IFN-γ enhances cell-mediated cytotoxicity against keratinocytes via JAK2/STAT1 in lichen planus

Shuai Shao, Lam C Tsoi, Mrinal K Sarkar, Xianying Xing, Ke Xue, Ranjitha Uppala, Celine C Berthier, Chang Zeng, Matthew Patrick, Allison C Billi, Joseph Fullmer, Maria A Beamer, Bethany Perez-White, Spiro Getsios, Andrew Schuler, John J Voorhees, Sung Choi, Paul Harms, J Michelle Kahlenberg, Johann E Gudjonsson, Shuai Shao, Lam C Tsoi, Mrinal K Sarkar, Xianying Xing, Ke Xue, Ranjitha Uppala, Celine C Berthier, Chang Zeng, Matthew Patrick, Allison C Billi, Joseph Fullmer, Maria A Beamer, Bethany Perez-White, Spiro Getsios, Andrew Schuler, John J Voorhees, Sung Choi, Paul Harms, J Michelle Kahlenberg, Johann E Gudjonsson

Abstract

Lichen planus (LP) is a chronic debilitating inflammatory disease of unknown etiology affecting the skin, nails, and mucosa with no current FDA-approved treatments. It is histologically characterized by dense infiltration of T cells and epidermal keratinocyte apoptosis. Using global transcriptomic profiling of patient skin samples, we demonstrate that LP is characterized by a type II interferon (IFN) inflammatory response. The type II IFN, IFN-γ, is demonstrated to prime keratinocytes and increase their susceptibility to CD8+ T cell-mediated cytotoxic responses through MHC class I induction in a coculture model. We show that this process is dependent on Janus kinase 2 (JAK2) and signal transducer and activator of transcription 1 (STAT1), but not JAK1 or STAT2 signaling. Last, using drug prediction algorithms, we identify JAK inhibitors as promising therapeutic agents in LP and demonstrate that the JAK1/2 inhibitor baricitinib fully protects keratinocytes against cell-mediated cytotoxic responses in vitro. In summary, this work elucidates the role and mechanisms of IFN-γ in LP pathogenesis and provides evidence for the therapeutic use of JAK inhibitors to limit cell-mediated cytotoxicity in patients with LP.

Conflict of interest statement

Competing interests: The authors declare no competing interests.

Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.

