Stem Cell Therapy—New Approach and Effective Treatment for Psoriasis
Keywords:
Psoriasis, stem cell therapy, auto-immune disease, Autoimmunity, Regenerative medicine, Immune modulationAbstract
The underlying mechanisms behind the development of psoriasis have become better understood in recent years. Key factors involved include the heightened reactivity of certain T-cell subsets (Th1 and Th17 cells), as well as dysregulation of regulatory T-cell (T-reg) function. Additionally, the complex interplay between immune cells, skin cells (keratinocytes), and the blood vessel endothelium has been shown to play an important role in the pathogenesis of psoriasis. Investigations have uncovered the impact that epidermal stem cells and their interactions with T cells have in psoriasis. Importantly, malfunctions across diverse stem cell types may constitute a central mechanism underlying the dysregulated inflammatory processes that characterize this condition. Nonetheless, a more thorough investigation of these mechanisms could present an opportunity to devise novel therapeutic approaches. In this publication, we reviewed the evidence regarding the role played by stem cells in the pathogenesis of psoriasis, as well as initial attempts at leveraging stem cells as a treatment modality.
References
Danielsen K, Olsen A, Wilsgaard T, Furberg AS. Is the prevalence of psoriasis increasing? A 30‐year follow‐up of a population‐based cohort. British Journal of Dermatology. 2013;168(6):1303–1310. DOI:10.1111/bjd.12230.
Parisi R, Symmons DP, Griffiths CE, Ashcroft DM. Global epidemiology of psoriasis: a systematic review of incidence and prevalence. Journal of Investigative Dermatology. 2013;133(2):377–385. DOI:10.1038/jid.2012.339
Singh S, Young P, Armstrong AW. An update on psoriasis and metabolic syndrome: a meta-analysis of observational studies. PloS one. 2017;12(7):e0181039. DOI:10.1371/journal.pone.0181039
Singh S, Taylor C, Kornmehl H, Armstrong AW. Psoriasis and suicidality: a systematic review and meta-analysis. Journal of the American Academy of Dermatology. 2017;77(3):425–440. e2. DOI:10.1016/j.jaad.2017.05.019
Randa H, Todberg T, Skov L, Larsen LS, Zachariae R. Health-related quality of life in children and adolescents with psoriasis: a systematic review and meta-analysis. Acta dermato-venereologica. 2017;97(5):555–563. DOI:10.2340/00015555-2600
Deng Y, Chang C, Lu Q. The inflammatory response in psoriasis: a comprehensive review. Clinical reviews in allergy & immunology. 2016;50:377–389. DOI:10.1007/s12016-016-8535-x
Martin DA, Towne JE, Kricorian G, et al. The emerging role of IL-17 in the pathogenesis of psoriasis: preclinical and clinical findings. Journal of Investigative Dermatology. 2013;133(1):17–26. DOI:10.1038/jid.2012.194
Kamstrup M, Faurschou A, Gniadecki R, Wulf HC. Epidermal stem cells-role in normal, wounded and pathological psoriatic and cancer skin. Current Stem Cell Research & Therapy. 2008;3(2):146–150. DOI:10.2174/157488808784223087
BERGSTRESSER PR, Richard Taylor J. Epidermal ‘turnover time’—a new examination. British Journal of Dermatology. 1977;96(5):503–506. DOI:10.1111/j.1365-2133.1977.tb07152.x
Zhang H, Hou W, Henrot L, et al. Modelling epidermis homoeostasis and psoriasis pathogenesis. Journal of The Royal Society Interface. 2015;12(103):20141071. DOI:10.1098/rsif.2014.1071
Jia H-Y, Shi Y, Luo L-F, et al. Asymmetric stem-cell division ensures sustained keratinocyte hyperproliferation in psoriatic skin lesions. International journal of molecular medicine. 2016;37(2):359–368. DOI:10.3892/ijmm.2015.2445
