Skip to main content
Clinical and Developmental Immunology logoLink to Clinical and Developmental Immunology
. 2012 Apr 23;2012:728480. doi: 10.1155/2012/728480

Recurrent Herpetic Stromal Keratitis in Mice: A Model for Studying Human HSK

Patrick M Stuart 1,*, Tammie L Keadle 2
PMCID: PMC3347728  PMID: 22593769

Abstract

Herpes simplex virus 1 (HSV-1) infection of the cornea leads to a potentially blinding disease, termed herpetic stromal keratitis (HSK) that is characterized by lesions of an immunoinflammatory nature. In spite of the fact that HSK typically presents as a recurrent disease due to reactivation of virus which latently infects the trigeminal ganglia, most murine studies of HSK have employed a primary and not recurrent model of the disease. This report documents the several recurrent models of HSK that have been developed and how data generated from these models differs in some important aspects from data generated following primary infection of the cornea. Chief among these differences is the fact that recurrent HSK takes place in the context of an animal that has a preexisting anti-HSV immune response, while primary HSK occurs in an animal that is developing such a response. We will document both differences and similarities that derive from this fundamental difference in these models with an eye towards possible vaccines and therapies that demonstrate promise in treating HSK.

1. Opening Comments

Herpetic stromal keratitis (HSK) is a potentially blinding corneal inflammation that accompanies herpes simplex virus type 1 (HSV-1) infection of the eye. The disease course in HSK begins with a primary infection by HSV followed by a period during which the virus enters latency in sensory and autonomic ganglia. Many studies have shown that clinical disease is the result of a cocktail of inflammatory cells, consisting of PMN's, macrophages, and T cells (both CD4+ and CD8+) that are recruited to the corneas of patients with HSK [14].

Most animal studies of HSK have focused on primary ocular infection. The major drawback with extrapolating data from primary HSV infection in mice is that it often does not manifest corneal lesions characteristic of human primary or recurrent HSK [5]. We believe that there are four advantages in using a recurrent model of HSK. The first is that recurrent human disease is most often associated with corneal scarring [6, 7]. Second, the clinical profile in the murine recurrent model mimics many of the symptoms observed in human disease [8]. Namely, that primary infection resulted in multiple epithelial dendrites, followed by diffuse stromal opacification, while recurrent infection presented clinical features that included microdendrites, focal stromal opacities, disciform endotheliitis, and corneal neovascularization, which were more similar to those observed in human disease. Third, the model allows reactivation to occur in the context of an immune host. This is also the case in humans where disease takes place in a host that has developed an adaptive immune response against HSV-1. While it is not suggested that such adaptive responses will be identical, they will likely be more similar than extending what occurs following primary infection, where an adaptive response is initially developing (most murine studies), to what is taking place in an “immune host” following reactivation. Finally, a recurrent model lends itself to testing the efficacy of HSV vaccines. Since human disease typically occurs following reactivation, identifying vaccines that are effective when used therapeutically would be very valuable and a recurrent model allows for that evaluation. This is critical as many vaccines which show efficacy in primary HSK models fail when tested in a reactivation paradigm [9].

There are several different protocols that have been used to reactivate mice from latency. One of the first methods used to reactivate mice latently infected was treating mice with the immunosuppressive drug cyclophosphamide (CycloP) [10]. Later Shimeld et al. developed a model in which mice are infected with the McKrae strain of HSV-1 and given passive immunization in the form of pooled serum. The eyes of these latently infected mice are exposed to UV-B irradiation at least 30 days following primary infection to induce reactivation [1114]. The reason for the addition of passive immunization is that this reduced the high incidence of mortality and also prevented acute HSK, which would lead to permanent corneal damage [13]. It was also noted that the latent infection was restricted to the ophthalmic part of the trigeminal ganglia [13]. When Shimeld et al. [12] and later Laycock et al. [14] compared UV-B irradiation to CycloP + dexamethasone, the incidence of virus shedding from the cornea was greater for the UV-B-treated mice and recurrent disease did was more easily apparent in UV-B-treated mice [12]. Different virus strains were compared for their ability to reactivate, and the McKrae strain of HSV-1 demonstrated the most consistent reactivation phenotype [1113]. In addition, various strains of mice have been tested for their ability to reactivate following UV-B irradiation and the NIH strain of mice consistently displays the highest rate of reactivation (70–90%) as determined by detection of infectious virus in tear film [9, 14, 15]. In addition, severity of disease also varies between different mouse strains with NIH and BALB/c mice showing severe disease and C57BL/6 mice with much less disease [9, 15, 16]. Other investigators have also developed other means of reactivating a latent HSV-1 infection by using hyperthermia shock [17, 18] and most recently sodium butyrate [19]. The hyperthermia shock model is used to determine reactivation within the infected ganglion neurons and thus is a very good model to study molecular events that occur there following reactivating stress [17, 18]. However it has not been exploited to study corneal disease. The sodium butyrate model, which does result in viral shedding from the corneal surface [19], has also not been exploited to study corneal disease. Thus, while each of these techniques has their own distinct advantages and disadvantages, the UV-B model has been used much more extensively for pathogenic and immunological studies of recurrent HSK. Thus this paper will restrict itself to the reactivation model whereby UV-B light is used to reactivate virus from latency. This was chosen as the vast majority of data concerning the pathogenesis of recurrent disease has used that model.

As a basis for comparison, a few factors should be mentioned. The first being that different investigators use a variety of strains of HSV-1 when studying primary HSK. The virus strains that have been used for these studies include, but are not restricted to, RE [4, 20, 21], Strain 17 [19], McKrae [8, 15, 22], CH394 [23], and KOS [15, 24]. In addition to a variety of HSV-1 strains being used, various mouse strains have also been used for these studies. Most studies employ C57BL/6 or BALB/c mouse strains or gene-targeted mice that are bred to these two strains. It should be noted that while infections with these various strains of HSV-1 display differences in corneal pathology and neurovirulence, the immunological components of corneal disease are remarkably similar when compared on the basis of virus strain used or the strain of mouse being infected as the following discussion will illustrate.

2. Characteristics of Herpetic Stromal Keratitis

As previously mentioned, HSK is at its core an inflammatory disease with chemokines involved in migration of leukocytes to sites of infection and inflammation [25, 26] and cytokines responsible for the activation of cells which mediate the cellular destruction following their activation. Primary models have shown that during disease, a diverse set of host inflammatory cells infiltrate the cornea following HSV-1 infection. Included among these cells are macrophages [8, 9], natural killer cells [27], T cells [8, 20, 2833], polymorphonuclear neutrophils (PMNs), which are the predominant cell type early during primary infection with HSV-1 [34, 35], and corneal Langerhans Cells [36]. While the exact mechanism for primary HSK has not been conclusively determined, the disease is believed to be the result of the interaction of virus and host immune cells and components and not due to direct viral cytolysis of corneal cells [23]. Since much fewer studies have employed the recurrent model, even less is known about the mechanisms responsible for recurrent HSK.