Figures

Fig. 1.. Transcriptomic profiling of LP lesions…
Fig. 1.. Transcriptomic profiling of LP lesions reflects an IFN-γ-dominant inflammation
(A) Principal components analysis of microarray data from LP (n=20), HLP (n=17) and healthy controls (n=24). (B) The Venn diagram displays the intersection (upper panel) and correlation (lower panel) of differentially expressed genes (FDR1.5 or IFNG, and IFNGR (D), and the IFN-response genes MX1, MX2, OAS1 and OASL (E) in our microarray data. (F) Further analysis and classification of differentially expressed genes in LP and HLP skin lesions to cytokine-induced genes. (G) QRT-PCR was employed to detect the IFNG, MX1, IL17A, and IL22 expression level in skin lesions of LP (n=22) and HLP (n=16) and normal controls (NC, n=23). One-way ANOVA. Data are presented as the mean ± SEM of measurements obtained in each sample. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Fig. 2.. IFN-γ increases keratinocyte susceptibility to…
Fig. 2.. IFN-γ increases keratinocyte susceptibility to cell-mediated cytotoxicity
(A) Keratinocytes from one donor were co-cultured with CD3/CD28 microbeads-activated PBMCs from a second donor (n=2) and cell death was evaluated by Annexin-V PI staining. The representative flow cytometry data (left panel) and statistical analysis (right panel) are shown.(B) TUNEL staining and statistical analysis of TUNEL-positive cells was used to detect keratinocyte cell death. DAPI was used for nuclear staining (scale bar, 100μm). (C) PBMCs were primed with IFN-γ and then treated with CD3/CD28 microbeads, and then added to unstimulated keratinocyte cultures. (D) CD3/CD28 microbeads-activated PBMCs were pre-incubated with anti-CD4, anti-CD8, or anti-Nkp44 blocking antibody for 1 h, then added to in vitro co-culture model, and keratinocyte death was evaluated by Annexin-V PI staining. Data were analyzed using one-way ANOVA followed by Dunnett’s posttest. Data are presented as the mean ± SD of measurements obtained in triplicate or quadruplicate experiments. *P < 0.05, **P < 0.01, ****p < 0.0001.
Fig. 3.. Cytotoxic responses to IFN-γ-primed keratinocytes…
Fig. 3.. Cytotoxic responses to IFN-γ-primed keratinocytes are MHC class I dependent.
(A) The mRNA expression level of all MHC molecules from microarray data from LP (n=20), HLP (n=17), and healthy control skin (n=24). (B) Representative immunofluorescence staining of MHC class I and MHC class II in LP/HLP lesions (n=3) and controls (n=3). DAPI was used to show nuclear staining (Scale bar, 100μm). (C) QRT-PCR showed mRNA expression of MHC I and II molecules in keratinocytes stimulated with IFN-γ (10ng/ml). (D) IFN-γ-primed keratinocyte were pre-incubated with anti-MHC class I and II monoclonal antibody for 1 h, and then co-cultured with CD3/CD28 activated PBMCs. Statistical analysis was done using one-way ANOVA followed by Dunnett’s posttest. Data are presented as the mean ± SD of measurements obtained in triplicate or quadruplicate experiments. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Fig. 4.. IFN-γ signals through JAK2/STAT1 signaling…
Fig. 4.. IFN-γ signals through JAK2/STAT1 signaling in keratinocytes to induce MHC class I expression
(A) Induced modules network analysis of the differentially expressed genes shared by LP and HLP. Node size correlates with the number of connected nodes and edges. Nodes with 5 connections are marked larger. (B) Expression level of STAT1-6 in LP (n=20), HLP (n=17) and healthy control skin (n=24) in our microarray data. (C) Representative immunofluorescence staining of STAT1 and phosphorylated (p)-STAT1 in LP/HLP skin lesions and controls (four samples were stained and analyzed per group). DAPI was used for nuclear staining (scale bar, 100μm). (D) Keratinocytes were stimulated with IFN-γ (10 ng/ml) for indicated time. Western blot for JAK1, JAK2, p-JAK1, p-JAK2, STAT1, STAT2, p-STAT1, and p-STAT2 are shown. (E) mRNA expression of MHC I molecules in JAK1, JAK2, STAT1, and STAT2 KO cells after 24hr IFN-γ (10 ng/ml) stimulation. One-way ANOVA. Data are presented as the mean ± SD of measurements obtained in triplicate experiments. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Fig. 5.. Targeting JAK signaling protects keratinocytes…
Fig. 5.. Targeting JAK signaling protects keratinocytes from cell-mediated cytotoxicity
(A) JAK2 and STAT1 KO cells were primed with IFN-γ and co-cultured with CD3/CD28 activated PBMCs. Keratinocyte cell-death was evaluated for Annexin-V positivity by flow cytometry. (B) The representative flow cytometry data and analysis of keratinocyte cell-death in the co-culture model with or without baricitinib. (C) The mRNA expression of MHC I molecules in IFN-γ-induced keratinocytes with or without baricitinib for 24 h. One-way ANOVA. Data are presented as the mean ± SD of measurements obtained in triplicate or quadruplicate experiments. *P < 0.05, ***P < 0.001, ****P < 0.0001.
Fig. 6.. High IFN-γ responses characterizes diseases…
Fig. 6.. High IFN-γ responses characterizes diseases with prominent epidermal cell death.
(A) Representative TUNEL staining showing cell death in skin lesions of LP, CLE, psoriasis, and normal controls. DAPI was used for nuclear staining (scale bar, 100μm). (B) The mRNA expression of IFNG and MHC I molecules in chronic plaque psoriasis (PV) (n=12 or 7), LP (n=38 or 10), CLE (n=21 or 12) skin lesions, and healthy control skin (NC, n=11 or 15). One-way ANOVA. Data are presented as the mean ± SD of measurements obtained in each sample.(c) Representative immunofluorescence staining of MHC I and MHC II, STAT1 and p-STAT1 in PV and CLE skin lesions and normal controls. These staining were performed on more than 3 patient samples for each group. DAPI was used to highlight nuclear staining (scale bar, 100μm).