Franssen ME, Zeeuwen PL, Vierwinden G, van de Kerkhof PC, Schalkwijk J, Van Erp PE. Phenotypical and functional differences in germinative subpopulations derived from normal and psoriatic epidermis. Journal of Investigative Dermatology. 2005;124(2):373–383. DOI:10.1111/j.0022-202X.2004.23612.x
Watarai A, Amoh Y, Maejima H, Hamada Y, Katsuoka K. Nestin expression is increased in the suprabasal epidermal layer in psoriasis vulgaris. Acta dermato-venereologica. 2013;93(1)DOI:10.2340/00015555-1420
Nograles K, Zaba L, Guttman‐Yassky E, et al. Th17 cytokines interleukin (IL)‐17 and IL‐22 modulate distinct inflammatory and keratinocyte‐response pathways. British Journal of Dermatology. 2008;159(5):1092–1102. DOI:10.1111/j.1365-2133.2008.08769.x
Zhou C, Kuang M, Tao Y, et al. Nynrin preserves hematopoietic stem cell function by inhibiting the mitochondrial permeability transition pore opening. Cell Stem Cell. 2024;31(9):1359–1375. e8. DOI:10.1016/j.stem.2024.06.007
Saburi E, Abazari MF, Hassannia H, et al. The use of mesenchymal stem cells in the process of treatment and tissue regeneration after recovery in patients with Covid-19. Gene. 2021;777:145471. DOI:10.1016/j.gene.2021.145471
Velier M, Simoncini S, Abellan M, et al. Adipose-derived stem cells from systemic sclerosis patients maintain pro-angiogenic and antifibrotic paracrine effects in vitro. Journal of Clinical Medicine. 2019;8(11):1979. DOI:10.3390/jcm8111979
Ryan JM, Barry FP, Murphy JM, Mahon BP. Mesenchymal stem cells avoid allogeneic rejection. Journal of inflammation. 2005;2:1–11. DOI:10.1186/1476-9255-2-8
Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105(4):1815–1822. DOI:10.1182/blood-2004-04-1559
Hu MS, Borrelli MR, Lorenz HP, Longaker MT, Wan DC. Mesenchymal stromal cells and cutaneous wound healing: a comprehensive review of the background, role, and therapeutic potential. Stem cells international. 2018;2018DOI:10.1155/2018/6901983
Kozlowska U, Krawczenko A, Futoma K, et al. Similarities and differences between mesenchymal stem/progenitor cells derived from various human tissues. World journal of stem cells. 2019;11(6):347. DOI:10.4252/wjsc.v11.i6.347
Ribeiro A, Laranjeira P, Mendes S, et al. Mesenchymal stem cells from umbilical cord matrix, adipose tissue and bone marrow exhibit different capability to suppress peripheral blood B, natural killer and T cells. Stem cell research & therapy. 2013;4:1–16. DOI:10.1186/scrt336
Gao F, Chiu S, Motan D, et al. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell death & disease. 2016;7(1):e2062–e2062. DOI:10.1038/cddis.2015.327
Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends in pharmacological sciences. 2020;41(9):653–664. DOI:10.1016/j.tips.2020.06.009
An Y, Zhang Q, Ren Y, Yang S, Zhang Q. BML-111 Modulates and Alleviates p38/MAPK Signaling Pathway and Th1/Th2/Th17 Cytokine Response in Murine Psoriasis-Like Dermatitis. Discovery medicine. 2024;36(189):2026–2036. DOI:10.24976/Discov.Med.202436189.186
Gao H, Wang L, Lyu Y, et al. The P2X7R/NLRP3 inflammasome axis suppresses enthesis regeneration through inflammatory and metabolic macrophage-stem cell cross-talk. Science Advances. 2025;11(17):eadr4894. DOI:10.1126/sciadv.adr4894
Saburi E, Saburi A, Ghanei M. Promising role for Gc-MAF in cancer immunotherapy: from bench to bedside. Caspian Journal of Internal Medicine. 2017;8(4):228. DOI:10.22088/cjim.8.4.228
J Salgado A, L Reis R, Sousa N, M Gimble J. Adipose tissue derived stem cells secretome: soluble factors and their roles in regenerative medicine. Current Stem Cell Research & Therapy. 2010;5(2):103–110. DOI:10.2174/157488810791268564