3. Role of Chemokines in HSK

Chemokines are important factors in viral infections [37] and HSK [21, 3840]. These are small proteins which are made by resident tissue cells and/or immunocompetent cells and whose primary function is to direct the movement or chemotaxis of cells that bear receptors for the chemokine that is being produced. Thus during infection chemokines are released by cells at the site of infection to activate the migration of particular cells bearing appropriate chemokine receptors to that site of infection. Chemokines have also been implicated in activation and polarization of certain immunocompetent cells [41]. Due to these activities, they are potential targets for therapeutic intervention to reduce or prevent disease (see Table 1 for cells responsive to chemokines and references related to HSK). During primary HSK it has been shown that the production of IL-6 stimulates resident corneal cells to produce CCL3 and CXCL2 [42]. Studies have shown that targeting of CCL3 significantly reduces disease [39, 43], suggesting that it plays a role in corneal pathology during primary HSK. In contrast, when CCL3 is neutralized or absent during recurrent HSK, the resulting disease is worse [40]. Likewise, targeting CCL2 reduced primary [4446] but did not affect recurrent disease [40]. Eo et al. [45] coimmunized mice with a plasmid encoding gB of HSV along with various plasmids containing CCL1, CCL3, CCL4, and CXCL2, which are known to be produced by the cornea [42, 45, 47], to determine if the adaptive immune response generated was affected by the presence of specific chemokines. Their data indicates that the chemokines CCL1 and CCL4 biased immunity to a Th2-like response, while CXCL2 and CCL3 activated a Th1 response. Mikloska et al. observed increased levels of CCL4 and, to a lesser extent, CCL3 and CCL5 in human vesicle fluid from patients with herpetic corneal lesions [48]. Similar studies evaluating CXCL1 expression have shown that infection of corneal-derived cells with HSV-1 leads to significant increases in CXCL1 production, which stimulates neutrophil chemotaxis [47, 49]. It has been reported that CXCL10 is very important in recruiting activated T cells into sites of inflammation [50]. However, CXCL10 has been shown to be a mediator of IL-12-mediated antiangiogenesis during primary HSK [51, 52]. Thus it is possible that CXCL10 might be involved in both promoting disease, by attracting activated CD4+ T cells, and in preventing disease by inhibiting corneal angiogenesis. The role of murine CXCL1, CXCL10, and IL-6 play during recurrent HSK has not been determined; however, they are currently being investigated in our laboratory at this time.

Table 1.

Chemokine Chemotactic activity Reference to HSK
CCL1 (Kit ligand) Monocytes, NK cells, DC [45]
CCL2 (MCP-1) Monocytes, T cells, DC [40, 4446]
CCL3 (MIP-1α) Neutrophils [39, 40, 42, 43, 45, 48]
CCL4 (MIP-1β) Monocytes, NK cells [45]
CCL5 (Rantes) Eosinophils, NK cells, T cells [48]
CXCL1 (KC) Neutrophils [47, 49]
CXCL2 (MIP-2) Neutrophils [42, 45]
CXCL10 (IP-10) Monocytes, NK cells, T cells, DC [5052]

4. Cytokine Profile Expressed during HSK

In addition to chemokines, HSK is also the result of the action of various cytokines. Some of these cytokines (IL-1, IL-6, IL-17, and TNFα) are found early following virus infection [38, 5355] and some (IL-1 and TNFα) found shortly after reactivation [56]. Staats and Lausch detected increased levels for proinflammatory cytokines, IL-1α, and IL-6 following primary HSV-1 infection [53]. In addition, targeting IL-1 during primary HSV-1 significantly reduced disease indicating its central role following acute infection [54]. During recurrent HSK our laboratory also detected significant increases in both IL-1, and TNFα [56]. Furthermore, when these cytokines were neutralized in vivo, they were shown to be required for the development of recurrent HSK [56].

Following this proinflammatory period, a somewhat overlapping set of cytokines (IFN-γ, IL-12, IL-17, IL-4 and IL-10) are found later during primary HSK [38, 53, 54, 57, 58], while during this same time period recurrent HSK the cytokines IFN-γ, IL-12, IL-4, and IL-10 are reported to be expressed [5961]. These cytokines can be very informative as they will be indicative of the phenotype of the T cell that is orchestrating the response at that time point. The profile of cytokines made by antigen-specific T cells in an immune response indicates whether it is mediated by Th1, Th2, or Th17 T cells. A Th1 response is characterized by the production of IL-2, IL-12, and IFN-γ [62], Th2 cells produce IL-4, IL-5, IL-6, and IL-10 [62], while Th17 cells produce IL-17A, IL-17F, and IL-22 [63]. It has been reported that significant levels of IFN-γ, IL-2, and TNF-α/β, but not IL-4 or IL-10, are found in mouse corneas with primary HSK, suggesting that CD4+ T cells in the eye are Th1 [31]. Similar conclusions were derived from studies demonstrating that treatment with anti-IFN-γ and anti-IL-2 antibodies, but not with anti-IL-4 antibodies, protected mice from primary HSK [64]. It has been reported that IL-2 not only stimulates Th1 development but also is chemotactic for and maintains the viability of polymorphonuclear neutrophils [65], which are an early component of primary HSK [34, 35]. Further evidence supporting Th1 cell involvement comes from studies using STAT4 KO mice [66]. STAT4 is a transcription factor that is activated by the presence of IL-12 and directs the polarization of T cells to become Th1 cells [67, 68]. When infected with HSV-1, these STAT4 KO mice did not develop significant primary HSK lesions, though they were more susceptible to developing encephalitis [66]. In contrast to these results, it has been shown that when IFN-γ KO mice were used to confirm the role of this cytokine during primary HSK surprisingly displayed similar disease to that seen in wild-type mice [67]. This was surprising as one would have expected that such mice would display less corneal disease as IFN-γ is an integral cytokine associated with Th1 responses [62, 68]. Interestingly, a very similar phenotype was observed in mice undergoing recurrent HSK [69]. In these studies both IFN-γ KO mice and wild-type mice treated with anti-IFN-γ were subjected to recurrent HSK and the disease was indistinguishable from recurrent HSK in control antibody-treated wild-type mice [69].

In a completely different set of studies to determine the role of IL-10 during HSK, it was reported that intraocular treatment of mice with IL-10 reduced corneal disease from 95% to 36% [43, 70]. This treatment, while not effecting the production of IL-1α in the cornea, led to a 10-fold reduction in IL-2 and a 50-fold reduction in IL-6 [70]. It has also been reported that IL-10 DNA administration during primary HSK significantly reduced disease [71]. Our laboratory also evaluated the role that IL-10 plays during recurrent HSK, and similar to what was reported during primary HSK [43, 70], lack of IL-10, as determined by neutralizing IL-10 or using IL-10 KO mice, resulted in very severe corneal disease [72]. Furthermore, treating mice with recombinant IL-10 led to significantly reduced corneal disease [72]. Taken together, these studies, both in primary HSK and recurrent HSK, demonstrate that IL-10 is associated with amelioration of disease [72].

Within the past several years, a new T-cell player has been added to the mix, namely, Th17 cells. These cells were termed Th17 because of their production of the cytokine IL-17 [63]. They have been shown to be involved in autoimmunity and host defense [63]. It was first reported that mice lacking IL-17 receptor displayed reduced neutrophil infiltrate and less corneal disease [73]. This observation was followed by a report that showed that treatment with Resolvin E1 reduced clinical disease and at the same time reduced the numbers of both Th1 and Th17 cells in the cornea [74], further indicating that Th17 cells might be playing a role in primary HSK. That role was further supported when IL-17 KO mice and wild type mice treated with neutralizing antibodies were infected with HSV-1. These studies demonstrated that primary HSK was significantly inhibited thus indicating that both Th1 and Th17 cells likely act in concert to produce the corneal lesions seen during primary HSK [54]. The role that Th17 cells, and thus the cytokine IL-17, play in recurrent HSK has yet to be evaluated.

