immune cells such as CD4 T cells could play a key role, as their depletion resulted in reversing nerve damage (77). Findings from another study suggest that the molecule involved in cell migration, semaphorin 7A might play a role in the corneal nerves degeneration and regeneration process in HSV-1 infected mice (78). The cytokine IL-6 produced during the inflammatory response to HSV-1 infection in the cornea might also be responsible for causing corneal sensory nerve damage (79).

Concluding Remarks

Stromal keratitis (SK) caused by HSV-1 corneal infection is a debilitating disease and one of the major causes of vision loss due to an infectious agent. As T cells are the primary orchestrators of SK, steps to improve the host environment which favors Treg over pathogenic Th1/Th17 cells is likely to help ease the severity of SK lesions. In addition, it is becoming increasingly clear from recent developments that metabolism plays a key role in immune function. Thus, as discussed in this review, understanding the events involved in pathogenesis along with key molecules and metabolic pathways involved in inflammation and applying this knowledge to develop better therapies might help control SK in the future.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

Research was supported by National Institutes of Health Grant R01EY005093 to BR.

Conflict of Interest Statement

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

Acknowledgments

We also thank the department of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville for supporting the research work through the COE funds to NR.

References

1. Looker KJ, Magaret AS, May MT, Turner KM, Vickerman P, Gottlieb SL, et al. Global and regional estimates of prevalent and incident herpes simplex virus type 1 infections in 2012. PLoS ONE. (2015) 10:e0140765. doi: 10.1371/journal.pone.0140765

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Johnston C, Gottlieb SL, Wald A. Status of vaccine research and development of vaccines for herpes simplex virus. Vaccine. (2016) 34:2948–52. doi: 10.1016/j.vaccine.2015.12.076

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Whitley RJ, Roizman B. Herpes simplex virus infections. Lancet. (2001) 357:1513–8. doi: 10.1016/S0140-6736(00)04638-9

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Liesegang TJ. Herpes simplex virus epidemiology and ocular importance. Cornea. (2001) 20:1–13. doi: 10.1097/00003226-200101000-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Rowe AM, St Leger AJ , Jeon S, Dhaliwal DK, Knickelbein JE, Hendricks RL. Herpes keratitis. Prog Retin Eye Res. (2013) 32:88–101. doi: 10.1016/j.preteyeres.2012.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Young RC, Hodge DO, Liesegang TJ, Baratz KH. Incidence, recurrence, and outcomes of herpes simplex virus eye disease in olmsted county, minnesota, 1976-2007: the effect of oral antiviral prophylaxis. Arch Ophthalmol. (2010) 128:1178–83. doi: 10.1001/archophthalmol.2010.187

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Stanzel TP, Diaz JD, Mather R, Wong IG, Margolis TP, Gritz DC. The epidemiology of herpes simplex virus eye disease in Northern California. Ophthalmic Epidemiol. (2014) 21:370–7. doi: 10.3109/09286586.2014.966848

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Biswas PS, Rouse BT. Early events in HSV keratitis–setting the stage for a blinding disease. Microbes Infect. (2005) 7:799–810. doi: 10.1016/j.micinf.2005.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Roizman B, Whitley RJ. An inquiry into the molecular basis of HSV latency and reactivation. Annu Rev Microbiol. (2013) 67:355–74. doi: 10.1146/annurev-micro-092412-155654

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Koyuncu OO, MacGibeny MA, Enquist LW. Latent versus productive infection: the alpha herpesvirus switch. Future Virol. (2018) 13:431–43. doi: 10.2217/fvl-2018-0023

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Koyuncu OO, Hogue IB, Enquist LW. Virus infections in the nervous system. Cell Host Microbe. (2013) 13:379–93. doi: 10.1016/j.chom.2013.03.010

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Knickelbein JE, Hendricks RL, Charukamnoetkanok P. Management of herpes simplex virus stromal keratitis: an evidence-based review. Surv Ophthalmol. (2009) 54:226–34. doi: 10.1016/j.survophthal.2008.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Gimenez F, Suryawanshi A, Rouse BT. Pathogenesis of herpes stromal keratitis–a focus on corneal neovascularization. Prog Retin Eye Res. (2013) 33:1–9. doi: 10.1016/j.preteyeres.2012.07.002

CrossRef Full Text | Google Scholar

14. Rajasagi NK, Rouse BT. Application of our understanding of pathogenesis of herpetic stromal keratitis for novel therapy. Microbes Infect. (2018) 20:526–30. doi: 10.1016/j.micinf.2017.12.014

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Doymaz MZ, Rouse BT. Herpetic stromal keratitis: an immunopathologic disease mediated by CD4+ T lymphocytes. Invest Ophthalmol Vis Sci. (1992) 33:2165–73.