Ferreira JR, Teixeira GQ, Santos SG, Barbosa MA, Almeida-Porada G, Gonçalves RM. Mesenchymal stromal cell secretome: influencing therapeutic potential by cellular pre-conditioning. Frontiers in immunology. 2018;9:2837. DOI:10.3389/fimmu.2018.02837
Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue engineering. 2001;7(2):211–228. DOI:10.1089/107632701300062859
Mizuno H, Tobita M, Uysal AC. Concise review: adipose‐derived stem cells as a novel tool for future regenerative medicine. Stem cells. 2012;30(5):804–810. DOI:10.1002/stem.1076
Schneider S, Unger M, Van Griensven M, Balmayor ER. Adipose-derived mesenchymal stem cells from liposuction and resected fat are feasible sources for regenerative medicine. European journal of medical research. 2017;22:1–11. DOI:10.1186/s40001-017-0258-9
Patrikoski M, Mannerström B, Miettinen S. Perspectives for clinical translation of adipose stromal/stem cells. Stem cells international. 2019;2019DOI:10.1155/2019/5858247
Izadpanah R, Kaushal D, Kriedt C, et al. Long-term in vitro expansion alters the biology of adult mesenchymal stem cells. Cancer research. 2008;68(11):4229–4238. DOI:10.1158/0008-5472.CAN-07-5272
Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem cells. 2006;24(5):1294–1301. DOI:10.1634/stemcells.2005-0342
Chen Y, Yu Q, Hu Y, Shi Y. Current research and use of mesenchymal stem cells in the therapy of autoimmune diseases. Current Stem Cell Research & Therapy. 2019;14(7):579–582. DOI:10.2174/1574888X14666190429141421
Abedi M, Alavi-Moghadam S, Payab M, et al. Mesenchymal stem cell as a novel approach to systemic sclerosis; current status and future perspectives. Cell Regeneration. 2020;9:1–19. DOI:10.1186/s13619-020-00058-0
Bateman ME, Strong AL, Gimble JM, Bunnell BA. Concise review: using fat to fight disease: a systematic review of nonhomologous adipose-derived stromal/stem cell therapies. Stem cells. 2018;36(9):1311–1328. DOI:10.1002/stem.2847
Álvaro-Gracia JM, Jover JA, García-Vicuña R, et al. Intravenous administration of expanded allogeneic adipose-derived mesenchymal stem cells in refractory rheumatoid arthritis (Cx611): results of a multicentre, dose escalation, randomised, single-blind, placebo-controlled phase Ib/IIa clinical trial. Annals of the rheumatic diseases. 2017;76(1):196–202. DOI:10.1136/annrheumdis-2015-208918
Zhou T, Li H-Y, Liao C, Lin W, Lin S. Clinical efficacy and safety of mesenchymal stem cells for systemic lupus erythematosus. Stem cells international. 2020;2020DOI:10.1155/2020/6518508
Wang L, Zhu C-y, Ma D-x, et al. Efficacy and safety of mesenchymal stromal cells for the prophylaxis of chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation: a meta-analysis of randomized controlled trials. Annals of Hematology. 2018;97:1941–1950. DOI:10.1007/s00277-018-3384-8
Lalu MM, McIntyre L, Pugliese C, et al. Safe ty of cell therapy with mesenchymal stromal cells (safecell): A systematic review and meta-analysis of clinical trials. PloS one. 2012;7(10):e47559. DOI:10.1371/journal.pone.0047559
Petrou P, Kassis I, Levin N, et al. Beneficial effects of autologous mesenchymal stem cell transplantation in active progressive multiple sclerosis. Brain. 2020;143(12):3574–3588. DOI:10.1093/brain/awaa333
Cao Y, Ding Z, Han C, Shi H, Cui L, Lin R. Efficacy of mesenchymal stromal cells for fistula treatment of Crohn’s disease: a systematic review and meta-analysis. Digestive diseases and sciences. 2017;62:851–860. DOI:10.1007/s10620-017-4453-x
William TT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation. 2003;75(3):389–397. DOI:10.1097/01.TP.0000045055.63901.A9
Beyth S, Borovsky Z, Mevorach D, et al. Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness. Blood. 2005;105(5):2214–2219. DOI:10.1182/blood-2004-07-2921