5. T-Cell Functions during HSK

As the aforementioned research implies, T cells are critical to the development of corneal lesions during both primary and recurrent HSK. In fact, T-cell deficient mice do experience HSK [28], unless T cells are adoptively transferred [29]. During primary HSK, the preponderance of data suggests that CD4+ T cells of the Th1 subset are the likely mediators of disease [30, 31, 64, 70, 71] while CD8+ T cells play a minor role in disease [22, 30, 31, 33, 75] or possibly are involved in reducing disease [15]. Studies attempting to determine if the classical CD4+ FoxP3+ Treg cells are involved in reducing corneal disease have not conclusively demonstrated that they do so in vivo [4]. Another means of determining the functional role of T cells is the measurement of DTH responses, which are most often mediated by the Th1 subset of CD4+ T cells. Using this readout, decreased disease is most often associated with reduced DTH responses [76, 77], though in one case where both Th1 and Th2 responses were allowed to develop, HSK was reduced, but DTH was unaltered [78]. During recurrent HSK, the T-cell profile is somewhat different. Whether by targeting these T-cell subsets by antibody depletion or by using CD4KO and CD8KO mice, these mice expressed a decreased HSK disease phenotype as compared to wild-type and untreated mice [79]. Thus it would appear that both CD4+ and CD8+ T cells contribute to clinical disease during recurrent HSK [79]. In addition, when the cytokine pattern was investigated, both Th1 and Th2 cytokines were present in the cornea shortly following reactivation [60]. These results should not be surprising as it should be recalled that recurrent HSK occurs in mice that have developed an immune response against HSV-1. That immune response will include antigen-specific CD4+ T cells of both Th1 and Th2 subsets as well as HSV-1-specific CD8+ T cells. Thus the mixed T-cell infiltrate may, at least in part, explain why focal stromal opacities, rather than the diffuse opacity, as typically seen in primary HSK, characterize recurrent HSK in mice [8]. That said, while there is a report implicating cytotoxic T cells in primary disease [80], recurrent disease is most often associated with strong DTH responses [72, 81].

Since T cells clearly play a central role in mediating disease, identifying those factors that are involved in their activation is very important. A critical factor for activation of T cells is the engagement of costimulatory molecules. The best characterized of these costimulatory interactions are between CD28 or CTLA-4 on the T cell and B7-1 or B7-2 expressed by the APC [82]. Studies have shown that blocking the CD28 interaction with CTLA-4Ig significantly reduced primary HSK by inhibiting Th1 but not Th2 cells [83]. When only B7-2 was blocked, there was a significant reduction in CD8+ cytotoxic T-cell response and these mice experienced delayed viral clearance at the cornea [84]. However, these treated mice did not display an impact on corneal disease, suggesting that naïve CD4+ T cells are being costimulated via B7-1 [84]. The Hendricks laboratory demonstrated that targeting the CD40-CD154 interaction changed the cellular nature of the inflammatory response but did not result in reduced primary HSK [85], while targeting the OX40-OX40L interaction did not significantly alter primary HSK in any way [86]. When the CD137-CD137L costimulatory pathway was targeted, reduced primary HSK was observed [87]. However, it is interesting to note that this same group later demonstrated that this interaction led to increased CD8+ T-cell responses that better controlled HSV-1 infection of the skin [88]. We are currently addressing the role that costimulation plays in recurrent HSK. We hypothesize that since recurrent HSK occurs in an animal that possesses an anti-HSV-1 immune response, we believe that CD28-mediated costimulation will primarily be associated with the initial T-cell activation that occurs shortly after infection and likely is not involved in subsequent stimulation of preexisting anti-HSV-1 T cells that will be present when reactivation of virus from latency occurs. However, it is possible that other costimulatory interactions that occur subsequent to CD28 activation might be important in recurrent HSK. It will be those interactions (CD40-CD154 and CD137-CD137L) that are the focus of ongoing studies in our laboratory.

6. Role of T Cells in Maintaining Viral Latency

In addition to their role in mediating corneal disease, T cells have also been implicated in maintaining viral latency in the infected trigeminal ganglia (TG). This role for T cells comes primarily from work done in the laboratory of Robert Hendricks, who reported that CD8+ T cells surround latently infected TG neurons in mice infected with HSV-1 [8992]. In a similar fashion, clinical studies performed on recently deceased individual who know to have infections with HSV-1, found both CD8+ and CD4+ T cells surrounding neurons that are also stained for HSV-1 antigens [93, 94]. These cells expressed surface markers indicating that they were late effector memory T cells. As was seen in mice, they expressed granzyme markers but no cytolytic function was detected [94]. It has since been reported in murine studies that the production of IFNγ, granzyme, and perforin by T cells is important to this maintenance of latency [95, 96]. It was further shown that these infected neurons were not undergoing apoptosis, but that the target of granzyme B released by T cells was the HSV-1 immediate early protein, ICP4 [95], which is essential for further viral gene expression and thus production of infectious virus [97]. More recently the TCR usage of these T cells found in latently infected C57BL/6 TG was characterized [98]. It was demonstrated that the majority were specific for a glycoprotein B peptide, but that CD8+ T cells specific for at least 18 other subdominant determinants could be found [98]. One caveat to these studies is that mice lacking CD8+ T cells display a similar latency phenotype as normal mice [14]. None the less, taken together, these observations indicate that CD8+ T cells are likely involved in maintaining latency but that other factors are also involved.

7. Vaccination in HSK

Many attempts have been made to develop a vaccine that is effective in preventing HSK. While most vaccines prevent primary HSK in animal models when given prior to infection [99103], they typically fail when delivered therapeutically to prevent recurrent HSK [9, 104]. Nesburn et al. reported that periocular vaccination of latently infected rabbits with recombinant HSV-2 gB/gD in MTP-PE adjuvant resulted in a 2-3-fold decrease in spontaneous corneal viral shedding but had no effect on corneal scarring [105]. They also reported that periocular vaccination with gD1 or gD2 inhibited recurrent dendritic keratitis [106]. Likewise, we and others have shown that vaccination with certain HSV-1 components limits both viral pathogenesis and prevents primary and recurrent corneal disease only when administered prior to infection [9, 104]. If administered following infection these vaccines are ineffective in preventing recurrent disease [9]. These studies illustrate the difficulty of developing effective vaccines for both primary and recurrent infections of HSV. The development of such a vaccine would be of great clinical significance since most patients who present with HSK are latently infected [1, 6, 7]. This was illustrated by a clinical trial in which the vaccine was only effective in women who had never been infected with either HSV-1 or HSV-2 and ineffective in those who had a history of infections with either HSV-1 or HSV-2 [107]. That said, we reported that vaccination with a vhs-defective mutant of HSV-1 [108], which had previously been shown to significantly reduce the rate of reactivation [103, 104], would also reduce recurrent HSK when constructed with [109] or without ICP8 [81]. The important thing about these studies was that the vaccine was administered intraperitoneally after infection and thus would have therapeutic value for latently infected individuals. It should be noted that another vaccine construct that consisted of defective vhs and ICP8 to which was cloned B7-1 or B7-2 was also effective prophylactically [110] and might also show efficacy when used therapeutically since it is very similar to previous vaccine constructs [81, 109]. Similar results were also seen in studies by Richards et al. who nasally vaccinated latently infected mice with a mixture of HSV-1 glycoproteins and recombinant E. coli heat-labile enterotoxin B subunit as an adjuvant [111].

