Sjögren-Like Lacrimal Keratoconjunctivitis in Germ-Free Mice

Changjun Wang, Mahira Zaheer, Fang Bian, Darin Quach, Alton G Swennes, Robert A Britton, Stephen C Pflugfelder, Cintia S de Paiva, Changjun Wang, Mahira Zaheer, Fang Bian, Darin Quach, Alton G Swennes, Robert A Britton, Stephen C Pflugfelder, Cintia S de Paiva

Abstract

Commensal bacteria play an important role in the formation of the immune system but their role in the maintenance of immune homeostasis at the ocular surface and lacrimal gland remains poorly understood. This study investigated the eye and lacrimal gland phenotype in germ-free and conventional C57BL/6J mice. Our results showed that germ-free mice had significantly greater corneal barrier disruption, greater goblet cell loss, and greater total inflammatory cell and CD4⁺ T cell infiltration within the lacrimal gland compared to the conventionally housed group. A greater frequency of CD4⁺IFN-γ⁺ cells was observed in germ-free lacrimal glands. Females exhibited a more severe phenotype compared to males. Adoptive transfer of CD4⁺ T cells isolated from female germ-free mice into RAG1KO mice transferred Sjögren-like lacrimal keratoconjunctivitis. Fecal microbiota transplant from conventional mice reverted dry eye phenotype in germ-free mice and decreased CD4⁺IFN-γ⁺ cells to levels similar to conventional C57BL/6J mice. These findings indicate that germ-free mice have a spontaneous lacrimal keratoconjunctivitis similar to that observed in Sjögren syndrome patients and demonstrate that commensal bacteria function in maintaining immune homeostasis on the ocular surface. Thus, manipulation of intestinal commensal bacteria has the potential to become a novel therapeutic approach to treat Sjögren Syndrome.

Keywords: Sjögren syndrome; commensal bacteria; dry eye; fecal transplant; germ-free mice; goblet cell.

Conflict of interest statement

All authors state that they have no conflicts of interest.

Figures

Figure 1
Figure 1
Sjögren-like Lacrimal Keratoconjunctivitis on germ-free mice. Conventionally housed mice with complex microbiota served as control (CON) and were compared to germ-free (GF) mice at 8 weeks of age. Both sexes were pooled. (A) Corneal Oregon-Green dextran fluorescence intensity score. Bar graphs show means ± SEM of two independent experiments with four to five animals per experiment (final n = 8–10 animals, mixed sex); (B) Representative images of corneas stained with Oregon Green dextran (OGD); (C) Number of Periodic acid–Schiff (PAS)+ conjunctival goblet cells counted in paraffin-embedded sections expressed as number per millimeter. Bar graph show means ± SEM of two independent experiments with three animals per group, yielding a final sample of six right eyes for each group); (D) Representative images of conjunctiva sections stained with PAS used to generate the bar graph in (C); (E) Total lacrimal gland infiltration measured in haematoxylin and eosin (H&E) stained sections as shown in (F) (n = 6 right lacrimal gland); (F) Representative images of H&E-stained sections of lacrimal gland. Black rectangular insets are a high magnification of dotted square; (G) Flow cytometric analysis of CD4+ and CD8+ T cells and B220+ B cells in lacrimal gland. Right and left extraorbital lacrimal glands from one mouse per group were excised and pooled into a single tube, yielding a final sample of 12 individual lacrimal gland samples divided into two independent experiments with six samples per experiment. Bar graphs show means ± SEM; (H) Tear epidermal growth factor (EGF) concentrations were measured by enzyme-linked immunosorbent assay. Tear washings from both right and left eyes from one mouse per group were collected and pooled into a single tube, yielding a final sample of 12 individual samples per group and divided into three independent experiments with four samples per experiment) Mann-Whitney U test was used for germ-free vs. conventional mice comparisons.
Figure 2
Figure 2
DC frequency and production of IL-12 is altered in germ-free mice. Single cell suspensions of the conjunctiva (CJ), cervical lymph nodes (CLN) and lacrimal glands (LG) were prepared and stained for antigen-presenting cell and macrophage markers followed by intracellular staining for IL-12. Bar graphs are shown as means ± SEM of a representative experiment with 4–5 samples per group. The experiment was repeated once with similar results. (A) Accumulative data showing frequency of CD11b+CD11c+ and F4/80+CD11b+ cells among alive CD45+ gated cells; (B) Accumulative data showing frequency of IL-12+ among viable CD45+ gated cells; (C) Representative overlaid histogram of IL-12 staining in conjunctival CD11b+CD11c+ and F4/80+CD11b+ cells; (D) Accumulative data showing median fluorescence intensity (MIF) of IL-12 in positive cells; (E) Relative fold expression changes of IL-12 in full-thickness conjunctival biopsies. Bar graphs show means ± SD of five samples per group, biological replicates from two independent experiments were averaged. FMO = fluorescence minus one; GF = germ-free; CON = conventional mice; * p < 0.05; ** p < 0.01; *** p < 0.001. Mann–Whitney U test was used for GF vs. CON comparison test.