Nussbaum L, Chen Y, Ogg G. Role of regulatory T cells in psoriasis pathogenesis and treatment. British Journal of Dermatology. 2021;184(1):14–24. DOI:10.1111/bjd.19380
Cordiali-Fei P, Bianchi L, Bonifati C, et al. Immunologic biomarkers for clinical and therapeutic management of psoriasis. Mediators of Inflammation. 2014;2014DOI:10.1155/2014/236060
Diluvio L, Romiti M, Angelini F, et al. Infliximab therapy induces increased polyclonality of CD4+ CD25+ regulatory T cells in psoriasis. British Journal of Dermatology. 2010;162(4):895–897. DOI:10.1111/j.1365-2133.2010.09650.x
Ma HL, Napierata L, Stedman N, et al. Tumor necrosis factor α blockade exacerbates murine psoriasis‐like disease by enhancing Th17 function and decreasing expansion of Treg cells. Arthritis & Rheumatism: Official Journal of the American College of Rheumatology. 2010;62(2):430–440. DOI:10.1002/art.27203
Yang J, Zhu J, Lu S, Qin H, Zhou W. Transdermal psoriasis treatment inspired by tumor microenvironment-mediated immunomodulation and advanced by exosomal engineering. Journal of Controlled Release. 2025;382:113664. DOI:10.1016/j.jconrel.2025.113664
Shu M, Fan Z, Huang B, Wang C. Retrospective analysis of clinical features of pembrolizumab induced psoriasis. Investigational New Drugs. 2025:1–6. DOI:10.1007/s10637-025-01536-5
Zhao S, Sun W, Sun J, Peng L, Wang C. Clinical features, treatment, and outcomes of nivolumab induced psoriasis. Investigational New Drugs. 2025;43(1):42–49. DOI:10.1007/s10637-024-01494-4
Kashif S, Al-Mani SY, Almani NY. A Rare Psoriasis Drug Eruption Induced by Levetiracetam: A Case Study from Saudi Arabia. International Journal of Pharmacology. 2024;20(4):704–708. DOI:10.3923/ijp.2024.704.708
Atabati H, Esmaeili SA, Saburi E, et al. Probiotics with ameliorating effects on the severity of skin inflammation in psoriasis: Evidence from experimental and clinical studies. Journal of cellular physiology. 2020;235(12):8925–8937. DOI:10.1002/jcp.29737
Erichsen C, Jensen P, Kofoed K. Biologic therapies targeting the interleukin (IL)-23/IL-17 immune axis for the treatment of moderate-to-severe plaque psoriasis: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2020;34(1):30–38. DOI:10.1111/jdv.15879
Rosenzwajg M, Lorenzon R, Cacoub P, et al. Immunological and clinical effects of low-dose interleukin-2 across 11 autoimmune diseases in a single, open clinical trial. Annals of the rheumatic diseases. 2019;78(2):209–217. DOI:10.1136/annrheumdis-2018-214229
Bovenschen HJ, Van De Kerkhof PC, Van Erp PE, Woestenenk R, Joosten I, Koenen HJ. Foxp3+ regulatory T cells of psoriasis patients easily differentiate into IL-17A-producing cells and are found in lesional skin. Journal of Investigative Dermatology. 2011;131(9):1853–1860. DOI:10.1038/jid.2011.139
Bluestone JA, Tang Q. Treg cells—the next frontier of cell therapy. Science. 2018;362(6411):154–155. DOI:10.1126/science.aau2688
Adkins D, Abidi M, Brown R, et al. Resolution of psoriasis after allogeneic bone marrow transplantation for chronic myelogenous leukemia: late complications of therapy. Bone marrow transplantation. 2000;26(11):1239–1241. DOI:10.1038/sj.bmt.1702703
Snowden J, Heaton D. Development of psoriasis after syngeneic bone marrow transplant from psoriatic donor: further evidence for adoptive autoimmunity. British Journal of Dermatology. 1997;137(1):130–132. DOI:10.1046/j.1365-2133.1997.17821869.x
Zhang K, Li X, Yin G, Liu Y, Tang X. Functional characterization of T cells differentiated in vitro from bone marrow‐derived CD34+ cells of psoriatic patients with family history. Experimental Dermatology. 2010;19(8):e128–e135. DOI:10.1111/j.1600-0625.2009.01016.x
