BMB Reports 2024; 57(11): 465-471  https://doi.org/10.5483/BMBRep.2024-0039
Stromal cells and epigenetics: emerging key players of chronic inflammatory skin diseases
Jihye Kim1,* & Michael Detmar2,*
1Department of Biopharmaceutical Engineering, Hannam University, Daejeon 34054, Korea, 2Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH Zurich), Zurich 8093, Switzerland
Correspondence to:

Michael Detmar, Tel: +41-44-633-7361; Fax: +41-44-633-1364; E-mail: michael.detmar@pharma.ethz.ch; Jihye Kim, Tel: +82-42-629-8828; Fax: +82-42-629-8708; E-mail: jihye.kim@hnu.kr

Received: January 31, 2024; Revised: March 28, 2024; Accepted: May 21, 2024; Published online: August 8, 2024.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
Epigenetic alterations play a crucial role in developmental processes, tissue regeneration, and cellular differentiation. Epigenetic changes are dynamically reversible. Various drugs that target DNA methyltransferases or histone deacetylases have demonstrated their ability to restore normal epigenetic patterns in a number of diseases. While the involvement of epigenetic modifications has been identified in chronic inflammatory diseases, their specific impact on skin inflammation in stromal cells remains unclear. This mini-review explores the role of stromal cells in chronic inflammatory skin diseases, focusing on epigenetic modifications of stromal cells such as fibroblasts, lymphatic, and blood vascular endothelial cells in both healthy and diseased skin. We also provide an overview of recent findings that highlight the contribution of stromal cells, including fibroblasts, to inflammatory and remodeling processes through epigenetic changes in the context of chronic inflammatory conditions. Investigating epigenetic reprogramming of stromal cells might lead to novel strategies for treating chronic inflammatory skin diseases.
Keywords: Chronic inflammation, Endothelial cells, Epigenetics, Fibroblasts
INTRODUCTION

Stromal cells are a heterogeneous and indispensable group of cells that provide structural and functional support for various tissues and organs throughout the body. Beyond their conventional structural responsibilities, they play a pivotal role in maintaining the integrity and function of their resident tissues (1). For example, stromal cells can produce and organize extracellular matrix (ECM), a complex network of proteins and carbohydrates crucial for endowing tissues with mechanical resilience and architectural framework (2). Stromal cells can interact with neighboring cells, including epithelial cells and immune cells, thereby contributing to the maintenance of tissue or organ homeostasis (3). Importantly, stromal cells also play a role in various physiological/pathological processes, including inflammation (4-6), wound healing (7), and cancer progression (8). Thus, a comprehensive understanding of the regulation of stromal cell function in both homeostasis and dysfunction is crucial for a better understanding of skin physiology and disease pathogenesis.

Epigenetic changes refer to modifications that affect how genes are turned on/off without changing the genetic code itself. One of the significant features of epigenetic changes is their potential for reversibility. This has implications for therapeutic interventions and strategies to modify gene expression patterns to treat diseases. Thus, drugs targeting epigenetic modifications have been developed and considered as new potential therapeutic tools to ameliorate various diseases, including cancer (9-11) and chronic inflammatory diseases (6, 12-14). The field of epigenetics has grown rapidly in recent years. Ongoing research continues to uncover complexities of epigenetic regulation and mechanisms underlying various biological processes and diseases.

Recently, several studies have discovered the important role of stromal cells in pathological conditions such as cancers and chronic inflammatory diseases that could be regulated by epigenetic modifications (14-16). For example, endothelial cells from pulmonary arterial hypertension show extensive remodeling at active enhancers, while very few transcriptomic changes are observed, indicating an important role of epigenetic changes in disease-relevant stromal cells (15). Thus, it is of great interest to investigate epigenetic changes of stromal cells and their impacts on pathological states that might be reversed by epigenetic reprogramming.

In this mini-review, we will cover the role of stromal cells and epigenetic changes of stromal cells in both healthy skin and chronic inflammatory skin diseases.