PubMed Abstract | Google Scholar

16. Thomas J, Gangappa S, Kanangat S, Rouse BT. On the essential involvement of neutrophils in the immunopathologic disease: herpetic stromal keratitis. J Immunol. (1997) 158:1383.

PubMed Abstract | Google Scholar

17. Daheshia M, Kanangat S, Rouse BT. Production of key molecules by ocular neutrophils early after herpetic infection of the cornea. Exp Eye Res. (1998) 67:619–24. doi: 10.1006/exer.1998.0565

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Niemialtowski MG, Rouse BT. Predominance of Th1 cells in ocular tissues during herpetic stromal keratitis. J Immunol. (1992) 149:3035–9.

PubMed Abstract | Google Scholar

19. Epstein RJ, Hendricks RL, Stulting RD. Interleukin-2 induces corneal neovascularization in A/J mice. Cornea. (1990) 9:318–23. doi: 10.1097/00003226-199010000-00009

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Hendricks RL, Tumpey TM, Finnegan A. IFN-gamma and IL-2 are protective in the skin but pathologic in the corneas of HSV-1-infected mice. J Immunol. (1992) 149:3023–8.

PubMed Abstract | Google Scholar

21. Tang Q, Hendricks RL. Interferon gamma regulates platelet endothelial cell adhesion molecule 1 expression and neutrophil infiltration into herpes simplex virus-infected mouse corneas. J Exp Med. (1996) 184:1435–47. doi: 10.1084/jem.184.4.1435

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Tang Q, Chen W, Hendricks RL. Proinflammatory functions of IL-2 in herpes simplex virus corneal infection. J Immunol. (1997) 158:1275–83.

PubMed Abstract | Google Scholar

23. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 Cells. Annu Rev Immunol. (2009) 27:485–517. doi: 10.1146/annurev.immunol.021908.132710

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Damsker JM, Hansen AM, Caspi RR. Th1 and Th17 cells: adversaries and collaborators. Ann N Y Acad Sci. (2010) 1183:211–21. doi: 10.1111/j.1749-6632.2009.05133.x

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Xu S, Cao X. Interleukin-17 and its expanding biological functions. Cell Mol Immunol. (2010) 7:164–74. doi: 10.1038/cmi.2010.21

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Suryawanshi A, Veiga-Parga T, Rajasagi NK, Reddy PB, Sehrawat S, Sharma S, et al. Role of IL-17 and Th17 cells in herpes simplex virus-induced corneal immunopathology. J Immunol. (2011) 187:1919–30. doi: 10.4049/jimmunol.1100736

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Suryawanshi A, Veiga-Parga T, Reddy PB, Rajasagi NK, Rouse BT. IL-17A differentially regulates corneal vascular endothelial growth factor (VEGF)-A and soluble VEGF receptor 1 expression and promotes corneal angiogenesis after herpes simplex virus infection. J Immunol. (2012) 188:3434–46. doi: 10.4049/jimmunol.1102602

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Maertzdorf J, Osterhaus AD, Verjans GM. IL-17 expression in human herpetic stromal keratitis: modulatory effects on chemokine production by corneal fibroblasts. J Immunol. (2002) 169:5897–903. doi: 10.4049/jimmunol.169.10.5897

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Hendricks RL, Tumpey TM. Contribution of virus and immune factors to herpes simplex virus type I-induced corneal pathology. Invest Ophthalmol Vis Sci. (1990) 31:1929–39.