Figure 3
Figure 3
Increased percentage of Th1+ cells in lacrimal gland of germ-free mice. Percentage of CD4+Foxp3+, Th1 (CD4+IFN-γ+), Th2 (CD4+IL-13+), Th17 (CD4+IL-17+) cells in freshly isolated cervical lymph nodes (CLN) and lacrimal glands (LG) by flow cytometry. Each experiment group consisted of three-to-four samples; showing mean ± SEM of two independent experiments (final n = seven to eight per group). CLN of the same animal were pooled; right and left extraorbital LGs of the same animal were pooled. * p < 0.05 conventional vs. germ-free comparison using Mann-Whitney U test.
Figure 4
Figure 4
CD4+T Cells from germ-free mice transfer Sjögren-like lacrimal Keratoconjunctivitis to immunodeficient RAG1KO mice. CD4+T cells were isolated from spleens and cervical lymph nodes (CLN) from conventionally housed (CON) and germ-free (GF) mice and adoptively transferred (→AT) into RAG1KO mice. Disease severity parameters were evaluated five weeks later. Bar graphs show means ± SEM of a representative experiment with 18–20 samples per group. (A) Corneal barrier function measured by Oregon-Green dextran fluorescence intensity score. Bar graphs show means ± SEM of two independent experiments with four animals per experiment (final n = eight animals, female sex). Dotted line demonstrates OGD fluorescence intensity score in naïve RAG1KO mice; (B) Number of PAS+ conjunctival goblet cells counted in paraffin-embedded sections expressed as number per millimeter. Bar graphs show means ± SEM of two independent experiments with three animals per group, yielding a final sample of six right eyes for each group. Dotted line demonstrates goblet cell density in naïve RAG1KO mice; (C) Representative images of conjunctiva sections stained with PAS (purple cells) used to generate the bar graph in (B). Black rectangular insets are a high magnification of the small demarcated area; (D) Total lacrimal gland infiltration measured in H&E stained sections as shown in (F) (n = six right lacrimal glands); (E) Representative images of haematoxylin and eosin (H&E)-stained sections. Black rectangular insets are a high magnification of small demarcated area; (F) Percentage of total CD4+ T, Th1 (CD4+IFN-γ+), Th2 (CD4+IL-13+), Th17 (CD4+IL-17+) cells in freshly isolated cervical lymph nodes (CLN) and lacrimal glands (LG) by flow cytometry five weeks post-adoptive transfer. Each experiment group consisted of three-to-four samples; showing mean ± SEM of two independent experiments. CLN of the same animal were pooled; right and left extraorbital LGs of the same animal were pooled. Bar graphs show means ± SEM of two independent experiments with four animals per experiment (final n = eight animals, female sex); (G) Relative fold expression changes of MHC II, IL-, TNF-α and Caspase 3 in LG. Bar graphs show means ± SD of six samples per group, biological replicates from two independent experiments were averaged. * p < 0.05; *** p < 0.001Mann-Whitney U GF vs. CON comparison test.
Figure 5
Figure 5
Reconstitution of germ-free mice with commensal bacteria reverses the dry eye phenotype. (AC) Four-week-old female germ-free mice were colonized with a fecal slurry from normal mice (a pool of three mice) by intragastric gavage and sacrificed at eight weeks of age (germ-free + Fecal gavage, GF + FG). (A) Comparison of α diversity between starting inoculum (C57BL/6J) and germ-free conventionalized mice (n = seven) using measures of observed OTUs and Shannon’s diversity index; (B) Relative abundance of major bacterial phyla in the starting inoculum (C57BL/6J pool) used to conventionalize mice and in the gut microbiota from conventionalized germ-free mice that received fecal gavage (n = seven) at eight weeks of age; (C) Corneal Oregon-Green dextran fluorescence intensity score. Bar graphs show means ± SEM of two independent experiments with four animals per experiment (final n = eight animals, female sex). Mann-Whitney U comparison test; (D) Number of PAS+ conjunctival goblet cells counted in paraffin-embedded sections expressed as number per millimeter. Bar graphs show means ± SEM of two independent experiments with three-four animals per group, yielding a final sample of seven right eyes for each group). Mann-Whitney U comparison test; (E) Representative images of conjunctiva sections stained with PAS used to generate the bar graph in (D); (F) Flow cytometry analysis showing the percentage of total CD4+, Th1 (CD4+IFN-γ+), Th17 (CD4+IL-17+) cells in freshly isolated cervical lymph nodes (CLN) and lacrimal glands (LG) in adoptive transfer (→AT) recipients of either germ-free or germ-free + FG CD4+T cells. Each experiment group consisted of three-to-four samples; showing means ± SEM of two independent experiments, yielding a final sample of seven to eight per group. Mann-Whitney U comparison test; (G) Fecal transplant during desiccating stress (DS) rescues goblet cells. Female conventional C57BL/6 mice were left untreated (naïve mice) or received a cocktail of antibiotics (ABX) for seven days. On the morning of the 8th day, mice were switched to normal water and subjected to desiccating stress (DS) for ten days (DS10) and randomized to receive either PBS or oral gavage of fecal material or were left non-stressed. Mice under DS were sacrificed after ten days, and the number of PAS+ cells in the conjunctiva was determined. n = five animals/group. Kruskal-Wallis test followed by Tukey’s post hoc test. NS = non-significant.