ROLE OF STROMAL CELLS IN THE SKIN

Stromal cells in the skin mainly consist of fibroblasts, lymphatic endothelial cells, and blood vascular endothelial cells. The term “stromal” originates from the Greek word stroma, meaning “bed covering” that refers to a structural or connective role of a tissue or organ. In the skin, stromal cells are positioned in the dermal layer below the epidermis release growth factors that can stimulate cellular division. This process ensures continuous regeneration of the epidermis from its basal layer, while the uppermost skin cells are consistently shedding from the body. The turnover time of healthy human skin is approximately 28 days (17). Furthermore, stromal cells contribute to inflammatory responses and regulate the quantity of cells accumulating in the inflamed region of a tissue (18). For example, extracellular matrix components such as collagen, laminin, and fibronectin derived from stromal cells undergo remodeling in response to external stimuli or during pathological processes (19-22). These stromal cells in the dermis consist of various cell types, including fibroblasts and endothelial cells of lymphatic/blood vessels. Especially fibroblasts, which are among key stromal cell types in the dermis, play an essential role in shaping the microenvironment by synthesizing and remodeling ECM proteins and by secreting cytokines, chemokines, and growth factors (23-26). Stromal cells can impact the homeostasis of adjacent cells such as immune cells through cell-cell communications. They can also modulate inflammatory infiltration (22).

CONTRIBUTION OF STROMAL CELLS TO CHRONIC INFLAMMATORY SKIN DISEASE

Many studies have reported that not only immune cells and epidermal cells, but also stromal cells in the skin, such as dermal fibroblasts and endothelial cells, contribute to the pathogenesis of chronic inflammatory skin diseases such as atopic dermatitis and psoriasis (4, 5, 27, 28). Atopic dermatitis is a Th2 mediated chronic inflammatory disease. In atopic dermatitis, dermal fibroblasts interact with immune cells, triggering a substantial cytokine/chemokine response to NOD2/TLR2 ligands (29). Endothelial cells are highly involved in the pathogenesis of AD since immune cells actively communicate with endothelial cells by releasing and stimulating inflammatory mediators. T cell-endothelial cell interactions are important for establishing acute AD, suggesting that they are pivotal targets for treating AD (30). Psoriasis is a Th17 mediated chronic inflammatory skin disease. In psoriasis, activated fibroblasts can enhance local T-cell persistence (29, 31), alter the expression of ECM proteins (27, 32), and stimulate endothelial cells (33). Activated vascular endothelium then up-regulates adhesion molecules and cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), IL-6, and IL-17 families. Adhesion molecules (e.g., VCAM-1) are important for coordinating the process of leukocyte rolling, attachment, and migration into the skin (34-36). Recently, single-cell analysis has been used to comprehensively characterize specific cell types in atopic dermatitis and psoriasis (Fig. 1). For example, He et al. have identified a COL6A5/18A1-positive fibroblast subset that can release cytokines such as CCL2, CCL19, and IL-32 important for AD pathogenesis in AD lesional skin (30). In psoriasis, recent studies have revealed the heterogeneity of fibroblast subpopulation (SFRP2+), showing transition from a profibrotic state to a pro-inflammatory state in psoriatic skin, including producers of ECM components and cytokine-induced, non-matrix producing, pro-inflammatory compartments (38). In addition, He et al. (30) have discovered that each endothelial cell subtype shows psoriasis-specific interactions between cis-regulatory enhancers and promoters which reveal dysregulated gene regulatory networks in psoriasis. These results suggest that specific transcriptional responses and epigenetic signatures of endothelial cells lining different vessel compartments are involved in psoriasis (6).