PubMed Abstract | Google Scholar

30. Mott KR, Perng GC, Osorio Y, Kousoulas KG, Ghiasi H. A recombinant herpes simplex virus type 1 expressing two additional copies of gK is more pathogenic than wild-type virus in two different strains of mice. J Virol. (2007) 81:12962–72. doi: 10.1128/JVI.01442-07

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Jaggi U, Wang S, Tormanen K, Matundan H, Ljubimov AV, Ghiasi H. Role of Herpes Simplex Virus Type 1 (HSV-1) Glycoprotein K (gK) Pathogenic CD8(+) T Cells in Exacerbation of Eye Disease. Front Immunol. (2018) 9:2895. doi: 10.3389/fimmu.2018.02895

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Saied AA, Chouljenko VN, Subramanian R, Kousoulas KG. A replication competent HSV-1(McKrae) with a mutation in the amino-terminus of glycoprotein K (gK) is unable to infect mouse trigeminal ganglia after cornea infection. Curr Eye Res. (2014) 39:596–603. doi: 10.3109/02713683.2013.855238

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Zhu J, Peng T, Johnston C, Phasouk K, Kask AS, Klock A, et al. Immune surveillance by CD8alphaalpha+ skin-resident T cells in human herpes virus infection. Nature. (2013) 497:494–7. doi: 10.1038/nature12110

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Khanna KM, Lepisto AJ, Decman V, Hendricks RL. Immune control of herpes simplex virus during latency. Curr Opin Immunol. (2004) 16:463–9. doi: 10.1016/j.coi.2004.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Verjans GM, Hintzen RQ, van Dun JM, Poot A, Milikan JC, Laman JD, et al. Selective retention of herpes simplex virus-specific T cells in latently infected human trigeminal ganglia. Proc Natl Acad Sci U.S.A. (2007) 104:3496–501. doi: 10.1073/pnas.0610847104

PubMed Abstract | CrossRef Full Text | Google Scholar

36. St Leger AJ, Hendricks RL. CD8+ T cells patrol HSV-1-infected trigeminal ganglia and prevent viral reactivation. J Neurovirol. (2011) 17:528–34. doi: 10.1007/s13365-011-0062-1

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Liu T, Khanna KM, Carriere BN, Hendricks RL. Gamma interferon can prevent herpes simplex virus type 1 reactivation from latency in sensory neurons. J Virol. (2001) 75:11178–84. doi: 10.1128/JVI.75.22.11178-11184.2001

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Knickelbein JE, Khanna KM, Yee MB, Baty CJ, Kinchington PR, Hendricks RL. Noncytotoxic lytic granule-mediated CD8+ T cell inhibition of HSV-1 reactivation from neuronal latency. Science. (2008) 322:268–71. doi: 10.1126/science.1164164

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Rouse BT, Sehrawat S. Immunity and immunopathology to viruses: what decides the outcome? Nat Rev Immunol. (2010) 10:514–26. doi: 10.1038/nri2802

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Veiga-Parga T, Sehrawat S, Rouse BT. Role of regulatory T cells during virus infection. Immunol Rev. (2013) 255:182–96. doi: 10.1111/imr.12085

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. (2012) 30:531–64. doi: 10.1146/annurev.immunol.25.022106.141623

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Ohkura N, Kitagawa Y, Sakaguchi S. Development and maintenance of regulatory T cells. Immunity. (2013) 38:414–23. doi: 10.1016/j.immuni.2013.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. (2008) 8:523–32. doi: 10.1038/nri2343

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. (2009) 30:636–45. doi: 10.1016/j.immuni.2009.04.010

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Suvas S, Azkur AK, Kim BS, Kumaraguru U, Rouse BT. CD4+CD25+ regulatory T cells control the severity of viral immunoinflammatory lesions. J Immunol. (2004) 172:4123–32. doi: 10.4049/jimmunol.172.7.4123

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Sehrawat S, Suvas S, Sarangi PP, Suryawanshi A, Rouse BT. In vitro-generated antigen-specific CD4+ CD25+ Foxp3+ regulatory T cells control the severity of herpes simplex virus-induced ocular immunoinflammatory lesions. J Virol. (2008) 82:6838–51. doi: 10.1128/JVI.00697-08

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Veiga-Parga T, Suryawanshi A, Mulik S, Gimenez F, Sharma S, Sparwasser T, et al. On the role of regulatory T cells during viral-induced inflammatory lesions. J Immunol. (2012) 189:5924–33. doi: 10.4049/jimmunol.1202322