Figure 5
Figure 5
Reconstitution of germ-free mice with commensal bacteria reverses the dry eye phenotype. (AC) Four-week-old female germ-free mice were colonized with a fecal slurry from normal mice (a pool of three mice) by intragastric gavage and sacrificed at eight weeks of age (germ-free + Fecal gavage, GF + FG). (A) Comparison of α diversity between starting inoculum (C57BL/6J) and germ-free conventionalized mice (n = seven) using measures of observed OTUs and Shannon’s diversity index; (B) Relative abundance of major bacterial phyla in the starting inoculum (C57BL/6J pool) used to conventionalize mice and in the gut microbiota from conventionalized germ-free mice that received fecal gavage (n = seven) at eight weeks of age; (C) Corneal Oregon-Green dextran fluorescence intensity score. Bar graphs show means ± SEM of two independent experiments with four animals per experiment (final n = eight animals, female sex). Mann-Whitney U comparison test; (D) Number of PAS+ conjunctival goblet cells counted in paraffin-embedded sections expressed as number per millimeter. Bar graphs show means ± SEM of two independent experiments with three-four animals per group, yielding a final sample of seven right eyes for each group). Mann-Whitney U comparison test; (E) Representative images of conjunctiva sections stained with PAS used to generate the bar graph in (D); (F) Flow cytometry analysis showing the percentage of total CD4+, Th1 (CD4+IFN-γ+), Th17 (CD4+IL-17+) cells in freshly isolated cervical lymph nodes (CLN) and lacrimal glands (LG) in adoptive transfer (→AT) recipients of either germ-free or germ-free + FG CD4+T cells. Each experiment group consisted of three-to-four samples; showing means ± SEM of two independent experiments, yielding a final sample of seven to eight per group. Mann-Whitney U comparison test; (G) Fecal transplant during desiccating stress (DS) rescues goblet cells. Female conventional C57BL/6 mice were left untreated (naïve mice) or received a cocktail of antibiotics (ABX) for seven days. On the morning of the 8th day, mice were switched to normal water and subjected to desiccating stress (DS) for ten days (DS10) and randomized to receive either PBS or oral gavage of fecal material or were left non-stressed. Mice under DS were sacrificed after ten days, and the number of PAS+ cells in the conjunctiva was determined. n = five animals/group. Kruskal-Wallis test followed by Tukey’s post hoc test. NS = non-significant.

References

    1. Nolan J. Evaluation of conjunctival and nasal bacterial cultures before intra-ocular operations. Br. J. Ophthalmol. 1967;51:483–485. doi: 10.1136/bjo.51.7.483.
    1. Fahmy J.A., Moller S., Bentzon M.W. Bacterial flora in relation to cataract extraction. II. Peroperative flora. Acta Ophthalmol. 1975;53:476–494. doi: 10.1111/j.1755-3768.1975.tb01178.x.
    1. Schabereiter-Gurtner C., Maca S., Rolleke S., Nigl K., Lukas J., Hirschl A., Lubitz W., Barisani-Asenbauer T. 16S rDNA-based identification of bacteria from conjunctival swabs by PCR and DGGE fingerprinting. Investig. Ophthalmol. Vis. Sci. 2001;42:1164–1171.
    1. Narayanan S., Miller W.L., McDermott A.M. Expression of human β-defensins in conjunctival epithelium: Relevance to dry eye disease. Investig. Ophthalmol. Vis. Sci. 2003;44:3795–3801. doi: 10.1167/iovs.02-1301.
    1. Zhou L., Huang L.Q., Beuerman R.W., Grigg M.E., Li S.F., Chew F.T., Ang L., Stern M.E., Tan D. Proteomic analysis of human tears: Defensin expression after ocular surface surgery. J. Proteome Res. 2004;3:410–416. doi: 10.1021/pr034065n.
