Complement and CD4+ T cells drive context-specific corneal sensory neuropathy

  1. Derek J Royer  Is a corresponding author
  2. Jose Echegaray-Mendez
  3. Liwen Lin
  4. Grzegorz B Gmyrek
  5. Rose Mathew
  6. Daniel R Saban
  7. Victor L Perez
  8. Daniel JJ Carr
  1. Duke University Medical Center, United States
  2. University of Oklahoma Health Sciences Center, United States
8 figures, 3 tables and 1 additional file

Figures

Figure 1 with 1 supplement
Complement C3 contributes to corneal denervation.

(A) Representative confocal images of cornea flat-mounts showing corneal nerves (βIII Tubulin, white) and T cells (CD3, green) in healthy uninfected (UI) and HSV-1-infected corneas 8 days post infection (p.i.). (B) Corneal mechanosensory function in WT and C3-/- mice following ocular HSV-1 infection (n = 6–8 mice/group; three independent experiments). (C) Viral titers shed in the tear film of WT and C3-/- mice at the indicated times p.i. (n = 5 mice per group; two independent experiments). Viral titers in the corneas and trigeminal ganglia (TG) of WT and C3-/- mice are shown at days 3 and 7 p.i. in (D) and (E), respectively. (F) Representative confocal images of stromal nerve fibers and T cells in healthy and HSV-infected corneas of WT and C3-/- mice as in (A). (G) Flow cytometry-based quantification of CD4+ and CD8+ T cells in HSV-infected corneas at day 7 p.i. (n = 7–8 mice/group; four independent experiments). Statistical differences were determined using two-way ANOVAs with Bonferroni posttests (B, C) or Student’s T tests (D, E, G).

https://doi.org/10.7554/eLife.48378.003
Figure 1—figure supplement 1
C3-/-mice retain corneal sensation through viral latency.

WT and C3-/- mice were infected with 1 × 104 p.f.u. HSV-1 McKrae per eye and corneal sensation thresholds measured at day 30 post infection when HSV-1 is considered to be latent in animal infection models (n = 6–12 eyes/group; Student’s T-test).

https://doi.org/10.7554/eLife.48378.004
Figure 2 with 1 supplement
T cells facilitate corneal sensation loss.

(A) Chemokine concentrations in HSV-1-infected corneas from WT and C3-/- mice at day five post infection (p.i.). (B) T cell expansion in the eye-draining mandibular lymph nodes of WT and C3-/- mice. For panels (A) and (B), dashed lines reflect the average value for uninfected WT controls (n = 5 mice per group; two independent experiments; Student’s T). (C) IFNγ expression following stimulation with PMA and ionomycin using T cells harvested from WT or C3-/- mice at day eight post infection (n = 7 unstimulated and 10 activated replicates from three independent experiments; one-way ANOVA, Bonferroni). (D) Adoptive transfer schematic and corneal sensation measurements in TCRα-/- mice following reconstitution with purified splenic T cells from HSV-infected WT and C3-/- mice (n = 5–9 TCRα-/- mice/group; three independent experiments; two-way ANOVA, Bonferroni).

https://doi.org/10.7554/eLife.48378.005
Figure 2—figure supplement 1
T cell engraftment in recipient TCRα-/- mice.

(A) T cell reconstitution was confirmed by evaluating the total number of T cells in the eye-draining mandibular lymph nodes of HSV-1-infected TCRα-/- mice. (B) Impact of exogenous T cell reconstitution in TCRα-/- mice as determined by HSV-1 titers in the trigeminal ganglia (TG). Data reflect summaries of 5–9 mice/group across three independent experiments (one-way ANOVA, Bonferroni).

https://doi.org/10.7554/eLife.48378.006
Figure 3 with 3 supplements
Antigen-specific CD4 T cells drive corneal sensation loss.