References

    1. Weston G, Payette M, Update on lichen planus and its clinical variants. Int J Womens Dermatol 1, 140–149 (2015).
    1. Gorouhi F, Davari P, Fazel N, Cutaneous and mucosal lichen planus: a comprehensive review of clinical subtypes, risk factors, diagnosis, and prognosis. Scientific World Journal 2014, 742826 (2014).
    1. Radwan-Oczko M, Zwyrtek E, Owczarek JE, Szczesniak D, Psychopathological profile and quality of life of patients with oral lichen planus. J Appl Oral Sci 26, e20170146 (2018).
    1. Alrashdan MS, Cirillo N, McCullough M, Oral lichen planus: a literature review and update. Arch Dermatol Res 308, 539–551 (2016).
    1. Giuliani M, Troiano G, Cordaro M, Corsalini M, Gioco G, Lo Muzio L, Pignatelli P, Lajolo C, Rate of malignant transformation of oral lichen planus: A systematic review. Oral Dis, (2018).
    1. Carrozzo M, Broccoletti R, Carbone M, Gandolfo S, Garzino P, Cascio G, Phenotypic analysis of peripheral blood cell immunity in Italian patients with different varieties of oral lichen planus. Bull Group Int Rech Sci Stomatol Odontol 39, 33–38 (1996).
    1. Popovska M, Grchevska L, Popovski V, Atanasovska-Stojanovska A, Minovska A, Belazelkoska Z, Radojkova-Nikolovska V, Mitik K, Strezovska S, T-cell subpopulations in lesions of oral lichen planus. Pril (Makedon Akad Nauk Umet Odd Med Nauki) 34, 143–149 (2013).
    1. Zhou XJ, Sugerman PB, Savage NW, Walsh LJ, Seymour GJ, Intra-epithelial CD8+ T cells and basement membrane disruption in oral lichen planus. J Oral Pathol Med 31, 23–27 (2002).
    1. Wang H, Zhang D, Han Q, Zhao X, Zeng X, Xu Y, Sun Z, Chen Q, Role of distinct CD4(+) T helper subset in pathogenesis of oral lichen planus. J Oral Pathol Med 45, 385–393 (2016).
    1. Ke Y, Dang E, Shen S, Zhang T, Qiao H, Chang Y, Liu Q, Wang G, Semaphorin4D Drives CD8(+) T-Cell Lesional Trafficking in Oral Lichen Planus via CXCL9/CXCL10 Upregulations in Oral Keratinocytes. J Invest Dermatol 137, 2396–2406 (2017).
    1. Burgdorf WH, Plewig G, Who described Civatte bodies? J Cutan Pathol 41, 340–346 (2014).
    1. Schroder K, Hertzog PJ, Ravasi T, Hume DA, Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol 75, 163–189 (2004).
    1. Patrick MT, Raja K, Miller K, Sotzen J, Gudjonsson JE, Elder JT, Tsoi LC, Drug Repurposing Prediction for Immune-Mediated Cutaneous Diseases using a Word Embedding-Based Machine Learning Approach. J Invest Dermatol, (2018).
    1. Dhillon S, Tofacitinib: A Review in Rheumatoid Arthritis. Drugs 77, 1987–2001 (2017).
    1. Okiyama N, Fujimoto M, Clinical perspectives and murine models of lichenoid tissue reaction/interface dermatitis. J Dermatol Sci 78, 167–172 (2015).
    1. Marshall A, Celentano A, Cirillo N, McCullough M, Porter S, Tissue-specific regulation of CXCL9/10/11 chemokines in keratinocytes: Implications for oral inflammatory disease. PLoS One 12, e0172821 (2017).
    1. Wu C, Xue Y, Wang P, Lin L, Liu Q, Li N, Xu J, Cao X, IFN-gamma primes macrophage activation by increasing phosphatase and tensin homolog via downregulation of miR-3473b. J Immunol 193, 3036–3044 (2014).