    1. McDermott A.M. Antimicrobial compounds in tears. Exp. Eye Res. 2013;117:53–61. doi: 10.1016/j.exer.2013.07.014.
    1. Jensen O.L., Gluud B.S., Birgens H.S. The concentration of lactoferrin in tears of normals and of diabetics. Acta Ophthalmol. 1986;64:83–87. doi: 10.1111/j.1755-3768.1986.tb06877.x.
    1. Vinding T., Eriksen J.S., Nielsen N.V. The concentration of lysozyme and secretory IgA in tears from healthy persons with and without contact lens use. Acta Ophthalmol. 1987;65:23–26. doi: 10.1111/j.1755-3768.1987.tb08485.x.
    1. Haynes R.J., Tighe P.J., Dua H.S. Antimicrobial defensin peptides of the human ocular surface. Br. J. Ophthalmol. 1999;83:737–741. doi: 10.1136/bjo.83.6.737.
    1. Willcox M.D. Characterization of the normal microbiota of the ocular surface. Exp. Eye Res. 2013;117:99–105. doi: 10.1016/j.exer.2013.06.003.
    1. Zegans M.E., van Gelder R.N. Considerations in understanding the ocular surface microbiome. Am. J. Ophthalmol. 2014;158:420–422. doi: 10.1016/j.ajo.2014.06.014.
    1. De Paiva C.S., Jones D.B., Stern M.E., Bian F., Moore Q.L., Corbiere S., Streckfus C.F., Hutchinson D.S., Ajami N.J., Petrosino J.F., et al. Altered Mucosal Microbiome Diversity and Disease Severity in Sjogren Syndrome. Sci. Rep. 2016;6:23561–23571. doi: 10.1038/srep23561.
    1. Huang Y., Yang B., Li W. Defining the normal “core microbiome” of conjunctival microbial communities. Clin. Microbiol. Infect. 2016;22:643. doi: 10.1016/j.cmi.2016.04.008.
    1. Graham J.E., Moore J.E., Jiru X., Moore J.E., Goodall E.A., Dooley J.S., Hayes V.E., Dartt D.A., Downes C.S., Moore T.C. Ocular pathogen or commensal: A PCR-based study of surface bacterial flora in normal and dry eyes. Investig. Ophthalmol. Vis. Sci. 2007;48:5616–5623. doi: 10.1167/iovs.07-0588.
    1. Ozkan J., Nielsen S., Diez-Vives C., Coroneo M., Thomas T., Willcox M. Temporal Stability and Composition of the Ocular Surface Microbiome. Sci. Rep. 2017;7:9880. doi: 10.1038/s41598-017-10494-9.
    1. St Leger A.J., Desai J.V., Drummond R.A., Kugadas A., Almaghrabi F., Silver P., Raychaudhuri K., Gadjeva M., Iwakura Y., Lionakis M.S., et al. An Ocular Commensal Protects against Corneal Infection by Driving an Interleukin-17 Response from Mucosal γδ T Cells. Immunity. 2017;47:148–158. doi: 10.1016/j.immuni.2017.06.014.
    1. Kugadas A., Christiansen S.H., Sankaranarayanan S., Surana N.K., Gauguet S., Kunz R., Fichorova R., Vorup-Jensen T., Gadjeva M. Impact of Microbiota on Resistance to Ocular Pseudomonas aeruginosa-Induced Keratitis. PLoS Pathog. 2016;12:e1005855. doi: 10.1371/journal.ppat.1005855.
    1. Kugadas A., Gadjeva M. Impact of Microbiome on Ocular Health. Ocul. Surf. 2016;14:342–349. doi: 10.1016/j.jtos.2016.04.004.
    1. Kugadas A., Wright Q., Geddes-McAlister J., Gadjeva M. Role of Microbiota in Strengthening Ocular Mucosal Barrier Function through Secretory IgA. Investig. Ophthalmol. Vis. Sci. 2017;58:4593–4600. doi: 10.1167/iovs.17-22119.
    1. Macia L., Tan J., Vieira A.T., Leach K., Stanley D., Luong S., Maruya M., Ian McKenzie C., Hijikata A., Wong C., et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 2015;6:6734. doi: 10.1038/ncomms7734.
    1. Bourassa M.W., Alim I., Bultman S.J., Ratan R.R. Butyrate, neuroepigenetics and the gut microbiome: Can a high fiber diet improve brain health? Neurosci. Lett. 2016;625:56–63. doi: 10.1016/j.neulet.2016.02.009.
    1. Ghaisas S., Maher J., Kanthasamy A. Gut microbiome in health and disease: Linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol. Ther. 2015;158:52–62. doi: 10.1016/j.pharmthera.2015.11.012.
    1. Kigerl K.A., Hall J.C., Wang L., Mo X., Yu Z., Popovich P.G. Gut dysbiosis impairs recovery after spinal cord injury. J. Exp. Med. 2016 doi: 10.1084/jem.20151345.