The mechanism responsible for the therapeutic success of these vaccine constructs has not been fully defined. However, the mechanism could be similar to what was reported in studies involving Theiler's virus- (TMEV-) mediated demyelinating disease in mice. This disease, which is associated with a Th1 antiviral response, was shown to be significantly reduced in infected mice following vaccination with TMEV-coupled spleen cells [112, 113]. These investigators went on to demonstrate that protection is accompanied by a preferential reduction in the Th1 antiviral response and a concomitant increase in the Th2-mediated antiviral response [113]. Thus it appears that protection involves alteration of a Th1-mediated immune response to one primarily mediated by Th2 cells. Results from our vaccination studies using a vhs defective replication incompetent virus show similar changes as evidenced by lower DTH responses and increased anti-HSV-1 neutralizing antibody titers in vaccinated mice as compared to mice receiving a control vaccine [81]. Ocular HSV-1 infection typically stimulates an immune response that protects the animal from lethal disease by this virus but also leads to a significant inflammatory response in the cornea that can result in corneal damage. A similar type of immune response also protects latently infected animals that are reactivated from lethal viral disease but can result in significant corneal disease. The mechanism underlying this alteration in the immune response might involve the activation of T regulatory cells. Both conventional CD4+ T cells [114, 115] and CD8+ T cells [15] have been implicated in regulating the anti-HSV-1 response during primary HSK. However, when Devito and Hendricks tested this, they did not observe an association for T regulatory cells within the cornea and decreased corneal lesions [4]. Thus the case for T regulatory involvement in reducing or resolving primary HSK remains to be established.

8. Concluding Remarks

Ocular disease associated with HSV-1 infection (HSK) is the leading cause of infectious blindness and is clearly the result of an immune-mediated inflammatory attack of the cornea. We present data comparing primary and recurrent murine models that are used to study this disease. As should be evident from this discussion, these two model systems, while sharing many characteristics, are not the same. We have learned a lot about this disease from studies of primary HSK. These studies have identified many factors that are responsible for the corneal damage associated with this disease. In fact, these studies have also suggested many potential therapeutic means of treating primary HSK. However, we would argue that without testing these therapies in a recurrent model of HSK, we might be chasing things that ultimately will not prove useful in the clinic. Consequently, due to the paucity of studies investigating recurrent HSK, much research remains to be performed. Particularly concerning those aspects of primary HSK that are reflected in recurrent HSK and also those things that are not, we contend that a better understanding of the immunological factors, both cellular and cell-free, that underlie the development of recurrent HSK will enable us to identify potential therapies that might prove to be most effective in treating human HSK.

Acknowledgments

This work was supported by the National Institutes of Health Grants EY11885, EY16352, and EY21247 and an unrestricted grant from Research to Prevent Blindness to the Department of Ophthalmology.