    1. Sikorski M, Bobek M, Zrubek H, Marcinkiewicz J, Dynamics of selected MHC class I and II molecule expression in the course of HPV positive CIN treatment with the use of human recombinant IFN-gamma. Acta Obstet Gynecol Scand 83, 299–307 (2004).
    1. Zhou F, Chen J, Zhao KN, Human papillomavirus 16-encoded E7 protein inhibits IFN-gamma-mediated MHC class I antigen presentation and CTL-induced lysis by blocking IRF-1 expression in mouse keratinocytes. J Gen Virol 94, 2504–2514 (2013).
    1. Lauffer F, Jargosch M, Krause L, Garzorz-Stark N, Franz R, Roenneberg S, Bohner A, Mueller NS, Theis FJ, Schmidt-Weber CB, Biedermann T, Eyerich S, Eyerich K, Type I Immune Response Induces Keratinocyte Necroptosis and Is Associated with Interface Dermatitis. J Invest Dermatol 138, 1785–1794 (2018).
    1. Liu Y, Liu G, Liu Q, Tan J, Hu X, Wang J, Wang Q, Wang X, The cellular character of liquefaction degeneration in oral lichen planus and the role of interferon gamma. J Oral Pathol Med 46, 1015–1022 (2017).
    1. Weber B, Schlapbach C, Stuck M, Simon HU, Borradori L, Beltraminelli H, Simon D, Distinct interferon-gamma and interleukin-9 expression in cutaneous and oral lichen planus. J Eur Acad Dermatol Venereol 31, 880–886 (2017).
    1. Al-Mohaya MA, Al-Otaibi L, Al-Harthi F, Al Bakr E, Arfin M, Al-Asmari A, Association of genetic polymorphisms in interferon-gamma, interleukin-6 and transforming growth factor-beta1 gene with oral lichen planus susceptibility. BMC Oral Health 16, 76 (2016).
    1. Eyerich K, Eyerich S, Immune response patterns in non-communicable inflammatory skin diseases. J Eur Acad Dermatol Venereol 32, 692–703 (2018).
    1. Ion DI, Setterfield JF, Oral Lichen Planus. Prim Dent J 5, 40–44 (2016).
    1. Unlu B, Tursen U, Autoimmune skin diseases and the metabolic syndrome. Clin Dermatol 36, 67–71 (2018).
    1. Luis-Montoya P, Yamamoto-Furusho JK, Vega-Memije E, Rodriguez-Carreon A, Ruiz-Morales JA, Vargas-Alarcon G, Dominguez-Soto L, Granados J, HLA-DRB1*0101 is associated with the genetic susceptibility to develop lichen planus in the Mexican Mestizo population. Arch Dermatol Res 299, 405–407 (2007).
    1. Farthing PM, Cruchley AT, Expression of MHC class II antigens (HLA DR, DP and DQ) by keratinocytes in oral lichen planus. J Oral Pathol Med 18, 305–309 (1989).
    1. Choi YS, Kim Y, Yoon HJ, Baek KJ, Alam J, Park HK, Choi Y, The presence of bacteria within tissue provides insights into the pathogenesis of oral lichen planus. Sci Rep 6, 29186 (2016).
    1. Yildirim B, Senguven B, Demir C, Prevalence of herpes simplex, Epstein Barr and human papilloma viruses in oral lichen planus. Med Oral Patol Oral Cir Bucal 16, e170–174 (2011).
    1. Baima B, Sticherling M, Apoptosis in different cutaneous manifestations of lupus erythematosus. Br J Dermatol 144, 958–966 (2001).
    1. French LE, Tschopp J, Fas-mediated cell death in toxic epidermal necrolysis and graft- versus-host disease: potential for therapeutic inhibition. Schweiz Med Wochenschr 130, 1656–1661 (2000).