(A) Adoptive transfer schematic and corneal sensation measurements in TCRα-/- mice following reconstitution with purified splenic CD4 or CD8 T cells from HSV-infected WT mice (n = 6–7 TCRα-/- mice/group; two independent experiments; two-way ANOVA, Bonferroni). (B) Corneal sensation measurements at baseline and day seven post infection (p.i.) in WT and OT-II mice following ocular HSV-1 infection (n = 4–6 mice/group; three independent experiments). (C) Percentage of CXCR3-expressing CD4 T cells in peripheral blood from WT and OT-II mice at day 7 p.i.; (n = 5 mice/group; two independent experiments). (D) Verification of CD4 T cell infiltration into corneas of WT and OT-II mice at day 7 p.i. (n = 3 mice/group; two independent experiments). Data in panels B-D were analyzed using Student’s T tests.

https://doi.org/10.7554/eLife.48378.007
Figure 3—figure supplement 1
Engraftment of donor T cells into TCRα-/- mice.

T cell reconstitution was confirmed by evaluating the total number of T cells in the eye-draining mandibular lymph nodes (MLN) of HSV-1-infected TCRα-/- mice by flow cytometry 8 days post HSV-1 infection (n = 6–7 TCRα-/- mice/group; two independent experiments; one-way ANOVA, Bonferroni).

https://doi.org/10.7554/eLife.48378.008
Figure 3—figure supplement 2
Antigen-specific donor CD4 T cells drive sensation loss during HSV-1 keratitis.

Adoptive transfer schematic and corneal sensation measurements at baseline and day seven post infection in HSV-1-infected TCRα-/- mice following reconstitution with purified splenic CD4 T cells from HSV-infected WT or OT-II mice (n= 5 TCRα-/- mice/group; two independent experiments; one-way ANOVA comparing each timepoint, Bonferroni posttest).

https://doi.org/10.7554/eLife.48378.009
Figure 3—figure supplement 3
Active corneal HSV-1 infection is required to drive sensation loss in herpetic keratitis.

Corneal sensation in TCRα-/- mice following reconstitution with purified CD4 T cells from HSV-1 infected WT mice as in Figure 3A, with recipient TCRα-/- mice undergoing mock infection (corneal scratch injury). T cell engraftment was confirmed by flow cytometry using MLN from TCRα-/- mice 14 days after mock infection (n = 3–5 TCRα-/- mice/group; two independent experiments; two-way ANOVA, Student’s T test).

https://doi.org/10.7554/eLife.48378.010
Corneal HSV-1 infection enhances local complement synthesis.

Gene expression of complement effectors (A), receptors (B), and regulators (C) upregulated in the corneas of B6 mice during acute HSV-1 infection (n = 7 WT mice/group; two independent experiments; Kruskal-Wallis, Dunn’s multiple comparisons test). (D) Relative C3 expression among selected cornea-resident and infiltrating cell subsets at day three post-infection (n = 3–4 pooled samples from two mice each for cell subsets or three independent cornea pairs; two independent experiments; one-way ANOVA, Bonferroni; ND, not detected/amplification cycle >35). Final PCR products were resolved on an agarose gel to verify amplification. Data are relative to GAPDH expression and normalized to uninfected control samples for panels A-C or to purified CSF1R-expressing peripheral blood monocytes/macrophages in panel D.

https://doi.org/10.7554/eLife.48378.011
Figure 5 with 2 supplements
Local C3 depletion prevents HSV-associated corneal sensation loss.

B6 mice were given PBS (vehicle) or 5.0 μg cobra venom factor (CVF) via subconjunctival injection to degrade C3, and ocularly infected with HSV-1 18 hr later. C3 depletion was maintained by daily topical treatment (eyedrop) containing 0.5 μg CVF. (A) Corneal sensation following HSV-1 infection in animals treated with CVF or PBS (n = 5–11 mice/group; three independent experiments; two-way ANOVA, Bonferroni). Impact of CVF treatment on C3 protein concentrations in the cornea (B) and serum (C) (n= 3 cornea pairs, 5–9 serum samples/timepoint; 2–3 independent experiments; one-way ANOVA, Tukey). (D) Corneal edema measurements (central corneal thickness) determined via spectral domain optical coherence tomography (SD-OCT) on uninfected (UI) or HSV-1 infected mice treated with CVF or PBS at day 5 p.i. (n = 4–5 mice/group; two experiments; one-way ANOVA, Newman-Keuls). (E) Impact of CVF treatment on total leukocyte (CD45+) and monocyte/macrophage (CSF1R+) infiltration into the corneas of CVF and PBS-treated MaFIA (CSF1R-GFP) mice at day 3 p.i. (n = 5–6 mice/group; two independent experiments; Student’s T). (F) Representative flow plots showing cell populations in healthy and HSV-1 corneas from panel E. (G) Impact of CVF treatment on leukocyte infiltration into the corneas of HSV-1 infected mice at day 7 p.i. (n = 4–5 mice/group; two independent experiments; Student’s T). Note: Total CD45+ graph in panel G reflects data from two technical replicates. Dashed lines in panels E and G reflect cell number recorded in healthy uninfected mice.