References

  • 1.Pepose JS, Leib DA, Stuart PM, Easty EL. Herpes simplex virus diseases: anterior segment of the eye. In: Pepose JS, Holland GAN, Wilhelmus KR, editors. Ocular Infection and Immunity. St. Louis, Mo, USA: Mosby; 1996. pp. 905–932. [Google Scholar]
  • 2.Thomas J, Gangappa S, Kanangat S, Rouse BT. On the essential involvement of neutrophils in the immunopathologic disease: herpetic stromal keratitis. Journal of Immunology. 1997;158(3):1383–1391. [PubMed] [Google Scholar]
  • 3.Maertzdorf J, Verjans GM, Remeijer L, van der Kooi A, Osterhaus AD. Restricted T cell receptor beta-chain variable region protein use by cornea-derived CD4+ and CD8+ herpes simplex virus-specific T cells in patients with herpetic stromal keratitis. Journal of Infectious Diseases. 2003;187(4):550–558. doi: 10.1086/367991. [DOI] [PubMed] [Google Scholar]
  • 4.Divito SJ, Hendricks RL. Activated inflammatory infiltrate in HSV-1-infected corneas without herpes stromal keratitis. Investigative Ophthalmology and Visual Science. 2008;49(4):1488–1495. doi: 10.1167/iovs.07-1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rinne JR, Abghari SZ, Stulting RD. The severity of herpes simplex viral keratitis in mice does not reflect the severity of disease in humans. Investigative Ophthalmology and Visual Science. 1992;33(2):268–272. [PubMed] [Google Scholar]
  • 6.Liesegang TJ. Herpes simplex virus epidemiology and ocular importance. Cornea. 2001;20(1):1–13. doi: 10.1097/00003226-200101000-00001. [DOI] [PubMed] [Google Scholar]
  • 7.Wilhelmus KR, Coster DJ, Donovan HC. Prognostic indicators of herpetic keratitis. Analysis of a five-year observation period after corneal ulceration. Archives of Ophthalmology. 1981;99(9):1578–1582. doi: 10.1001/archopht.1981.03930020452009. [DOI] [PubMed] [Google Scholar]
  • 8.Miller JK, Laycock KA, Umphress JA, Hook KK, Stuart PM, Pepose JS. A comparison of recurrent and primary herpes simplex keratitis in NIH inbred mice. Cornea. 1996;15(5):497–504. [PubMed] [Google Scholar]
  • 9.Keadle TL, Laycock KA, Miller JK, et al. Efficacy of a recombinant glycoprotein D subunit vaccine on the development of primary and recurrent ocular infection with herpes simplex virus type 1 in mice. Journal of Infectious Diseases. 1997;176(2):331–338. doi: 10.1086/514049. [DOI] [PubMed] [Google Scholar]
  • 10.Cook SD, Paveloff MJ, Doucet JJ, Cottingham AJ, Sedarati F, Hill JM. Ocular herpes simplex virus reactivation in mice latently infected with latency-associated transcript mutants. Investigative Ophthalmology and Visual Science. 1991;32(5):1558–1561. [PubMed] [Google Scholar]
  • 11.Shimeld C, Hill T, Blyth B, Easty D. An improved model of recurrent herpetic eye disease in mice. Current Eye Research. 1989;8(11):1193–1205. doi: 10.3109/02713688909000044. [DOI] [PubMed] [Google Scholar]
  • 12.Shimeld C, Hill TJ, Blyth WA, Easty DL. Reactivation of latent infection and induction of recurrent herpetic eye disease in mice. Journal of General Virology. 1990;71(2):397–404. doi: 10.1099/0022-1317-71-2-397. [DOI] [PubMed] [Google Scholar]
  • 13.Shimeld C, Hill TJ, Blyth WA, Easty DL. Passive immunization protects the mouse eye from damage after herpes simplex virus infection by limiting spread of virus in the nervous system. Journal of General Virology. 1990;71(3):681–687. doi: 10.1099/0022-1317-71-3-681. [DOI] [PubMed] [Google Scholar]
  • 14.Laycock KA, Lee SF, Brady RH, Pepose JS. Characterization of a murine model of recurrent herpes simplex viral keratitis induced by ultraviolet B radiation. Investigative Ophthalmology and Visual Science. 1991;32(10):2741–2746. [PubMed] [Google Scholar]
  • 15.Stuart PM, Summers B, Morris JE, Morrison LA, Leib DA. CD8+ T cells control corneal disease following ocular infection with herpes simplex virus type 1. Journal of General Virology. 2004;85(7):2055–2063. doi: 10.1099/vir.0.80049-0. [DOI] [PubMed] [Google Scholar]
  • 16.Morris JE, Zobell S, Yin XT, et al. Mice with mutations in fas and fas ligand demonstrate increased herpetic stromal keratitis following corneal infection with HSV-1. Journal of Immunology. 2012;188(2):793–799. doi: 10.4049/jimmunol.1102251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sawtell NM, Thompson RL. Rapid in vivo reactivation of herpes simplex virus in latently infected murine ganglionic neurons after transient hyperthermia. Journal of Virology. 1992;66(4):2150–2156. doi: 10.1128/jvi.66.4.2150-2156.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sawtell NM. Quantitative analysis of herpes simplex virus reactivation in vivo demonstrates that reactivation in the nervous system is not inhibited at early times postinoculation. Journal of Virology. 2003;77(7):4127–4138. doi: 10.1128/JVI.77.7.4127-4138.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Neumann DM, Bhattacharjee PS, Hill JM. Sodium butyrate: a chemical inducer of in vivo reactivation of herpes simplex virus type 1 in the ocular mouse model. Journal of Virology. 2007;81(11):6106–6110. doi: 10.1128/JVI.00070-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Newell CK, Martin S, Sendele D, Mercadal CM, Rouse BT. Herpes simplex virus-induced stromal keratitis: role of T-lymphocyte subsets in immunopathology. Journal of Virology. 1989;63(2):769–775. doi: 10.1128/jvi.63.2.769-775.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Su YH, Yan XT, Oakes JE, Lausch RN. Protective antibody therapy is associated with reduced chemokine transcripts in herpes simplex virus type 1 corneal infection. Journal of Virology. 1996;70(2):1277–1281. doi: 10.1128/jvi.70.2.1277-1281.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ghiasi H, Cai S, Perng GC, Nesburn AB, Wechsler SL. Both CD4+ and CD8+ T cells are involved in protection against HSV-1 induced corneal scarring. British Journal of Ophthalmology. 2000;84(4):408–412. doi: 10.1136/bjo.84.4.408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kintner RL, Brandt CR. The effect of viral inoculum level and host age on disease incidence, disease severity, and mortality in a murine model of ocular HSV-1 infection. Current Eye Research. 1995;14(2):145–152. doi: 10.3109/02713689508999926. [DOI] [PubMed] [Google Scholar]
  • 24.Bhattacharjee PS, Neumann DM, Foster TP, et al. Effective treatment of ocular HSK with a human apolipoprotein E mimetic peptide in a mouse eye model. Investigative Ophthalmology and Visual Science. 2008;49(10):4263–4268. doi: 10.1167/iovs.08-2077. [DOI] [PubMed] [Google Scholar]
  • 25.Strieter RM, Standiford TJ, Huffnagle GB, Colletti LM, Lukacs NW, Kunkel SL. “The good, the bad, and the ugly”: the role of chemokines in models of human disease. Journal of Immunology. 1996;156(10):3583–3586. [PubMed] [Google Scholar]
  • 26.Taub DD, Conlon K, Lloyd AR, Oppenheim JJ, Kelvin DJ. Preferential migration of activated CD4+ and CD8+ T cells in response to MIP-1α and MIP-1β . Science. 1993;260(5106):355–358. doi: 10.1126/science.7682337. [DOI] [PubMed] [Google Scholar]
  • 27.Tamesis RR, Messmer EM, Rice BA, Dutt JE, Foster CS. The role of natural killer cells in the development of herpes simplex virus type 1 induced stromal keratitis in mice. Eye. 1994;8(3):298–306. doi: 10.1038/eye.1994.61. [DOI] [PubMed] [Google Scholar]
  • 28.Metcalf JF, Hamilton DS, Reichert RW. Herpetic keratitis in athymic (nude) mice. Infection and Immunity. 1979;26(3):1164–1171. doi: 10.1128/iai.26.3.1164-1171.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Russell RG, Nasisse MP, Larsen HS, Rouse BT. Role of T-lymphocytes in the pathogenesis of herpetic stromal keratitis. Investigative Ophthalmology and Visual Science. 1984;25(8):938–944. [PubMed] [Google Scholar]