    1. Saito N, Qiao H, Yanagi T, Shinkuma S, Nishimura K, Suto A, Fujita Y, Suzuki S, Nomura T, Nakamura H, Nagao K, Obuse C, Shimizu H, Abe R, An annexin A1-FPR1 interaction contributes to necroptosis of keratinocytes in severe cutaneous adverse drug reactions. Sci Transl Med 6, 245ra295 (2014).
    1. Sugerman PB, Satterwhite K, Bigby M, Autocytotoxic T-cell clones in lichen planus. Br J Dermatol 142, 449–456 (2000).
    1. Gueiros LA, Gondak R, Jorge Junior J, Coletta RD, Carvalho Ade A, Leao JC, de Almeida OP, Vargas PA, Increased number of Langerhans cells in oral lichen planus and oral lichenoid lesions. Oral Surg Oral Med Oral Pathol Oral Radiol 113, 661–666 (2012).
    1. Kastelan M, Prpic Massari L, Gruber F, Zamolo G, Zauhar G, Coklo M, Rukavina D, The role of perforin-mediated apoptosis in lichen planus lesions. Arch Dermatol Res 296, 226–230 (2004).
    1. Wenzel J, Peters B, Zahn S, Birth M, Hofmann K, Kusters D, Tomiuk S, Baron JM, Merk HF, Mauch C, Krieg T, Bieber T, Tuting T, Bosio A, Gene expression profiling of lichen planus reflects CXCL9+-mediated inflammation and distinguishes this disease from atopic dermatitis and psoriasis. J Invest Dermatol 128, 67–78 (2008).
    1. Wenzel J, Lucas S, Zahn S, Mikus S, Metze D, Stander S, von Stebut E, Hillen U, Bieber T, Tuting T, CXCR3 <-> ligand-mediated skin inflammation in cutaneous lichenoid graft-versus-host disease. J Am Acad Dermatol 58, 437–442 (2008).
    1. Brant JM, Aguiar MC, Grandinetti HA, Rodrigues LV, Vasconcelos AC, A comparative study of apoptosis in reticular and erosive oral lichen planus. Braz Dent J 23, 564–569 (2012).
    1. Tobon-Arroyave SI, Villegas-Acosta FA, Ruiz-Restrepo SM, Vieco-Duran B, Restrepo-Misas M, Londono-Lopez ML, Expression of caspase-3 and structural changes associated with apoptotic cell death of keratinocytes in oral lichen planus. Oral Dis 10, 173–178 (2004).
    1. Kumari S, Pasparakis M, Epithelial Cell Death and Inflammation in Skin. Curr Top Microbiol Immunol 403, 77–93 (2017).
    1. Zhou F, Molecular mechanisms of IFN-gamma to up-regulate MHC class I antigen processing and presentation. Int Rev Immunol 28, 239–260 (2009).
    1. Cinats A, Heck E, Robertson L, Janus Kinase Inhibitors: A Review of Their Emerging Applications in Dermatology. Skin Therapy Lett 23, 5–9 (2018).
    1. Mullard A, FDA approves Eli Lilly’s baricitinib. Nat Rev Drug Discov 17, 460 (2018).
    1. Smolen JS, Genovese MC, Takeuchi T, Hyslop DL, Macias WL, Rooney T, Chen L, Dickson CL, Riddle Camp J, Cardillo TE, Ishii T, Winthrop KL, Safety Profile of Baricitinib in Patients with Active Rheumatoid Arthritis with over 2 Years Median Time in Treatment. J Rheumatol, (2018).