Yang L, Li B, Dang E, Jin L, Fan X, Wang G. Impaired function of regulatory T cells in patients with psoriasis is mediated by phosphorylation of STAT3. Journal of dermatological science. 2016;81(2):85–92. DOI:10.1016/j.jdermsci.2015.11.007
Terras S, Gambichler T, Moritz RK, Altmeyer P, Lambert J. Immunohistochemical analysis of FOXP 3+ regulatory T cells in healthy human skin and autoimmune dermatoses. International journal of dermatology. 2014;53(3):294–299. DOI:10.1111/j.1365-4632.2012.05808.x
Soler DC, Sugiyama H, Young AB, Massari JV, McCormick TS, Cooper KD. Psoriasis patients exhibit impairment of the high potency CCR5+ T regulatory cell subset. Clinical Immunology. 2013;149(1):111–118. DOI:10.1016/j.clim.2013.06.007
Zhang K, Li X, Yin G, Liu Y, Niu X, Hou R. Functional characterization of CD4+ CD25+ regulatory T cells differentiated in vitro from bone marrow‐derived haematopoietic cells of psoriasis patients with a family history of the disorder. British Journal of Dermatology. 2008;158(2):298–305. DOI:10.1111/j.1365-2133.2007.08359.x
Eedy D, Burrows D, Bridges J, Jones F. Clearance of severe psoriasis after allogenic bone marrow transplantation. BMJ: British Medical Journal. 1990;300(6729):908. DOI:10.1136/bmj.300.6729.908
Woods A, Mant M. Amelioration of severe psoriasis with psoriatic arthritis for 20 years after allogeneic haematopoietic stem cell transplantation. Annals of the rheumatic diseases. 2006;65(5):697–697. DOI:10.1136/ard.2005.039479
Masszi T, Farkas A, Remenyi P, et al. Ten-year remission of psoriasis after allogeneic but not autologous bone marrow transplantation. Dermatology. 2006;212(1):88. DOI:10.1159/000089032
Kaffenberger BH, Wong HK, Jarjour W, Andritsos LA. Remission of psoriasis after allogeneic, but not autologous, hematopoietic stem-cell transplantation. Journal of the American Academy of Dermatology. 2013;68(3):489–492. DOI:10.1016/j.jaad.2012.08.021
Jowitt S, Yin J. Psoriasis and bone marrow transplantation. BMJ: British Medical Journal. 1990;300(6736):1398. DOI:10.1136/bmj.300.6736.1398-c
Yin J, Jowitt S. Resolution of immune-mediated diseases following allogeneic bone marrow transplantation for leukaemia. Bone marrow transplantation. 1992;9(1):31–33.
Yokota A, Hukazawa M, Nakaseko C, et al. Resolution of psoriasis vulgaris following allogeneic bone marrow transplantation for aplastic anemia. [Rinsho Ketsueki] The Japanese Journal of Clinical Hematology. 1996;37(1):35–39.
Kishimoto Y, Yamamoto Y, Ito T, et al. Transfer of autoimmune thyroiditis and resolution of palmoplantar pustular psoriasis following allogeneic bone marrow transplantation. Bone marrow transplantation. 1997;19(10):1041–1043. DOI:10.1038/sj.bmt.1700789
Snowden JA, Kearney P, Kearney A, et al. Long‐term outcome of autoimmune disease following allogeneic bone marrow transplantation. Arthritis & Rheumatism: Official Journal of the American College of Rheumatology. 1998;41(3):453–459. DOI:10.1002/1529-0131(199803)41
Slavin S, Nagler A, Varadi G, Or R. Graft vs autoimmunity following allogeneic non-myeloablative blood stem cell transplantation in a patient with chronic myelogenous leukemia and severe systemic psoriasis and psoriatic polyarthritis. Experimental Hematology. 2000;28(7):853–857. DOI:10.1016/s0301-472x(00)00172-7
Chakrabarti S, Handa SK, Bryon RJ, Griffiths MJ, Milligan DW. Will mixed chimerism cure autoimmune diseases after a nonmyeloablative stem cell transplant? Transplantation. 2001;72(2):340–342. DOI:10.1097/00007890-200107270-00032
Kanamori H, Tanaka M, Kawaguchi H, et al. Resolution of psoriasis following allogeneic bone marrow transplantation for chronic myelogenous leukemia: case report and review of the literature. American journal of hematology. 2002;71(1):41–44. DOI:10.1002/ajh.10169