  • 30.Doymaz MZ, Rouse BT. Herpetic stromal keratitis: an immunopathologic disease mediated by CD4+ T lymphocytes. Investigative Ophthalmology and Visual Science. 1992;33(7):2165–2173. [PubMed] [Google Scholar]
  • 31.Niemialtowski MG, Rouse BT. Predominance of Th1 cells in ocular tissues during herpetic stromal keratitis. Journal of Immunology. 1992;149(9):3035–3039. [PubMed] [Google Scholar]
  • 32.Hendricks RL, Tao MSP, Glorioso JC. Alterations in the antigenic structure of two major HSV-1 glycoproteins, gC and gB, influence immune regulation and susceptibility to murine herpes keratitis. Journal of Immunology. 1989;142(1):263–269. [PubMed] [Google Scholar]
  • 33.Hendricks RL, Tumpey TM. Contribution of virus and immune factors to herpes simplex virus type I-induced corneal pathology. Investigative Ophthalmology and Visual Science. 1990;31(10):1929–1939. [PubMed] [Google Scholar]
  • 34.Tumpey TM, Chen SH, Oakes JE, Lausch RN. Neutrophil-mediated suppression of virus replication after herpes simplex virus type 1 infection of the murine cornea. Journal of Virology. 1996;70(2):898–904. doi: 10.1128/jvi.70.2.898-904.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Thomas J, Gangappa S, Kanangat S, Rouse BT. On the essential involvement of neutrophils in the immunopathologic disease: herpetic stromal keratitis. Journal of Immunology. 1997;158(3):1383–1391. [PubMed] [Google Scholar]
  • 36.Miller JK, Laycock KA, Nash MM, Pepose JS. Corneal Langerhans cell dynamics after herpes simplex virus reactivation. Investigative Ophthalmology and Visual Science. 1993;34(7):2282–2290. [PubMed] [Google Scholar]
  • 37.Cook DN, Beck MA, Coffman TM, et al. Requirement of MIP-1α for an inflammatory response to viral infection. Science. 1995;269(5230):1583–1585. doi: 10.1126/science.7667639. [DOI] [PubMed] [Google Scholar]
  • 38.Tumpey TM, Cheng H, Cook DN, Smithies O, Oakes JE, Lausch RN. Absence of macrophage inflammatory protein-1α prevents the development of blinding herpes stromal keratitis. Agricultural and Forest Meteorology. 1999;97(5):275–292. doi: 10.1128/jvi.72.5.3705-3710.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yan XT, Tumpey TM, Kunkel SL, Oakes JE, Lausch RN. Role of MBP-2 in neutrophil migration and tissue injury in the herpes simplex virus-1-infected cornea. Investigative Ophthalmology and Visual Science. 1998;39(10):1854–1862. [PubMed] [Google Scholar]
  • 40.Stuart PM, Morris JE, Sidhu M, Keadle TL. CCL3 protects mice from corneal pathology during recurrent HSV-1 infection. Frontiers in Bioscience. 2008;13(12):4407–4415. doi: 10.2741/3013. [DOI] [PubMed] [Google Scholar]
  • 41.Colantonio L, Recalde H, Sinigaglia F, D’Ambrosio D. Modulation of chemokine receptor expression and chemotactic responsiveness during differentiation of human naive T cells into Th1 or Th2 cells. European Journal of Immunology. 2002;32(5):1264–1273. doi: 10.1002/1521-4141(200205)32:5<1264::AID-IMMU1264>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
  • 42.Fenton RR, Molesworth-Kenyon S, Oakes JE, Lausch RN. Linkage of IL-6 with neutrophil chemoattractant expression in virus-induced ocular inflammation. Investigative Ophthalmology and Visual Science. 2002;43(3):737–743. [PubMed] [Google Scholar]
  • 43.Tumpey TM, Cheng H, Yan XT, Oakes JE, Lausch RN. Chemokine synthesis in the HSV-1-infected cornea and its suppression by interleukin-10. Journal of Leukocyte Biology. 1998;63(4):486–492. doi: 10.1002/jlb.63.4.486. [DOI] [PubMed] [Google Scholar]
  • 44.Kim B, Sarangi PP, Lee Y, Kaistha SD, Lee S, Rouse BT. Depletion of MCP-1 increases development of herpetic stromal keratitis by innate immune modulation. Journal of Leukocyte Biology. 2006;80(6):1405–1415. doi: 10.1189/jlb.0406295. [DOI] [PubMed] [Google Scholar]
  • 45.Eo SK, Lee S, Chun S, Rouse BT. Modulation of immunity against herpes simplex virus infection via mucosal genetic transfer of plasmid DNA encoding chemokines. Journal of Virology. 2001;75(2):569–578. doi: 10.1128/JVI.75.2.569-578.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lee SK, Choi BK, Kang WJ, et al. MCP-1 derived from stromal keratocyte induces corneal infiltration of CD4+ T cells in herpetic stromal keratitis. Molecules and Cells. 2008;26(1):67–73. [PubMed] [Google Scholar]
  • 47.Duan R, Remeijer L, van Dun JM, Osterhaus AD, Verjans GM. Granulocyte macrophage colony-stimulating factor expression in human herpetic stromal keratitis: Implications for the role of neutrophils in HSK. Investigative Ophthalmology and Visual Science. 2007;48(1):277–284. doi: 10.1167/iovs.06-0053. [DOI] [PubMed] [Google Scholar]
  • 48.Mikloska Z, Danis VA, Adams S, Lloyd AR, Adrian DL, Cunningham AL. In vivo production of cytokines and β (C-C) chemokines in human recurrent herpes simplex lesions—do herpes simplex virus—infected keratinocytes contribute to their production? Journal of Infectious Diseases. 1998;177(4):827–838. doi: 10.1086/515236. [DOI] [PubMed] [Google Scholar]
  • 49.Li H, Zhang J, Kumar A, Zheng M, Atherton SS, Yu FS. Herpes simplex virus 1 infection induces the expression of proinflammatory cytokines, interferons and TLR7 in human corneal epithelial cells. Immunology. 2006;117(2):167–176. doi: 10.1111/j.1365-2567.2005.02275.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD. IFN-γ-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. Journal of Immunology. 2002;168(7):3195–3204. doi: 10.4049/jimmunol.168.7.3195. [DOI] [PubMed] [Google Scholar]
  • 51.Lee S, Zheng M, Deshpande S, Eo SK, Hamilton TA, Rouse BT. IL-12 suppresses the expression of ocular immunoinflammatory lesions by effects on angiogenesis. Journal of Leukocyte Biology. 2002;71(3):469–476. [PubMed] [Google Scholar]
  • 52.Carr DJ, Chodosh J, Ash J, Lane TE. Effect of anti-CXCL10 monoclonal antibody on herpes simplex virus type 1 keratitis and retinal infection. Journal of Virology. 2003;77(18):10037–10046. doi: 10.1128/JVI.77.18.10037-10046.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Staats HF, Lausch RN. Cytokine expression in vivo during murine herpetic stromal keratitis: effect of protective antibody therapy. Journal of Immunology. 1993;151(1):277–283. [PubMed] [Google Scholar]
  • 54.Biswas PS, Banerjee K, Kim B, Rouse BT. Mice transgenic for IL-1 receptor antagonist protein are resistant to herpetic stromal keratitis: possible role for IL-1 in herpetic stromal keratitis pathogenesis. Journal of Immunology. 2004;172(6):3736–3744. doi: 10.4049/jimmunol.172.6.3736. [DOI] [PubMed] [Google Scholar]
  • 55.Suryawanshi A, Veiga-Parga T, Rajasagi NK, et al. Role of il-17 and th17 cells in herpes simplex virus-induced corneal immunopathology. Journal of Immunology. 2011;187(4):1919–1930. doi: 10.4049/jimmunol.1100736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Keadle TL, Laycock KA, Pepose JS, Stuart PM. Proinflammatory cytokines IL-1 and TNF-α are required for recurrent herpetic keratitis in NIH mice. Investigative Ophthalmology & Visual Science. 2000;41:96–102. [PubMed] [Google Scholar]
  • 57.Babu JS, Kanangat S, Rouse BT. T cell cytokine mRNA expression during the course of the immunopathologic ocular disease herpetic stromal keratitis. Journal of Immunology. 1995;154(9):4822–4829. [PubMed] [Google Scholar]
  • 58.Kanangat S, Thomas J, Gangappa S, Babu JS, Rouse BT. Herpes simplex virus type 1-mediated up-regulation of IL-12 (p40) mRNA expression: implications in immunopathogenesis and protection. Journal of Immunology. 1996;156(3):1110–1116. [PubMed] [Google Scholar]
