Serum and Tissue CXCL9 Levels in Patients with Vitiligo Before and After Phototherapy: A Case-Control Study

Main Article Content

Marwa Abdallah
Samah Farag https://orcid.org/0000-0001-6775-5385
Ebtihal Hassan
Ahmed Elhawatky https://orcid.org/0000-0003-3454-8878

Keywords

vitiligo, chemokines, interferon gamma, CXCL9

Abstract

Background: Vitiligo is a common pigmentation disease that affects 1–2% of the global population. It is a genetic disease that is triggered by an environmental factor resulting in an autoimmune disease. The predominance of the T-helper 1 (Th1) pattern favors the development of vitiligo. Interferon-gamma (IFN-γ) is the most important cytokine that is associated with the Th1 immune response. IFN-γ induces the release of chemokine which is called CXCL9.


Objectives: Evaluation of the effects of narrow band ultraviolet B (nbUVB) phototherapy on the serum and tissue levels of CXCL9 among vitiligo patients.


Patients and methods: We recruited in our study two groups; one group of twenty patients complaining of nonsegmental vitiligo and the other is a control group composed of another age and sex-matched twenty healthy controls, we assessed the serum level of CXCL9 in all subjects before the study and the patient group was reassessed after treatment with nbUVB for 12 weeks. We also determined tissue level of CXCL9 in patients before and after the phototherapy for 12 weeks from suction blister fluid.


Results: We detected that serum levels of CXCL9 were higher in patients with vitiligo compared to healthy matched controls. nbUVB sessions for twelve weeks were done, and we found that serum and tissue levels of CXCL9 after treatment were decreased.


Conclusion: Serum and tissue CXCL9 can be used as a marker for disease activity and potentiality for the response to treatment. Therefore, targeting the inhibition of the Interferon-γ-chemokine axis may help in treating the disease activity.

References

1-Valle Y, Korobko I, Sigova J, et al. Patient-reported outcomes:A 5-year long study reveals previously unreported therapeutic, demographic, socio-economic, and other correlations in vitiligo. Dermatologic Therapy. 2018; 31:e12620.

2-Taïeb A, Meurant JM. Should we prioritise psychological interventions in the management of vitiligo? J Eur Acad Dermatol Venereol. 2018; 32(12):2053–4.

3-Ezzedine K, Eleftheriadou V, Whitton M, van Geel N. Vitiligo. Lancet. 2015; 386(9988): 74–84.

4-Kanwar AJ, Parsad D, De D. Mucosal involvement in vitiligo: a comprehensive review of 241 cases. J Eur Acad Dermatol Venereol. 2011; 1361-3.

5-Nagarajan A, Masthan MK, Sankar LS, Narayanasamy AB, Elumalai R. Oral manifestations of vitiligo. Indian J Dermatol. 2015; 60(1):103.

6- Harris J. E. Chemical-Induced Vitiligo. Dermatologic clinics. 2017; 35(2): 151–161.

7- Sheth VM, Guo Y, Qureshi AA. Comorbidities associated with vitiligo: a ten-year retrospective study. Dermatology. 2013; 227:311–5.

8- Gan EY, Cario-Andre M, Pain C, et al. Follicular vitiligo: a report of 8 cases. J Am Acad Dermatol. 2016; 74:1178–84.

9- Laberge G, Mailloux CM, Gowan K et al. Early disease onset and increased risk of other autoimmune diseases in familial generalized vitiligo. Pigment Cell Res. 2005; 18:300–305.

10- Chen YT, Chen YJ, Hwang CY, et al. Comorbidity profiles in association with vitiligo: a nationwide population-based study in Taiwan. J Eur Acad Dermatol Venereol. 2015; 29:1362–9.

11- Gill L, Zarbo A, Isedeh P, Jacobsen G, Lim HW, Hamzavi I. Comorbid autoimmune diseases in patients with vitiligo: a cross-sectional study. J Am Acad Dermatol. 2016; 74:295–302.

12- Spritz RA. The genetics of generalized vitiligo: autoimmune pathways and an inverse relationship with malignant melanoma. Genom Med. 2010; 2(10):78.

13- Sandoval-Cruz M., Garcia-Carrasco M., Sanchez-Porras R. et al.
Immunopathogenesis of vitiligo. Autoimmune Rev. 2011; 10(12): 762-765.

14- Laddha N.C., Dwivedi M., Begum R. Increased TNF-α and its Promoter Polymorphisms Correlate with Disease Progression and Higher Susceptibility towards Vitiligo. PLoS ONE. 2012; 7: e52298.