    1. Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, Stone DL, Gao L, Klausmeier T, Foell D, de Jesus AA, Chapelle DC, Kim H, Dill S, Colbert RA, Failla L, Kost B, O’Brien M, Reynolds JC, Folio LR, Calvo KR, Paul SM, Weir N, Brofferio A, Soldatos A, Biancotto A, Cowen EW, Digiovanna JJ, Gadina M, Lipton AJ, Hadigan C, Holland SM, Fontana J, Alawad AS, Brown RJ, Rother KI, Heller T, Brooks KM, Kumar P, Brooks SR, Waldman M, Singh HK, Nickeleit V, Silk M, Prakash A, Janes JM, Ozen S, Wakim PG, Brogan PA, Macias WL, Goldbach-Mansky R, JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest 128, 3041–3052 (2018).
    1. Mucke J, Schneider M, Baricitinib for systemic lupus erythematosus. Lancet 392, 190–192 (2018).
    1. Papp KA, Menter MA, Raman M, Disch D, Schlichting DE, Gaich C, Macias W, Zhang X, Janes JM, A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor, in patients with moderate-to-severe psoriasis. Br J Dermatol 174, 1266–1276 (2016).
    1. Guttman-Yassky E, Silverberg JI, Nemoto O, Forman SB, Wilke A, Prescilla R, de la Pena A, Nunes FP, Janes J, Gamalo M, Donley D, Paik J, DeLozier AM, Nickoloff BJ, Simpson EL, Baricitinib in adult patients with moderate-to-severe atopic dermatitis: a phase 2 parallel, double-blinded, randomized placebo-controlled multiple-dose study. J Am Acad Dermatol, (2018).
    1. Jabbari A, Dai Z, Xing L, Cerise JE, Ramot Y, Berkun Y, Sanchez GA, Goldbach-Mansky R, Christiano AM, Clynes R, Zlotogorski A, Reversal of Alopecia Areata Following Treatment With the JAK1/2 Inhibitor Baricitinib. EBio Medicine 2, 351–355 (2015).
    1. Choi J, Cooper ML, Staser K, Ashami K, Vij KR, Wang B, Marsala L, Niswonger J, Ritchey J, Alahmari B, Achilefu S, Tsunoda I, Schroeder MA, DiPersio JF, Baricitinib-induced blockade of interferon gamma receptor and interleukin-6 receptor for the prevention and treatment of graft-versus-host disease. Leukemia, (2018).
    1. Davis AP, Grondin CJ, Lennon-Hopkins K, Saraceni-Richards C, Sciaky D, King BL, Wiegers TC, Mattingly CJ, The Comparative Toxicogenomics Database’s 10th year anniversary: update 2015. Nucleic Acids Res 43, D914–920 (2015).
    1. Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, Sajed T, Johnson D, Li C, Sayeeda Z, Assempour N, Iynkkaran I, Liu Y, Maciejewski A, Gale N, Wilson A, Chin L, Cummings R, Le D, Pon A, Knox C, Wilson M, DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res 46, D1074–D1082 (2018).
    1. Thorn CF, Klein TE, Altman RB, PharmGKB: the Pharmacogenomics Knowledge Base. Methods Mol Biol 1015, 311–320 (2013).
    1. Dickson MA, Hahn WC, Ino Y, Ronfard V, Wu JY, Weinberg RA, Louis DN, Li FP, Rheinwald JG, Human keratinocytes that express hTERT and also bypass a p16(INK4a)-enforced mechanism that limits life span become immortal yet retain normal growth and differentiation characteristics. Mol Cell Biol 20, 1436–1447 (2000).
    1. Sarkar MK, Hile GA, Tsoi LC, Xing X, Liu J, Liang Y, Berthier CC, Swindell WR, Patrick MT, Shao S, Tsou PS, Uppala R, Beamer MA, Srivastava A, Bielas SL, Harms PW, Getsios S, Elder JT, Voorhees JJ, Gudjonsson JE, Kahlenberg JM, Photosensitivity and type I IFN responses in cutaneous lupus are driven by epidermal-derived interferon kappa. Ann Rheum Dis, (2018).

Source: PubMed

3
Subskrybuj