EPIGENETIC MODICIFATIONS IN THE SKIN

Epigenetics is the study of changes in gene expression or cellular phenotype in the absence of changes in underlying DNA sequences. While DNA contains instructions for building and maintaining the body, epigenetic modifications influence how these instructions are interpreted and executed. Changes in DNA accessibility by epigenetic modification can be achieved through various mechanisms such as DNA modification, histone modification, and non-coding RNA (39, 40). DNA undergoes modifications through the addition of methyl groups and consequently influences gene expression (DNA modification). Hypermethylation results in gene silencing, whereas hypomethylation promotes active transcription (41). To study these modifications, various high-throughput techniques are commonly used to profile DNA methylation, including whole-genome bisulfite sequencing and Infinium human methylation bead-chip arrays (42) which also have been used to study skin diseases such as psoriasis (43-45). Another epigenetic process that governs chromatin structure and regulates gene expression is histone modification. Histone modifications alter chromatin packaging, determine its accessibility (open) or inaccessibility (closed), thereby influencing gene expression such as activation or repression (39, 46). To study histone modifications, chromatin immunoprecipitation sequencing to analyze histone-DNA interactions and assay for transposase-accessible chromatin (ATAC) sequencing to identify open areas in chromatin packaging are widely performed (47). Epigenetic modifications affect various processes in the skin, such as epidermal homeostasis and differentiation, development, and responses to environmental stimuli (48-52). In chronic inflammatory skin diseases, several histone methylation/acetylation marks have been recently reported in psoriasis, atopic dermatitis, and systemic lupus erythematosus (Table 1). Since epigenetic modifications are modifiable and reversible, epigenetic modifier drugs inhibiting histone acetyltransferases (HATs), histone deacetylases (HDACs), and DNA methyltransferases (DNMTs) that can reverse epigenetic signatures have been drawing attention as a new potential strategy for treating diseases (Fig. 2) (9, 10). There are currently 6 FDA-approved epigenetic modifying drugs, and more than 50 epigenetic drug candidates are being tested in preclinical and clinical trials (53). For examples, trichostatin A, an HDAC inhibitor, ameliorates the development of atopic-like dermatitis by increasing T regs and reducing IL-4 production (54). A485, a HAT inhibitor, has been reported to reduce psoriasis-like skin inflammation in vivo (14). Therefore, understanding epigenetic mechanisms in the skin might open avenues for developing novel therapeutic targets and enhancing comprehension of skin biology.

EPIGENETIC CHANGES OF STROMAL CELLS IN CHRONIC INFLAMMATORY DISEASES

Epigenetic changes play crucial roles not only in many biological processes, but also in various diseases, including cancer, chronic inflammatory diseases, and certain genetic disorders (13, 16, 55, 56). Many studies have focused on epigenetic changes of immune cells, keratinocytes, and whole skin tissues in chronic inflammatory diseases such as atopic dermatitis and psoriasis (43, 44, 57-60). However, several studies have recently focused on stromal cells, including fibroblasts and endothelial cells, undergoing epigenetic modifications that contribute to chronic inflammatory conditions in the skin. For example, a recent study has identified psoriatic endothelial cell subtype-specific cis-regulatory element connections (CREs) by 5’ single-cell profiling, providing insights into specific transcriptional responses and epigenetic signatures in psoriasis (6). In this study, MCAM and its binding partners specifically showed upregulation in blood capillaries (CD31+RGCC+ population) in psoriasis, together with vascular remodeling. There was a unique pairing of CREs in psoriasis, suggesting a potent contribution by transcriptomic and epigenetic repatterning of the psoriatic vasculature. Another study has suggested that upregulation of histone deacetylase-1 (HDAC-1) in psoriatic endothelial cells might potentially contribute to overexpression of VEGF that induces proliferation of endothelial cells (61). In addition, Kwon et al. have recently investigated interactions between mast cells, keratinocytes, and fibroblasts in an atopic dermatitis model, indicating a regulative role of the HDAC6-CXCL13 axis in the pathogenesis of atopic dermatitis (62). These results on transcriptional and epigenetic changes that underlie responses of stromal cells in chronic inflammatory skin diseases can suggest new therapeutic strategies to overcome limitations of current treatments, including relapse and treatment resistance.