15- Boniface K, Jacquemin C, Darrigade AS, Dessarthe B et al. Vitiligo Skin Is Imprinted with Resident Memory CD8 T Cells Expressing CXCR3. J Invest Dermatol. 2018; 138(2):355-364.

16- Ezzedine S, El Rewiny EM, Abozied AA, Samna SM. Evaluation of Serum Interferon-gamma Level in Vitiligo Patients. The Egyptian Journal of Hospital Medicine. 2018; 73(10):7806-13.

17- Bulat V, Situm M, Dediol I, Ljubicic I, et al. The mechanisms of action of phototherapy in the treatment of the most common dermatoses. Coll Antropol. 2011; 35 (2): 147–151.

18- Dong D, Jiang M, Xu X, Guan M, et al. The effects of NB-UVB on the hair follicle-derived neural crest stem cells differentiating into melanocyte lineage in vitro. J Dermatol Sci. 2012; 66: 20–28.

19- Mélik-Parsadaniantz S, Rostène W. "Chemokines and neuromodulation". Journal of Neuroimmunology. 2008; 198 (1–2): 62–8.

20- Tokunaga R, Zhang W, Naseem M, Puccini A, Berger MD, Soni S, McSkane M, Baba H, Lenz HJ. "CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - A target for novel cancer therapy". Cancer Treatment Reviews. 2018; 63: 40–47.

21- Smit MJ, Verdijk P, van der Raaij-Helmer EM, et al. CXCR3- mediated chemotaxis of human T cells is regulated by a Gi- and phospholipase C-dependent pathway and not via activation of MEK/p44/p42 MAPK nor Akt/PI-3 kinase. Blood. 2003; 102:1959-65.

22- Muthuswamy R., Urban J., Lee J. J., Reinhart T. A., Bartlett D., and Kalinski P. Ability of mature dendritic cells to interact with regulatory T cells is imprinted during maturation. Cancer Res. 2008; 68:5972–5978.

23- Ikeda A., Aoki N., Kido M., Iwamoto S., Nishiura H., Maruoka R., et al. Progression of autoimmune hepatitis is mediated by IL‐18‐producing dendritic cells and hepatic CXCL9 expression in mice. Hepatology. 2014; 60:224–236.

24- Tworek D., Kuna P., Mlynarski W., Gorski P., Pietras T., and Antczak A. MIG (CXCL9), IP‐10 (CXCL10) and I‐TAC (CXCL11) concentrations after nasal allergen challenge in patients with allergic rhinitis. Arc. Med. Sci. 2013; 9:849–853.

25- Antonelli A., Ferrari S. M., Fallahi P., Frascerra S., Santini E., Franceschini S. S., et al. Monokine induced by interferon gamma (IFNgamma) (CXCL9) and IFNgamma inducible T‐cell alpha‐chemoattractant (CXCL11) involvement in Graves’ disease and ophthalmopathy: modulation by peroxisome proliferator‐activated receptor‐gamma agonists. J. Clin. Endocrinol. Metabol. 2009; 94:1803–1809.

26- Vandercappellen J., Van Damme J., and Struyf S. The role of CXC chemokines and their receptors in cancer. Cancer Lett. 2008; 267:226–244.

27- Zohar Y, Wildbaum G, Novak R, Salzman AL, Thelen M, Alon R, Barsheshet Y, Karp CL, Karin N. "CXCL11-dependent induction of FOXP3-negative regulatory T cells suppresses autoimmune encephalomyelitis". The Journal of Clinical Investigation. 2014; 124 (5): 2009–22.

28- Lacotte S, Brun S, Muller S, et al. CXCR3, inflammation, and - autoimmune diseases. Ann N Y Acad Sci. 2009; 1173:310-7.

29- Chen, SC., de Groot, M., Kinsley, D. et al. Expression of chemokine receptor CXCR3 by lymphocytes and plasmacytoid dendritic cells in human psoriatic lesions. Arch Dermatol Res. 2010; 302: 113–123.

30- Su R, Nguyen ML, Agarwal MR, et al. Interferon-inducible chemokines reflect severity and progression in sarcoidosis. Respir Res. 2013; 14:121.

31- Antonelli A, Ferrari SM, Giuggioli D, et al. Chemokine (C-X-C motif) ligand (CXCL)10 in autoimmune diseases. Autoimmun Rev. 2014; 13:272-80.