Kojima R, Kami M, Kim S, et al. Induction of graft-versus-autoimmune (GVA) disease effect against refractory psoriasis by complete donor-type chimerism and graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Bone marrow transplantation. 2003;32(4):439–442. DOI:10.1038/sj.bmt.1704146
Ciurea SO, Hansrivijit P, Ciurea AM, et al. Curative potential of hematopoietic stem cell transplantation for advanced psoriasis. American journal of hematology. 2019;94(6):E176–E180. DOI:10.1002/ajh.25470
Cooley HM, Snowden JA, Grigg AP, Wicks IP. Outcome of rheumatoid arthritis and psoriasis following autologous stem cell transplantation for hematologic malignancy. Arthritis & Rheumatism: Official Journal of the American College of Rheumatology. 1997;40(9):1712–1715. DOI:10.1002/art.1780400923
Mohren M, Daikeler T, Benz D, Günaydin I, Kanz L, Kötter I. Myeloablative immunosuppressive treatment with autologous haematopoietic stem cell transplantation in a patient with psoriatic arthropathy and monoclonal gammopathy of undetermined significance. Annals of the rheumatic diseases. 2004;63(4):466–467. DOI:10.1136/ard.2003.010702
Braiteh F, Hymes SR, Giralt SA, Jones R. Complete remission of psoriasis after autologous hematopoietic stem-cell transplantation for multiple myeloma. Journal of clinical oncology. 2008;26(27):4511–4513. DOI:10.1200/JCO.2008.17.6560
Held K, Rahmetulla R, Loew T, Radhi M. Complete resolution of guttate psoriasis following autologous SCT for Ewing’s sarcoma in a pediatric patient. Bone marrow transplantation. 2012;47(12):1585–1586. DOI:10.1038/bmt.2012.68
Sung SM, Kimball AB. Remission of psoriasis 13 years after autologous stem cell transplant. Cutis. 2015;95(6):E7–E8.
Azevedo A, Gonçalves C, Selores M, Torres T. Remission of psoriasis after autologous stem cell transplantation-until when? European Journal of Dermatology. 2017;27(1):74–75. DOI:10.1684/ejd.2016.2893
Chen W, Ren G, Zuo K, Huang X. Complete remission of both immunoglobulin light chain amyloidosis and psoriasis after autologous hematopoietic stem cell transplantation: A case report. Medicine. 2018;97(50):e13589. DOI:10.1097/MD.0000000000013589
Chen H, Niu J-W, Ning H-M, et al. Treatment of psoriasis with mesenchymal stem cells. The American journal of medicine. 2016;129(3):e13–e14. DOI:10.1016/j.amjmed.2015.11.001
De Jesus MM, Santiago JS, Trinidad CV, et al. Autologous adipose-derived mesenchymal stromal cells for the treatment of psoriasis vulgaris and psoriatic arthritis: a case report. Cell Transplantation. 2016;25(11):2063–2069. DOI:10.3727/096368916X691998
Wang SG, Hsu NC, Wang SM, Wang FN. Successful treatment of plaque psoriasis with allogeneic gingival mesenchymal stem cells: a case study. Case reports in dermatological medicine. 2020;2020DOI:10.1155/2020/4617520
Chen M, Peng J, Xie Q, et al. Mesenchymal stem cells alleviate moderate-to-severe psoriasis by reducing the production of type I interferon (IFN-I) by plasmacytoid dendritic cells (pDCs). Stem cells international. 2019;2019DOI:10.1155/2019/6961052
Downloads
Published
Issue
Section
License
Copyright (c) 2025 Hanan Hassan Ahmed, Mohamed J. Saadh, Radhwan Abdul Kareem, Suhas Ballal, Shilpa Sharma, J. Guntaj, G. V. Siva Prasad, Waam Mohammed Taher, Mariem Alwan, Mahmood Jasem Jawad, Atheer Khdyair Hamad

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
Dermatology Practical & Conceptual applies a Creative Commons Attribution License (CCAL) to all works we publish (http://creativecommons.org/licenses/by-nc/4.0/). Authors retain the copyright for their published work.