    1. Luczynski P., McVey Neufeld K.A., Oriach C.S., Clarke G., Dinan T.G., Cryan J.F. Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int. J. Neuropsychopharmacol. 2016;19 doi: 10.1093/ijnp/pyw020.
    1. Maslowski K.M., Vieira A.T., Ng A., Kranich J., Sierro F., Yu D., Schilter H.C., Rolph M.S., Mackay F., Artis D., et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–1286. doi: 10.1038/nature08530.
    1. Dowds C.M., Blumberg R.S., Zeissig S. Control of intestinal homeostasis through crosstalk between natural killer T cells and the intestinal microbiota. Clin. Immunol. 2015;159:128–133. doi: 10.1016/j.clim.2015.05.008.
    1. Blumberg R., Powrie F. Microbiota, disease, and back to health: A metastable journey. Sci. Transl. Med. 2012;4:137rv7. doi: 10.1126/scitranslmed.3004184.
    1. Brandt J.E., Priori R., Valesini G., Fairweather D. Sex differences in Sjogren’s syndrome: A comprehensive review of immune mechanisms. Biol. Sex Differ. 2015;6:19. doi: 10.1186/s13293-015-0037-7.
    1. Schaumberg D.A., Sullivan D.A., Buring J.E., Dana M.R. Prevalence of dry eye syndrome among US women. Am. J. Ophthalmol. 2003;136:318–326. doi: 10.1016/S0002-9394(03)00218-6.
    1. McClellan A.J., Volpe E.A., Zhang X., Darlington G.J., Li D.Q., Pflugfelder S.C., de Paiva C.S. Ocular Surface Disease and Dacryoadenitis in Aging C57BL/6 Mice. Am. J. Pathol. 2014;184:631–643. doi: 10.1016/j.ajpath.2013.11.019.
    1. Bian F., Barbosa F.L., Corrales R.M., Pelegrino F.S., Volpe E.A., Pflugfelder S.C., de Paiva C.S. Altered balance of interleukin-13/interferon-γ contributes to lacrimal gland destruction and secretory dysfunction in CD25 knockout model of Sjogren’s syndrome. Arthritis Res. Ther. 2015;17:53. doi: 10.1186/s13075-015-0582-9.
    1. Yoshino K., Monroy D., Pflugfelder S.C. Cholinergic stimulation of lactoferrin and epidermal growth factor secretion by the human lacrimal gland. Cornea. 1996;15:617–621. doi: 10.1097/00003226-199611000-00013.
    1. Pelegrino F.S., Volpe E.A., Gandhi N.B., Li D.Q., Pflugfelder S.C., de Paiva C.S. Deletion of interferon-γ delays onset and severity of dacryoadenitis in CD25KO mice. Arthritis Res. Ther. 2012;14:R234. doi: 10.1186/ar4077.
    1. Cha S., Nagashima H., Brown V.B., Peck A.B., Humphreys-Beher M.G. Two NOD Idd-associated intervals contribute synergistically to the development of autoimmune exocrinopathy (Sjogren’s syndrome) on a healthy murine background. Arthritis Rheum. 2002;46:1390–1398. doi: 10.1002/art.10258.
    1. Cha S., Peck A.B., Humphreys-Beher M.G. Progress in understanding autoimmune exocrinopathy using the non-obese diabetic mouse: An update. Crit. Rev. Oral Biol. Med. 2002;13:5–16. doi: 10.1177/154411130201300103.
    1. Xiao Y., Coursey T.G., Li D.Q., de Paiva C.S., Tukler Henriksson J., Pflugfelder S.C. Conjunctival Goblet Cells modulate dendritic cell maturation and retinoic acid producing capacity. Investig. Ophthalmol. Vis. Sci. 2016;57:426.
    1. Contreras-Ruiz L., Masli S. Immunomodulatory cross-talk between conjunctival goblet cells and dendritic cells. PLoS ONE. 2015;10:e0120284. doi: 10.1371/journal.pone.0120284.
    1. Barbosa F.L., Xiao Y., Bian F., Coursey T.G., Ko B.Y., Clevers H., de Paiva C.S., Pflugfelder S.C. Goblet Cells Contribute to Ocular Surface Immune Tolerance-Implications for Dry Eye Disease. Int. J. Mol. Sci. 2017;18:978. doi: 10.3390/ijms18050978.
    1. Marko C.K., Menon B.B., Chen G., Whitsett J.A., Clevers H., Gipson I.K. Spdef null mice lack conjunctival goblet cells and provide a model of dry eye. Am. J. Pathol. 2013;183:35–48. doi: 10.1016/j.ajpath.2013.03.017.