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Lahl K, Sparwasser T. In vivo depletion of FoxP3+ Tregs using the DEREG mouse model. Methods Mol Biol. (2011) 707:157–72. doi: 10.1007/978-1-61737-979-6_10

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Sehrawat S, Suryawanshi A, Hirashima M, Rouse BT. Role of Tim-3/galectin-9 inhibitory interaction in viral-induced immunopathology: shifting the balance toward regulators. J Immunol. (2009) 182:3191–201. doi: 10.4049/jimmunol.0803673

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Reddy PBJ, Schreiber TH, Rajasagi NK, Suryawanshi A, Mulik S, Veiga-Parga T, et al. TNFRSF25 agonistic antibody and galectin-9 combination therapy controls herpes simplex virus-induced immunoinflammatory lesions. J Virol. (2012) 86:10606–20. doi: 10.1128/JVI.01391-12

CrossRef Full Text | Google Scholar

51. Sehrawat S, Rouse BT. Anti-inflammatory effects of FTY720 against viral-induced immunopathology: role of drug-induced conversion of T cells to become Foxp3+ regulators. J Immunol. (2008) 180:7636–47. doi: 10.4049/jimmunol.180.11.7636

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Gaddipati S, Estrada K, Rao P, Jerome AD, Suvas S. IL-2/anti-IL-2 antibody complex treatment inhibits the development but not the progression of herpetic stromal keratitis. J Immunol. (2015) 194:273–82. doi: 10.4049/jimmunol.1401285

CrossRef Full Text | Google Scholar

53. Brinkmann V. Sphingosine 1-phosphate receptors in health and disease: mechanistic insights from gene deletion studies and reverse pharmacology. Pharmacol Ther. (2007) 115:84–105. doi: 10.1016/j.pharmthera.2007.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Varanasi SK, Rajasagi NK, Jaggi U, Rouse BT. Role of IL-18 induced Amphiregulin expression on virus induced ocular lesions. Mucosal Immunol. (2018). doi: 10.1038/s41385-018-0058-8

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Zaiss DMW, Gause WC, Osborne LC, Artis D. Emerging functions of amphiregulin in orchestrating immunity, inflammation, and tissue repair. Immunity. (2015) 42:216–26. doi: 10.1016/j.immuni.2015.01.020

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Bailey-Bucktrout SL, Bluestone JA. Regulatory T cells: stability revisited. Trends Immunol. (2011) 32:301–6. doi: 10.1016/j.it.2011.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Zhou L, Chong MM, Littman DR. Plasticity of CD4+ T cell lineage differentiation. Immunity. (2009) 30:646–55. doi: 10.1016/j.immuni.2009.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Zhou X, Bailey-Bucktrout SL, Jeker LT, Penaranda C, Martinez-Llordella M, Ashby M, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. (2009) 10:1000–7. doi: 10.1038/ni.1774

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Bailey-Bucktrout SL, Martinez-Llordella M, Zhou X, Anthony B, Rosenthal W, Luche H, et al. Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response. Immunity. (2013) 39:949–62. doi: 10.1016/j.immuni.2013.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Hori S. Regulatory T cell plasticity: beyond the controversies. Trends Immunol. (2011) 32:295–300. doi: 10.1016/j.it.2011.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Sakaguchi S, Vignali DA, Rudensky AY, Niec RE, Waldmann H. The plasticity and stability of regulatory T cells. Nat Rev Immunol. (2013) 13:461–7. doi: 10.1038/nri3464

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Kitagawa Y, Sakaguchi S. Molecular control of regulatory T cell development and function. Curr Opin Immunol. (2017) 49:64–70. doi: 10.1016/j.coi.2017.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Lal G, Bromberg JS. Epigenetic mechanisms of regulation of Foxp3 expression. Blood. (2009) 114:3727–35. doi: 10.1182/blood-2009-05-219584

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Hori S. Lineage stability and phenotypic plasticity of Foxp3(+) regulatory T cells. Immunol Rev. (2014) 259:159–72. doi: 10.1111/imr.12175