  • 59.Shimeld C, Easty DL, Hill TJ. Reactivation of herpes simplex virus type 1 in the mouse trigeminal ganglion: an in vivo study of virus antigen and cytokines. Journal of Virology. 1999;73(3):1767–1773. doi: 10.1128/jvi.73.3.1767-1773.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Keadle TL, Usui N, Laycock KA, Kumano Y, Pepose JS, Stuart PM. Cytokine expression in murine corneas during recurrent herpetic stromal keratitis. Ocular Immunology and Inflammation. 2001;9(3):193–205. doi: 10.1076/ocii.9.3.193.3967. [DOI] [PubMed] [Google Scholar]
  • 61.Stumpf TH, Shimeld C, Easty DL, Hill TJ. Cytokine production in a murine model of recurrent herpetic stromal keratitis. Investigative Ophthalmology and Visual Science. 2001;42(2):372–378. [PubMed] [Google Scholar]
  • 62.Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annual Review of Immunology. 1989;7:145–173. doi: 10.1146/annurev.iy.07.040189.001045. [DOI] [PubMed] [Google Scholar]
  • 63.Graeber KE, Olsen NJ. Th17 cell cytokine secretion profile in host defense and autoimmunity. Inflammation Research. 2012;61(2):87–96. doi: 10.1007/s00011-011-0419-1. [DOI] [PubMed] [Google Scholar]
  • 64.Hendricks RL, Tumpey TM, Finnegan A. IFN-γ and IL-2 are protective in the skin but pathologic in the corneas of HSV-1-infected mice. Journal of Immunology. 1992;149(9):3023–3028. [PubMed] [Google Scholar]
  • 65.Tang Q, Chen W, Hendricks RL. Proinflammatory functions of IL-2 in herpes simplex virus corneal infection. Journal of Immunology. 1997;158(3):1275–1283. [PubMed] [Google Scholar]
  • 66.Banerjee K, Biswas PS, Rouse BT. Role of Stat4-mediated signal transduction events in the generation of aggressor CD4+ T cells in herpetic stromal keratitis pathogenesis. Journal of Interferon and Cytokine Research. 2007;27(1):65–75. doi: 10.1089/jir.2007.0077. [DOI] [PubMed] [Google Scholar]
  • 67.Bouley DM, Kanangat S, Wire W, Rouse BT. Characterization of herpes simplex virus type-1 infection and herpetic stromal keratitis development in IFN-γ knockout mice. Journal of Immunology. 1995;155(8):3964–3971. [PubMed] [Google Scholar]
  • 68.Jacobson NG, Szabo SJ, Weber-Nordt RM, et al. Interleukin 12 signaling in T helper type 1 (Th1) cells involves tyrosine phosphorylation of signal transducer and activator of transcription (Stat)3 and Stat4. Journal of Experimental Medicine. 1995;181(5):1755–1762. doi: 10.1084/jem.181.5.1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Keadle TL, Alexander DE, Leib DA, Stuart PM. Interferon gamma is not required for recurrent herpetic stromal keratitis. Virology. 2008;380(1):46–51. doi: 10.1016/j.virol.2008.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Tumpey TM, Elner VM, Chen SH, Oakes JE, Lausch RN. Interleukin-10 treatment can suppress stromal keratitis induced by herpes simplex virus type 1. Journal of Immunology. 1994;153(5):2258–2265. [PubMed] [Google Scholar]
  • 71.Daheshia M, Kuklin N, Kanangat S, Manickan E, Rouse BT. Suppression of ongoing ocular inflammatory disease by topical administration of plasmid DNA encoding IL-10. Journal of Immunology. 1997;159(4):1945–1952. [PubMed] [Google Scholar]
  • 72.Keadle TL, Stuart PM. Interleukin-10 (IL-10) ameliorates corneal disease in a mouse model of recurrent herpetic keratitis. Microbial Pathogenesis. 2005;38(1):13–21. doi: 10.1016/j.micpath.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 73.Molesworth-Kenyon SJ, Yin R, Oakes JE, Lausch RN. IL-17 receptor signaling influences virus-induced corneal inflammation. Journal of Leukocyte Biology. 2008;83(2):401–408. doi: 10.1189/jlb.0807571. [DOI] [PubMed] [Google Scholar]
  • 74.Rajasagi NK, Reddy PB, Suryawanshi A, Mulik S, Gjorstrup P, Rouse BT. Controlling herpes simplex virus-induced ocular inflammatory lesions with the lipid-derived mediator resolvin E1. Journal of Immunology. 2011;186(3):1735–1746. doi: 10.4049/jimmunol.1003456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lepisto AJ, Frank GM, Xu M, Stuart PM, Hendricks RL. CD8 T cells mediate transient herpes stromal keratitis in CD4-deficient mice. Investigative Ophthalmology and Visual Science. 2006;47(8):3400–3409. doi: 10.1167/iovs.05-0898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bauer D, Mrzyk S, Van Rooijen N, Steuhl KP, Heiligenhaus A. Incidence and severity of herpetic stromal keratitis: impaired by the depletion of lymph node macrophages. Experimental Eye Research. 2001;72(3):261–269. doi: 10.1006/exer.2000.0947. [DOI] [PubMed] [Google Scholar]
  • 77.Xia L, Zhang S, Zhou J, Li Y. A crucial role for B and T lymphocyte attenuator in preventing the development of CD4+ T cell-mediated herpetic stromal keratitis. Molecular Vision. 2010;16:2071–2083. [PMC free article] [PubMed] [Google Scholar]
  • 78.Heiligenhaus A, Li H, Schmitz A, Wasmuth S, Bauer D. Improvement of herpetic stromal keratitis with fumaric acid derivate is associated with systemic induction of T helper 2 cytokines. Clinical and Experimental Immunology. 2005;142(1):180–187. doi: 10.1111/j.1365-2249.2005.02896.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Keadle TL, Morris JL, Pepose JS, Stuart PM. CD4+ and CD8+ cells are key participants in the development of recurrent herpetic stromal keratitis in mice. Microbial Pathogenesis. 2002;32(6):255–262. doi: 10.1006/mpat.2002.0506. [DOI] [PubMed] [Google Scholar]
  • 80.Chang E, Galle L, Maggs D, Estes DM, Mitchell WJ. Pathogenesis of herpes simplex virus type 1-induced corneal inflammation in perforin-deficient mice. Journal of Virology. 2000;74(24):11832–11840. doi: 10.1128/jvi.74.24.11832-11840.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Keadle TL, Morrison LA, Morris JL, Pepose JS, Stuart PM. Therapeutic immunization with a virion host shutoff-defective, replication-incompetent herpes simplex virus type 1 strain limits recurrent herpetic ocular infection. Journal of Virology. 2002;76(8):3615–3625. doi: 10.1128/JVI.76.8.3615-3625.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kearney ER, Walunas TL, Karr RW, et al. Antigen-dependent clonal expansion of a trace population of antigen- specific CD4+ T cells in vivo is dependent on CD28 costimulation and inhibited by CTLA-4. Journal of Immunology. 1995;155(3):1032–1036. [PubMed] [Google Scholar]
  • 83.Gangappa S, Manickan E, Rouse BT. Control of herpetic stromal keratitis using CTLA4Ig fusion protein. Clinical Immunology and Immunopathology. 1998;86(1):88–94. doi: 10.1006/clin.1997.4460. [DOI] [PubMed] [Google Scholar]
  • 84.Osorio Y, Cai S, Ghiasi H. Treatment of mice with anti-CD86 mAb reduces CD8+ T cell-mediated CTL activity and enhances ocular viral replication in HSV-1-infected mice. Ocular Immunology and Inflammation. 2005;13(2-3):159–167. doi: 10.1080/09273940490518775. [DOI] [PubMed] [Google Scholar]
  • 85.Xu M, Lepisto AJ, Hendricks RL. CD154 signaling regulates the Th1 response to herpes simplex virus-1 and inflammation in infected corneas. Journal of Immunology. 2004;173(2):1232–1239. doi: 10.4049/jimmunol.173.2.1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lepisto AJ, Xu M, Yagita H, Weinberg AD, Hendricks RL. Expression and function of the OX40/OX40L costimulatory pair during herpes stromal keratitis. Journal of Leukocyte Biology. 2007;81(3):766–774. doi: 10.1189/jlb.0406293. [DOI] [PubMed] [Google Scholar]
  • 87.Seo SK, Park HY, Choi JH, et al. Blocking 4-1BB/4-1BB ligand interactions prevents herpetic stromal keratitis. Journal of Immunology. 2003;171(2):576–583. doi: 10.4049/jimmunol.171.2.576. [DOI] [PubMed] [Google Scholar]