    1. Coursey T.G., Henriksson J.T., Barbosa F.L., de Paiva C.S., Pflugfelder S.C. Interferon-γ-Induced Unfolded Protein Response in Conjunctival Goblet Cells as a Cause of Mucin Deficiency in Sjogren Syndrome. Am. J. Pathol. 2016;186:1547–1558. doi: 10.1016/j.ajpath.2016.02.004.
    1. Garcia-Posadas L., Hodges R.R., Li D., Shatos M.A., Storr-Paulsen T., Diebold Y., Dartt D.A. Interaction of IFN-γ with cholinergic agonists to modulate rat and human goblet cell function. Mucosal Immunol. 2016;9:206–217. doi: 10.1038/mi.2015.53.
    1. Atarashi K., Tanoue T., Shima T., Imaoka A., Kuwahara T., Momose Y., Cheng G., Yamasaki S., Saito T., Ohba Y., et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–341. doi: 10.1126/science.1198469.
    1. Niederkorn J.Y., Stern M.E., Pflugfelder S.C., de Paiva C.S., Corrales R.M., Gao J., Siemasko K. Desiccating Stress Induces T Cell-Mediated Sjogren’s Syndrome-Like Lacrimal Keratoconjunctivitis. J. Immunol. 2006;176:3950–3957. doi: 10.4049/jimmunol.176.7.3950.
    1. Coursey T.G., Gandhi N.B., Volpe E.A., Pflugfelder S.C., de Paiva C.S. Chemokine receptors CCR6 and CXCR3 are necessary for CD4+ T cell mediated ocular surface disease in experimental dry eye disease. PLoS ONE. 2013;8:e78508. doi: 10.1371/journal.pone.0078508.
    1. Coursey T.G., Bian F., Zaheer M., Pflugfelder S.C., Volpe E.A., de Paiva C.S. Age-related spontaneous lacrimal keratoconjunctivitis is accompanied by dysfunctional T regulatory cells. Mucosal Immunol. 2016;10:743. doi: 10.1038/mi.2016.83.
    1. de Paiva C.S., Villarreal A.L., Corrales R.M., Rahman H.T., Chang V.Y., Farley W.J., Stern M.E., Niederkorn J.Y., Li D.Q., Pflugfelder S.C. Dry Eye-Induced Conjunctival Epithelial Squamous Metaplasia Is Modulated by Interferon-γ. Investig. Ophthalmol. Vis. Sci. 2007;48:2553–2560. doi: 10.1167/iovs.07-0069.
    1. de Paiva C.S., Raince J.K., McClellan A.J., Shanmugam K.P., Pangelinan S.B., Volpe E.A., Corrales R.M., Farley W.J., Corry D.B., Li D.Q., et al. Homeostatic control of conjunctival mucosal goblet cells by NKT-derived IL-13. Mucosal Immunol. 2011;4:397–408. doi: 10.1038/mi.2010.82.
    1. Tazume S., Umehara K., Matsuzawa H., Yoshida T., Hashimoto K., Sasaki S. Immunological function of food-restricted germfree and specific pathogen-free mice. Exp. Anim. 1991;40:523–528. doi: 10.1538/expanim1978.40.4_523.
    1. Al-Asmakh M., Zadjali F. Use of Germ-Free Animal Models in Microbiota-Related Research. J. Microbiol. Biotechnol. 2015;25:1583–1588. doi: 10.4014/jmb.1501.01039.
    1. Kandori H., Hirayama K., Takeda M., Doi K. Histochemical, lectin-histochemical and morphometrical characteristics of intestinal goblet cells of germfree and conventional mice. Exp. Anim. 1996;45:155–160. doi: 10.1538/expanim.45.155.
    1. Moriyama M., Tanaka A., Maehara T., Furukawa S., Nakashima H., Nakamura S. T helper subsets in Sjogren’s syndrome and IgG4-related dacryoadenitis and sialoadenitis: A critical review. J. Autoimmun. 2014;51:81–88. doi: 10.1016/j.jaut.2013.07.007.
    1. Nakabayashi T., Letterio J.J., Geiser A.G., Kong L., Ogawa N., Zhao W., Koike T., Fernandes G., Dang H., Talal N. Up-regulation of cytokine mRNA, adhesion molecule proteins, and MHC class II proteins in salivary glands of TGF-β1 knockout mice: MHC class II is a factor in the pathogenesis of TGF-β1 knockout mice. J. Immunol. 1997;158:5527–5535.
    1. Hayashi T., Shimoyama N., Mizuno T. Destruction of salivary and lacrimal glands by Th1-polarized reaction in a model of secondary Sjogren’s syndrome in lupus-prone female NZB× NZWF1 mice. Inflammation. 2012;35:638–646. doi: 10.1007/s10753-011-9356-y.