https://doi.org/10.7554/eLife.48378.013
Figure 5—figure supplement 1
Impact of ocular cobra venom factor treatment on HSV-1 titers.

The ocular surface of B6 mice was treated with PBS or CVF and animals ocularly infected with HSV-1 as in Figure 5. Systemic effects of CVF treatment were evaluated in terms of viral burden in the corneas and trigeminal ganglia (TG) at days 3 (A) and 7 (B) post infection (p.i.). Data in reflect three cornea pairs and 8–9 TG specimens per timepoint; data are composite from two independent experiments and analyzed using Student’s T tests.

https://doi.org/10.7554/eLife.48378.014
Figure 5—figure supplement 2
Immunologic impacts of ocular cobra venom factor treatment.

The ocular surface of B6 mice was treated with PBS or CVF and animals ocularly infected with HSV-1 as in Figure 5. Immunologic impact of CVF treatment was assessed by flow cytometry on the eye-draining mandibular lymph nodes (MLN) (A) to assess CD4+, CD8+, and HSV-1 glycoprotein B (gB)-specific CD8+ T cell numbers or blood (B) to evaluate CD4 count and percentage of activated (CXCR3+) CD4 T cells at day 7 p.i. Data in panel A reflects 6–8 mice/group; panel B reflects five samples/group; data are composite from two independent experiments and analyzed using Student’s T tests.

https://doi.org/10.7554/eLife.48378.015
Chronic ocular allergy does not provoke frank corneal sensation loss.

(A) Schematic of allergic eye disease induced by ovalbumin (OVA) immunization followed by topical ocular OVA challenge. Healthy and OVA-challenged B6 mice were evaluated for signs of ophthalmic allergy (B) and corneal sensation thresholds (C). (D) Representative confocal images of cornea flat mounts from healthy and OVA-challenged mice showing nerves (βΙII Tubulin, red) and infiltrating T cells (CD3, green) in the central cornea at 20x magnification (scale bar = 100 μm). (E) Morphometric analysis of nerve densities in the central cornea based on confocal images. Normalization is based on the average volume in the healthy control group. (F) Quantification of total CD3+ cells per field of view. (G) Flow cytometry was used to confirm CD4 T cell infiltration into the corneas of OVA-challenged mice. Data in panels B – F reflect 5 to 6 mice per group; flow plots in panel G reflect pooled digests of corneas from three mice. Data in panels E and F were analyzed using Student’s T tests.

https://doi.org/10.7554/eLife.48378.016
CD4 T cells drive corneal sensation loss in ocular GVHD.

(A) Schematic of GVHD induction using T cell depleted bone marrow (TCD-BM) or TCD-BM with splenic T cells isolated from C57BL/6 donors and transferred into C3.SW-H2 b recipients. (B) Corneal sensation measurements in C3.SW-H2 b mice following reconstitution with BM only or with BM and 1.3x10 6 CD4, CD8, or CD4 and CD8 T cells. (C) External photographs showing CD4-dependent ocular surface morbidities consistent with corneal sensation loss. (D) Representative confocal images of cornea flat mounts from each group of mice at the study endpoint showing nerves (βΙII Tubulin, red) and infiltrating T cells (CD3, green) in the central cornea at 20x magnification (scale bar = 100 μm). (E) Morphometric analysis of nerve densities based on confocal images. Normalization is based on the average volume in the BM only control group. (F) Quantification of total CD3 + cells per field of view. Data reflect independent measurements of corneas from 3-4 mice per group across 2 experiments; sensation data were evaluated by two-way ANOVA with Bonferroni posttests; data in panels E and F were analyzed by one-way ANOVA with Bonferroni posttests. Figure was generated using Servier Medical Art (http://smart.servier.com/) under a Creative Commons 3.0 license.

https://doi.org/10.7554/eLife.48378.017
Figure 8 with 1 supplement
Local C3 depletion prevents corneal sensation loss in ocular GVHD.