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Bhela S, Varanasi SK, Jaggi U, Sloan SS, Rajasagi NK, Rouse BT. The plasticity and stability of regulatory T cells during viral-induced inflammatory lesions. J Immunol. (2017) 199:1342–52. doi: 10.4049/jimmunol.1700520

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Varanasi SK, Reddy PB, Bhela S, Jaggi U, Gimenez F, Rouse BT. Azacytidine treatment inhibits the progression of herpes stromal keratitis by enhancing regulatory T cell function. J Virol. (2017) 91:e02367-16. doi: 10.1128/JVI.02367-16

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Wright GP, Notley CA, Xue S-A, Bendle GM, Holler A, Schumacher TN, et al. Adoptive therapy with redirected primary regulatory T cells results in antigen-specific suppression of arthritis. Proc Natl Acad Sci USA. (2009) 106:19078. doi: 10.1073/pnas.0907396106

PubMed Abstract | CrossRef Full Text | Google Scholar

68. O'Neill LAJ, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol. (2016) 16:553. doi: 10.1038/nri.2016.70

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Bettencourt IA, Powell JD. Targeting metabolism as a novel therapeutic approach to autoimmunity, inflammation, and transplantation. J Immunol. (2017) 198:999. doi: 10.4049/jimmunol.1601318

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J. Immunol. (2011) 186:3299–303. doi: 10.4049/jimmunol.1003613

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Varanasi SK, Donohoe D, Jaggi U, Rouse BT. Manipulating glucose metabolism during different stages of viral pathogenesis can have either detrimental or beneficial effects. J Immunol. (2017) 199:1748–61. doi: 10.4049/jimmunol.1700472

CrossRef Full Text | Google Scholar

72. Rao P, Suvas S. Development of inflammatory hypoxia and prevalence of glycolytic metabolism in progressing herpes stromal keratitis lesions. J Immunol. (2019) 202:514–26. doi: 10.4049/jimmunol.1800422

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y, M, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. (2013) 341:569–73. doi: 10.1126/science.1241165

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Zeng H, Chi H. Metabolic control of regulatory T cell development and function. Trends Immunol. (2015) 36:3–12. doi: 10.1016/j.it.2014.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Yun H, Lathrop KL, Hendricks RL. A central role for sympathetic nerves in herpes stromal keratitis in mice. Invest Ophthalmol Vis Sci. (2016) 57:1749–56. doi: 10.1167/iovs.16-19183

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Hamrah P, Cruzat A, Dastjerdi MH, Zheng L, Shahatit BM, Bayhan HA, et al. Corneal sensation and subbasal nerve alterations in patients with herpes simplex keratitis: an in vivo confocal microscopy study. Ophthalmology. (2010) 117:1930–6. doi: 10.1016/j.ophtha.2010.07.010

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Yun H, Rowe AM, Lathrop KL, Harvey SA, Hendricks RL. Reversible nerve damage and corneal pathology in murine herpes simplex stromal keratitis. J Virol. (2014) 88:7870–80. doi: 10.1128/JVI.01146-14

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Chucair-Elliott AJ, Zheng M, Carr DJ. Degeneration and regeneration of corneal nerves in response to HSV-1 infection. Invest Ophthalmol Vis Sci. (2015) 56:1097–107. doi: 10.1167/iovs.14-15596

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Chucair-Elliott AJ, Jinkins J, Carr MM, Carr DJ. IL-6 contributes to corneal nerve degeneration after herpes simplex virus type I infection. Am J Pathol. (2016) 186:2665–78. doi: 10.1016/j.ajpath.2016.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: herpes stromal keratitis, CD4 T cells, metabolism, regulatory T cells, plasticity

Citation: Rajasagi NK and Rouse BT (2019) The Role of T Cells in Herpes Stromal Keratitis. Front. Immunol. 10:512. doi: 10.3389/fimmu.2019.00512

Received: 09 November 2018; Accepted: 26 February 2019;
Published: 19 March 2019.

Edited by:

Susmit Suvas, Wayne State University, United States

Reviewed by:

Paul R. Kinchington, University of Pittsburgh, United States
Homayon Ghiasi, Cedars-Sinai Medical Center, United States

Copyright © 2019 Rajasagi and Rouse. 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: Barry T. Rouse, btr@utk.edu

Disclaimer: 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.