32- Ding Q., Xia Y., Ding S., Lu P., Sun L., and Liu M. An alternatively spliced variant of CXCR3 mediates the metastasis of CD133+ liver cancer cells induced by CXCL9. Oncotarget. 2016; 7:14405–14414.

33- Clancy-Thompson E., Perekslis T. J., Croteau W., Alexander M. P., Chabanet T. B., Turk M. J., et al. Melanoma induces, and adenosine suppresses, CXCR3‐Cognate chemokine production and T‐cell infiltration of lungs bearing metastatic‐like disease. Cancer Immunol. Res 2015; 3:956–967.

34- Lili Y, Yi W, Ji Y et al. Global activation of CD8+ cytotoxic T lymphocytes correlates with an impairment in regulatory T cells in patients with generalized vitiligo. PLoS One. 2012; 7:e37513.

35- van den Boorn JG, Konijnenberg D, Dellemijn TA, et al. Autoimmune destruction of skin melanocytes by perilesional T cells from vitiligo patients. J Invest Dermatol. 2009; 129(9):2220–32.

36- Yang L, Wei Y, Sun Y, Shi W, Yang J, Zhu L, Li M. Interferon-gamma Inhibits Melanogenesis and Induces Apoptosis in Melanocytes: A Pivotal Role of CD8+ Cytotoxic T Lymphocytes in Vitiligo. Acta Derm Venereol. 2015; 95(6):664-70.

37- Rashighi M., Agarwal P., Richmond J.M., Harris T.H. et al. CXCL10 is critical for the progression and maintenance of depigmentation in a mouse model of vitiligo. Sci Transl Med. 2014; 6: 223ra23.

38- Richmond JM, Bangari DS, Essien KI, Currimbhoy SD, Groom JR, Pandya AG, Youd ME, Luster AD, Harris JE. Keratinocyte-Derived Chemokines Orchestrate T-Cell Positioning in the Epidermis during Vitiligo and May Serve as Biomarkers of Disease. J Invest Dermatol. 2017; 137(2):350-358.

39- Wang X.X., Wang Q.Q., Wu J.Q., Jiang M., Chen L., Zhang C.F., and Xiang L.H. Increased expression of CXCR3 and its ligands in patients with vitiligo and CXCL10 as a potential clinical marker for vitiligo. Br J Dermatol. 2016; 174: 1318-1326.

40- Strassner JP, Rashighi M, Ahmed Refat M, et al. Suction blistering the lesional skin of vitiligo patients reveals useful biomarkers of disease activity. J Am Acad Dermatol. 2017; 76:847-55

41-Nagarajan A, Masthan MK, Sankar LS, Narayanasamy AB, Elumalai R. Oral manifestations of vitiligo. Indian J Dermatol. 2015; 60(1):103.

42- van Geel N, Wolkerstorfer A, Lommerts JE, et al. Validation study of the Vitiligo Extent Score-plus. J Am Acad Dermatol. 2018; 78(5):1013-1015.

43- Grimes, P. E., & Miller, M. M. Vitiligo: Patient stories, self-esteem, and the psychological burden of disease. International journal of women's dermatology. 2018; 4(1): 32–37.

44- Maouia A, Sormani L, Youssef M, et al. Differential expres-sion of CXCL9, CXCL10, and IFN-γ in vitiligo and alopecia areata patients. Pigment Cell Melanoma Res. 2017; 30:259-61

45- Yang L, Yang S, Lei J, et al. Role of chemokines and the corresponding receptors in vitiligo: a pilot study. J Dermatol. 2018; 45: 31‐ 38.

46- Bhardwaj S, Rani S, Srivastava N, Kumar R, Parsad D. Increased systemic and epidermal levels of IL-17A and IL-1β promotes progression of non-segmental vitiligo. Cytokine. 2017; 91():153-161.

47- Liu, L. Y., Strassner, J. P., Refat, M. A., Harris, J. E., & King, B. A. Repigmentation in vitiligo using the Janus kinase inhibitor tofacitinib may require concomitant light exposure. J Am Acad Dermatol. 2017; 77(4): 675–682.e1.

48- Chahar, Y. S., Singh, P. K., Sonkar, V. K., Rajani, I., & Adil, M. Impact on Quality of Life in Vitiligo Patients Treated with Narrowband Ultraviolet B Phototherapy. Indian Journal of Dermatology. 2018; 63(5): 399–402.

Similar Articles

You may also start an advanced similarity search for this article.