(A) Experiment schematic of GVHD induction using T cell depleted bone marrow (TCD-BM) or TCD-BM with splenic T cells isolated from C57BL/6 donors and transferred into C3.SW-H2 b recipients. Each group was subsequently given a subconjunctival injection containing PBS (vehicle) or 5.0 μg cobra venom factor (CVF) to degrade C3. C3 depletion was maintained by topical treatment (eyedrop) containing 2.0 μg CVF twice weekly. (B) Systemic disease scores in each cohort (n=8 mice per group; two-way ANOVA, Bonferroni). (C) Impact of CVF treatment on serum C3 protein concentrations in each group at the experiment endpoint (n=5-6 samples/group; one-way ANOVA, Bonferroni). (D) Evaluation of CD4:CD8 ratios in secondary lymphoid organs by flow cytometry to confirm onset of GVHD at experimental endpoints (n=3-8 mice/group; 2 independent experiments; one-way ANOVA, Bonferroni). (E) Longitudinal corneal sensation measurements in each group of C3.SW-H2 b mice. (F) Representative confocal images of cornea flat mounts from each group of C3.SW-H2 b mice at the study endpoint showing nerves (βΙII Tubulin, red) and infiltrating T cells (CD3, green) in the central cornea at 20x magnification (scale bar = 100 μm). (G) Morphometric analysis of nerve densities based on confocal images. Normalization is based on the average volume in the BM only control group. (H) Quantification of total CD3 + cells per field of view. Data in panels E – H reflect 3-8 mice/group; 2 independent experiments; two-way ANOVA with Bonferroni posttests. Figure was generated using Servier Medical Art (http://smart.servier.com/) under a Creative Commons 3.0 license.

https://doi.org/10.7554/eLife.48378.019
Figure 8—figure supplement 1
Sex-biased effects of local C3 depletion on ocular GVHD severity.

Models of GVHD were established as described in Figure 8 with cohorts of bone marrow (BM) only controls and GVHD (BM + T cells) mice exposed to ocular treatment with PBS (vehicle) or cobra venom factor (CVF) to degrade complement C3. (A) Representative external photographs showing ocular surface morbidities in each treatment cohort from Figure 8 at the experiment endpoint. Note the sex-biased impact in the CVF-treated GVHD group where treatment limits eyelid swelling in female but not male mice. (B) Ophthalmic disease scores for each experimental group by sex (n = 5 females or three males per group; two-way ANOVA, Bonferroni).

https://doi.org/10.7554/eLife.48378.020

Tables

Table 1
Complement regulatory factor expression in the cornea during HSV-1 infection
https://doi.org/10.7554/eLife.48378.012
GeneUninfectedDay 2 p.i.Day 7 p.i.
C4bpNDNDND
Crry (CD46 homolog)1.193 ± 0.3380.817 ± 0.1721.137 ± 0.331
CD55 (DAF)1.087 ± 0.2011.035 ± 0.1990.791 ± 0.251
CD59a (MIRL)1.078 ± 0.1920.611 ± 0.1230.728 ± 0.191
CfiNDNDND
Cr1l1.026 ± 0.0960.7442 ± 0.2220.889 ± 0.133
  1. Data are expressed as mean ± SEM; n = 7 samples/group. Gene expression is standardized to internal GAPDH expression and relative to uninfected control tissue. Abbreviations: C4bp, C4 binding protein; CD, cluster of differentiation; Cfi, complement factor I, Cr1l, complement C3b/C4b receptor-1-like; DAF, decay accelerating factor; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; MIRL, membrane inhibitor of reactive lysis; ND, not detected (amplification cycle >35); p.i., post-infection.