    1. Cha S., Brayer J., Gao J., Brown V., Killedar S., Yasunari U., Peck A.B. A dual role for interferon-γ in the pathogenesis of Sjogren’s syndrome-like autoimmune exocrinopathy in the nonobese diabetic mouse. Scand. J. Immunol. 2004;60:552–565. doi: 10.1111/j.0300-9475.2004.01508.x.
    1. Saito I., Terauchi K., Shimuta M., Nishiimura S., Yoshino K., Takeuchi T., Tsubota K., Miyasaka N. Expression of cell adhesion molecules in the salivary and lacrimal glands of Sjogren’s syndrome. J. Clin. Lab. Anal. 1993;7:180–187. doi: 10.1002/jcla.1860070309.
    1. Tsubota K., Fukagawa K., Fujihara T., Shimmura S., Saito I., Saito K., Takeuchi T. Regulation of human leukocyte antigen expression in human conjunctival epithelium. Investig. Ophthalmol. Vis. Sci. 1999;40:28–34.
    1. Zhang X., Chen W., de Paiva C.S., Corrales R.M., Volpe E.A., McClellan A.J., Farley W.J., Li D.Q., Pflugfelder S.C. Interferon-γ exacerbates dry eye-induced apoptosis in conjunctiva through dual apoptotic pathways. Investig. Ophthalmol. Vis. Sci. 2011;52:6279–6285. doi: 10.1167/iovs.10-7081.
    1. Zhang X., Chen W., de Paiva C.S., Volpe E.A., Gandhi N.B., Farley W.J., Li D.Q., Niederkorn J.Y., Stern M.E., Pflugfelder S.C. Desiccating Stress Induces CD4+ T-Cell-Mediated Sjogren’s Syndrome-Like Corneal Epithelial Apoptosis via Activation of the Extrinsic Apoptotic Pathway by Interferon-γ. Am. J. Pathol. 2011;179:1807–1814. doi: 10.1016/j.ajpath.2011.06.030.
    1. Zhang X., de Paiva C.S., Su Z., Volpe E.A., Li D.Q., Pflugfelder S.C. Topical interferon-γ neutralization prevents conjunctival goblet cell loss in experimental murine dry eye. Exp. Eye Res. 2014;118:117–124. doi: 10.1016/j.exer.2013.11.011.
    1. Youinou P., Pers J.O. Disturbance of cytokine networks in Sjogren’s syndrome. Arthritis Res. Ther. 2011;13:227. doi: 10.1186/ar3348.
    1. Roescher N., Tak P.P., Illei G.G. Cytokines in Sjogren’s syndrome: Potential therapeutic targets. Ann. Rheum. Dis. 2010;69:945–948. doi: 10.1136/ard.2009.115378.
    1. Cho S.Y., Kim J., Lee J.H., Sim J.H., Cho D.H., Bae I.H., Lee H., Seol M.A., Shin H.M., Kim T.J., et al. Modulation of gut microbiota and delayed immunosenescence as a result of syringaresinol consumption in middle-aged mice. Sci. Rep. 2016;6:39026. doi: 10.1038/srep39026.
    1. Brookes S.M., Price E.J., Venables P.J., Maini R.N. Interferon-γ and epithelial cell activation in Sjogren’s syndrome. Br. J. Rheumatol. 1995;34:226–231. doi: 10.1093/rheumatology/34.3.226.
    1. Bulosan M., Pauley K.M., Yo K., Chan E.K., Katz J., Peck A.B., Cha S. Inflammatory caspases are critical for enhanced cell death in the target tissue of Sjogren’s syndrome before disease onset. Immunol. Cell Biol. 2008;87:81–90. doi: 10.1038/icb.2008.70.
    1. Clark D.A., Lamey P.J., Jarrett R.F., Onions D.E. A model to study viral and cytokine involvement in Sjogren’s syndrome. Autoimmunity. 1994;18:7–14. doi: 10.3109/08916939409014674.
    1. Daniels P.J., Gustafson S.A., French D., Wang Y., DePond W., McArthur C.P. Interferon-mediated block in cell cycle and altered integrin expression in a differentiated salivary gland cell line (HSG) cultured on Matrigel. J. Interferon Cytokine Res. 2000;20:1101–1109. doi: 10.1089/107999000750053771.
    1. Fox R.I., Kang H.I. Pathogenesis of Sjogren’s syndrome. Rheum. Dis. Clin. N. Am. 1992;18:517–538.
    1. Jin J.O., Kawai T., Cha S., Yu Q. Interleukin-7 enhances the Th1 response to promote the development of Sjogren’s syndrome-like autoimmune exocrinopathy in mice. Arthritis Rheum. 2013;65:2132–2142. doi: 10.1002/art.38007.