  • 88.Kim YH, Seo SK, Choi BK, et al. 4-1BB costimulation enhances HSV-1-specific CD8+ T cell responses by the induction of CD11c+CD8+ T cells. Cellular Immunology. 2005;238(2):76–86. doi: 10.1016/j.cellimm.2006.01.004. [DOI] [PubMed] [Google Scholar]
  • 89.Liu T, Tang Q, Hendricks RL. Inflammatory infiltration of the trigeminal ganglion after herpes simplex virus type 1 corneal infection. Journal of Virology. 1996;70(1):264–271. doi: 10.1128/jvi.70.1.264-271.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Liu T, Khanna KM, Carriere BN, Hendricks RL. Gamma interferon can prevent herpes simplex virus type 1 reactivation from latency in sensory neurons. Journal of Virology. 2001;75(22):11178–11184. doi: 10.1128/JVI.75.22.11178-11184.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Khanna K, Bonneau R, Kinchington P, Hendricks RL. Herpes simplex virus-specific memory CD8+ T cells are selectively activated and retained in latently infected sensory ganglia. Immunity. 2003;18(5):593–603. doi: 10.1016/s1074-7613(03)00112-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Decman V, Kinchington PR, Harvey SA, Hendricks RL. Gamma interferon can block herpes simplex virus type 1 reactivation from latency, even in the presence of late gene expression. Journal of Virology. 2005;79(16):10339–10347. doi: 10.1128/JVI.79.16.10339-10347.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hüfner K, Derfuss T, Herberger S, et al. Latency of α-herpes viruses is accompanied by a chronic inflammation in human trigeminal ganglia but not in dorsal root ganglia. Journal of Neuropathology and Experimental Neurology. 2006;65(10):1022–1030. doi: 10.1097/01.jnen.0000235852.92963.bf. [DOI] [PubMed] [Google Scholar]
  • 94.Verjans GM, Hintzen RQ, van Dun JM, et al. Selective retention of herpes simplex virus-specific T cells in latently infected human trigeminal ganglia. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(9):3496–3501. doi: 10.1073/pnas.0610847104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.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(5899):268–271. doi: 10.1126/science.1164164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Frank GM, Lepisto AJ, Freeman ML, Sheridan BS, Cherpes TL, Hendricks RL. Early CD4+ T cell help prevents partial CD8+ T cell exhaustion and promotes maintenance of herpes simplex virus 1 latency. Journal of Immunology. 2010;184(1):277–286. doi: 10.4049/jimmunol.0902373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.DeLuca NA, McCarthy AM, Schaffer PA. Isolation and characterization of deletion mutants of herpes simplex virus type 1 in the gene encoding immediate-early regulatory protein ICP4. Journal of Virology. 1985;56(2):558–570. doi: 10.1128/jvi.56.2.558-570.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.St. Leger AJ, Peters B, Sidney J, Sette A, Hendricks RL. Defining the herpes simplex virus-specific CD8+ T cell repertoire in C57BL/6 mice. Journal of Immunology. 2011;186(7):3927–3933. doi: 10.4049/jimmunol.1003735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ghiasi H, Bahri S, Nesburn AB, Wechsler SL. Protection against herpes simplex virus-induced eye disease after vaccination with seven individually expressed herpes simplex virus 1 glycoproteins. Investigative Ophthalmology and Visual Science. 1995;36(7):1352–1360. [PubMed] [Google Scholar]
  • 100.Heiligenhaus, Wells PA, Foster CS. Immunisation against HSV-1 keratitis with a synthetic gD peptide. Eye. 1995;9(1):89–95. doi: 10.1038/eye.1995.14. [DOI] [PubMed] [Google Scholar]
  • 101.Inoue Y, Ohashi Y, Shimomura Y, et al. Herpes simplex virus glycoprotein D. Protective immunity against murine herpetic keratitis. Investigative Ophthalmology and Visual Science. 1990;31(3):411–418. [PubMed] [Google Scholar]
  • 102.Nesburn AB, Burke RL, Ghiasi H, Slanina S, Wechsler SL. Intramuscular vaccination of rabbits with expressed HSV-2 glycoproteins B+D (gD2+gD2) protects against death following ocular HSV-1 challenge but not from eye disease. Investigative Ophthalmology & Visual Science. 1994;35:1679–1686. [Google Scholar]
  • 103.Walker J, Leib DA. Protection from primary infection and establishment of latency by vaccination with a herpes simplex virus type 1 recombinant deficient in the virion host shutoff (vhs) function. Vaccine. 1998;16(1):1–5. doi: 10.1016/s0264-410x(97)00164-3. [DOI] [PubMed] [Google Scholar]
  • 104.Walker J, Laycock KA, Pepose JS, Leib DA. Postexposure vaccination with a virion host shutoff defective mutant reduces UV-B radiation-induced ocular herpes simplex virus shedding in mice. Vaccine. 1998;16(1):6–8. doi: 10.1016/s0264-410x(97)00177-1. [DOI] [PubMed] [Google Scholar]
  • 105.Nesburn AB, Burke RL, Ghiasi H, Slanina S, Bahri S, Wechsler SL. Vaccine therapy for ocular herpes simplex virus (HSV) infection: periocular vaccination reduces spontaneous ocular HSV type 1 shedding in latently infected rabbits. Journal of Virology. 1994;68(8):5084–5092. doi: 10.1128/jvi.68.8.5084-5092.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Nesburn AB, Burke RL, Gbiasi H, Slanina SM, Wechsler SL. A therapeutic vaccine that reduces recurrent herpes simplex virus type 1 corneal disease. Investigative Ophthalmology and Visual Science. 1998;39(7):1163–1170. [PubMed] [Google Scholar]
  • 107.Stanberry LR, Spruance SL, Cunningham AL, et al. Glycoprotein-D-adjuvant vaccine to prevent genital herpes. The New England Journal of Medicine. 2002;347(21):1652–1661. doi: 10.1056/NEJMoa011915. [DOI] [PubMed] [Google Scholar]
  • 108.Strelow LI, Leib DA. Role of the virion host shutoff (vhs) of herpes simplex virus type 1 in latency and pathogenesis. Journal of Virology. 1995;69(11):6779–6786. doi: 10.1128/jvi.69.11.6779-6786.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Keadle TL, Walker J, Laycock KA, et al. Therapeutic vaccination with vhs- herpes simplex virus (HSV) reduces severity of recurrent herpetic stromal keratitis in mice. Journal of General Virology. 2002;83(10):2361–2365. doi: 10.1099/0022-1317-83-10-2361. [DOI] [PubMed] [Google Scholar]
  • 110.Schrimpf JE, Tu EM, Wang H, Wong YM, Morrison LA. B7 costimulation molecules encoded by replication-defective, vhs-deficient HSV-1 improve vaccine-induced protection against corneal disease. Plos ONE. 2011;6(8) doi: 10.1371/journal.pone.0022772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Richards CM, Case R, Hirst TR, Hill TJ, Williams NA. Protection against recurrent ocular herpes simplex virus type 1 disease after therapeutic vaccination of latently infected mice. Journal of Virology. 2003;77(12):6692–6699. doi: 10.1128/JVI.77.12.6692-6699.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Karpus WJ, Pope JG, Peterson JD, Dal Canto MC, Miller SD. Inhibition of Theiler’s virus-mediated demyelination by peripheral immune tolerance induction. Journal of Immunology. 1995;155(2):947–957. [PubMed] [Google Scholar]
  • 113.Karpus WJ, Peterson JD, Miller SD. Anergy in vivo: down-regulation of antigen-specific CD4+ Th1 but not Th2 cytokine responses. International Immunology. 1994;6(5):721–730. doi: 10.1093/intimm/6.5.721. [DOI] [PubMed] [Google Scholar]
  • 114.Sarangi PP, Sehrawat S, Suvas S, Rouse BT. IL-10 and natural regulatory T cells: two independent anti-inflammatory mechanisms in herpes simplex virus-induced ocular immunopathology. Journal of Immunology. 2008;180(9):6297–6306. doi: 10.4049/jimmunol.180.9.6297. [DOI] [PubMed] [Google Scholar]
  • 115.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. Journal of Immunology. 2008;180(11):7636–7647. doi: 10.4049/jimmunol.180.11.7636. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Developmental Immunology are provided here courtesy of Wiley

RESOURCES