Table 2
Ophthalmic Disease Scoring Guide for Ocular GVHD.
https://doi.org/10.7554/eLife.48378.018
CriteriaGrade 0Grade 1Grade 2Grade 3
Eyelid ClosureNoneMild squintingPartial lid closureFull lid closure
BlepharitisNoneMild lid margin edemaModerate lid margin edema with crustingGross lid edema, loss of lashes/eyelid fur
Meibomian Gland DiseaseNone≤4 plugs>4 plugs OR cystic plugs>4 plugs AND cystic plugs
Conjunctival
Chemosis
NoneMild edemaModerate edemaGross edema
Conjunctival HyperemiaNoneMild rednessBroad erythemaN/A
Mucoid DischargeNoneMild dischargeDischarge covers the ocular surfaceN/A
Corneal OpacityNoneEpithelial hazeFocal opacitiesWidespread opacity
Key resources table
Reagent type
(species) or
resource
DesignationSource or
reference
IdentifiersAdditional
information
Strain, strain background (Mus musculus, C57BL/6J)Wildtype C57BL/6 (WT)Jackson LaboratoriesStock # 000664
Strain, strain background (M. musculus, C57BL/6J)Complement C3 deficient (C3-/-)Jackson LaboratoriesStock # 029661
Strain, strain background (M. musculus, C57BL/6J)T cell receptor alpha deficient (TCRα-/-)Jackson LaboratoriesStock # 002116
Strain, strain background (M. musculus, C57BL/6J)Transgenic OVA-specific CD4 T cells (OT-II)Jackson LaboratoriesStock # 004194
Strain, strain background (M. musculus, C57BL/6J)Transgenic Csf1r-eGFP (MAFIA)Jackson LaboratoriesStock # 005070
Strain, strain background (M. musculus, C57BL/6J)C3.SW-H2bJackson LaboratoriesStock # 000438
Strain, strain background (Herpes simplex virus type 1, McKrae)HSV-1Macdonald et al., 2012; Watson et al., 2012N/AOriginally from Brian Gebhardt
Cell line (Cercopithecus aethiops)VeroAmerican Type Culture Collection (ATCC)Cat. # CCL-81see Materials and methods
Chemical compound, drugCobra Venom Factor, Naja naja kaouthiaMillipore-SigmaCat. # 233552
Commercial assay or kitAnti-mouse CD90.2 IMag particlesBDCat. # 551518
Commercial assay or kitAnti-mouse CD4 microbeadsMiltenyi BiotecCat. # 130-049-201
Commercial assay or kitAnti-mouse CD8 microbeadsMiltenyi BiotecCat. # 130-116-478
Commercial assay or kitAnti-mouse CD90.2 microbeadsMiltenyi BiotecCat. # 130-049-101
Commercial assay or kitAnti-mouse CD45 microbeadsMiltenyi BiotecCat. # 130-052-301
Commercial assay or kitAnti-mouse EpCAM microbeadsMiltenyi BiotecCat. # 130-105-958
AntibodyRabbit polyclonal IgG (anti-mouse beta-III tubulin)AbcamCat. # ab18207(1:500)
AntibodyAlexaFluor647 donkey polyclonal IgG
(anti-rabbit IgG)
Jackson ImmunoresearchCat. # 711-605-152(1:1000)
AntibodyAnti-mouse CD3e FITC
(Clone: 145–2 C11)
eBioscience (ThermoFisher)Cat. # 11-0031-82(1:500)
OtherRoche Liberase TLSigma AldrichCat. # 5401020001see Materials and methods
Commercial assay or kitC3 ELISAAbcamCat. # ab157711
Commercial assay or kitMilliplex MAP Luminex ArrayEMD MilliporeCustom Design
Recombinant DNA reagentiScript cDNA synthesis kitBioradCat. # 1708891
Recombinant DNA reagentSso Advanced SYBR Green qPCR SupermixBioradCat. # 1725270
OtherTrizolThermoFisherCat. # 15596026see Materials and methods
Software, algorithmPrism 6GraphPadN/A
Software, algorithmImaris x64 (8.2.1)BitplaneN/A

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Derek J Royer
  2. Jose Echegaray-Mendez
  3. Liwen Lin
  4. Grzegorz B Gmyrek
  5. Rose Mathew
  6. Daniel R Saban
  7. Victor L Perez
  8. Daniel JJ Carr
(2019)
Complement and CD4+ T cells drive context-specific corneal sensory neuropathy
eLife 8:e48378.
https://doi.org/10.7554/eLife.48378