    1. Ogawa N., Ping L., Zhenjun L., Takada Y., Sugai S. Involvement of the interferon-γ-induced T cell-attracting chemokines, interferon-γ-inducible 10-kd protein (CXCL10) and monokine induced by interferon-γ (CXCL9), in the salivary gland lesions of patients with Sjogren’s syndrome. Arthritis Rheum. 2002;46:2730–2741. doi: 10.1002/art.10577.
    1. Wu A.J., Chen Z.J., Tsokos M., O’Connell B.C., Ambudkar I.S., Baum B.J. Interferon-γ induced cell death in a cultured human salivary gland cell line. J. Cell Physiol. 1996;167:297–304. doi: 10.1002/(SICI)1097-4652(199605)167:2<297::AID-JCP14>;2-5.
    1. Rahimy E., Pitcher J.D., III, Pangelinan S.B., Chen W., Farley W.J., Niederkorn J.Y., Stern M.E., Li D.Q., Pflugfelder S.C., de Paiva C.S. Spontaneous autoimmune dacryoadenitis in aged CD25KO mice. Am. J. Pathol. 2010;177:744–753. doi: 10.2353/ajpath.2010.091116.
    1. Agrawal M., Aroniadis O.C., Brandt L.J., Kelly C., Freeman S., Surawicz C., Broussard E., Stollman N., Giovanelli A., Smith B., et al. The Long-term Efficacy and Safety of Fecal Microbiota Transplant for Recurrent, Severe, and Complicated Clostridium difficile Infection in 146 Elderly Individuals. J. Clin. Gastroenterol. 2015;50:403–407. doi: 10.1097/MCG.0000000000000410.
    1. Colman R.J., Rubin D.T. Fecal microbiota transplantation as therapy for inflammatory bowel disease: A systematic review and meta-analysis. J. Crohn’s Colitis. 2014;8:1569–1581. doi: 10.1016/j.crohns.2014.08.006.
    1. Kelly C.R., Ihunnah C., Fischer M., Khoruts A., Surawicz C., Afzali A., Aroniadis O., Barto A., Borody T., Giovanelli A., et al. Fecal microbiota transplant for treatment of Clostridium difficile infection in immunocompromised patients. Am. J. Gastroenterol. 2014;109:1065–1071. doi: 10.1038/ajg.2014.133.
    1. de Paiva C.S., Chotikavanich S., Pangelinan S.B., Pitcher J.D., III, Fang B., Zheng X., Ma P., Farley W.J., Siemasko K.S., Niederkorn J.Y., et al. IL-17 disrupts corneal barrier following desiccating stress. Mucosal Immunol. 2009;2:243–253. doi: 10.1038/mi.2009.5.
    1. You I.C., Bian F., Volpe E.A., de Paiva C.S., Pflugfelder S.C. Age-related conjunctival disease in the C57BL/6.NOD-Aec1Aec2 Mouse Model of Sjogren Syndrome develops independent of lacrimal dysfunction. Investig. Ophthalmol. Vis. Sci. 2015;56:2224–2233. doi: 10.1167/iovs.14-15668.
    1. Hill D.A., Siracusa M.C., Abt M.C., Kim B.S., Kobuley D., Kubo M., Kambayashi T., Larosa D.F., Renner E.D., Orange J.S., et al. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat. Med. 2012;18:538–546. doi: 10.1038/nm.2657.
    1. Britton R.A., Irwin R., Quach D., Schaefer L., Zhang J., Lee T., Parameswaran N., McCabe L.R. Probiotic L. reuteri treatment prevents bone loss in a menopausal ovariectomized mouse model. J. Cell. Physiol. 2014;229:1822–1830. doi: 10.1002/jcp.24636.
    1. Auchtung J.M., Robinson C.D., Britton R.A. Cultivation of stable, reproducible microbial communities from different fecal donors using minibioreactor arrays (MBRAs) Microbiome. 2015;3:42. doi: 10.1186/s40168-015-0106-5.
    1. Collins J., Auchtung J.M., Schaefer L., Eaton K.A., Britton R.A. Humanized microbiota mice as a model of recurrent Clostridium difficile disease. Microbiome. 2015;3:35. doi: 10.1186/s40168-015-0097-2.
    1. Schloss P.D., Westcott S.L., Ryabin T., Hall J.R., Hartmann M., Hollister E.B., Lesniewski R.A., Oakley B.B., Parks D.H., Robinson C.J., et al. Introducing mothur: Open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 2009;75:7537–7541. doi: 10.1128/AEM.01541-09.
    1. Kozich J.J., Westcott S.L., Baxter N.T., Highlander S.K., Schloss P.D. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl. Environ. Microbiol. 2013;79:5112–5120. doi: 10.1128/AEM.01043-13.
    1. McMurdie P.J., Holmes S. phyloseq: An R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE. 2013;8:e61217. doi: 10.1371/journal.pone.0061217.

Source: PubMed

3
구독하다