CONCLUSION

Here, we summarized stromal cells and their epigenetic changes known to play a significant role in both healthy skin and chronic inflammatory skin diseases, focusing on atopic dermatitis and psoriasis. Interestingly, recent studies have highlighted disease-specific cell subtypes and their interactions across cell types in healthy and diseased states in the skin by single-cell transcriptomic profiling (6, 31, 38, 63). Single-cell analysis compared to conventional bulk transcriptomics provides a more detailed characterization of various cell types such as stromal and immune cells (64-66), thus uncovering the subtype of each cell that might contribute to transcriptional and epigenetic signatures underlying chronic inflammatory skin diseases. For epigenetic changes, histone deacetylases (HDACs) can mediate the balance of acetylation/deacetylation together with histone acetyltransferases (HATs). Several studies have shown that aberrant expression of HDACs and HATs in chronic inflammatory skin diseases such as atopic dermatitis and psoriasis (59, 61, 67) might serve as a potential therapeutic target for treatment (68, 69). Therefore, multi-omics profiling, analyzing patterns of transcriptional and epigenetic changes, and their reprogramming via modifier drugs might potentially lead to clinical approaches to improve patient outcomes.

ACKNOWLEDGEMENTS

This work was supported by 2023 Hannam University Research Fund.

CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURES
Fig. 1. Heterogeneity of stromal cells in chronic inflammatory disease. Subpopulations of stromal cells, including fibroblasts, blood vascular endothelial cells (BECs), and lymphatic endothelial cells (LECs), identified from single-cell transcriptomics, display specific molecular repatterning, potentially leading to activated states. Activated stromal cells can trigger cytokine/chemokine responses, enhance T-cell persistence, and alter extracellular matrix (ECM) components relevant to the pathogenesis of chronic inflammatory skin diseases.
Fig. 2. Flexibility of epigenetic signatures by epigenetic reprogramming. DNA can undergo methylation at cytosine residues in a CpG context (DNA methylation). The DNA wraps around histone proteins to form structures called nucleosomes. Modifications to these histone proteins represent another layer of epigenetic regulations (histone acetylation/methylation). These modifications dictate whether the chromatin structure is open and accessible or closed and inaccessible. Acetylation of histone tails by histone acetyltransferase (HAT) activates genes, whereas histone deacetylation by deacetylases (HDACs) leads to gene silencing. DNA methyltransferase (DNMTs) adds methyl groups to the 5-position of cytosine residues in DNA, crucial for the process of DNA methylation, playing a significant role in maintaining genome stability.
TABLE

Key epigenetic modifications reported in chronic inflammatory skin diseases in recent years

Disease Modification Site Cell types Ref.
Psoriasis Histone methylation H3K9me3 Keratinocytes Chen C et al. (70)
H3K4me3 Keratinocytes Huang S et al. (71)
H3K4me1
H3K27me3 Keratinocytes Zhang T et al. (72)
Histone acetylation H3K27ac Skin tissue Masalha M et al. (73)
J Kim et al. (14)
H3K27ac CD4+ T cells Xia X et al. (74)
H3K9ac CD4+ T cells Xia X et al. (74)
Atopic dermatitis Histone acetylation H3K9ac Keratinocytes Traisaeng S et al. (75)
Systemic lupus erythematosus Histone methylation H3K4me1 CD4+ T cells Zhao M et al. (76)
H3K9me3 CD4+ T cells Zhao M et al. (77)
H3K27me3 CD4+ T cells Tsou PS et al. (78)
Histone acetylation H3K27ac CD4+ T cells Zhao M et al. (76)

Recent studies have reported histone modifications at specific sites in different cell types associated with psoriasis, atopic dermatitis, and systemic lupus erythematosus.


REFERENCES
  1. Manetti M (2021) Molecular morphology and function of stromal cells. Int J Mol Sci 22, 13422.
    Pubmed KoreaMed CrossRef
  2. Novoseletskaya E, Grigorieva O and Nimiritsky P et al (2020) Mesenchymal stromal cell-produced components of extracellular matrix potentiate multipotent stem cell response to differentiation stimuli. Front Cell Dev Biol 8, 555378.
    Pubmed KoreaMed CrossRef
  3. Mueller SN and Germain RN (2009) Stromal cell contributions to the homeostasis and functionality of the immune system. Nat Rev Immunol 9, 618-629.
    Pubmed KoreaMed CrossRef
  4. Nunomura S, Ejiri N and Kitajima M et al (2019) Establishment of a mouse model of atopic dermatitis by deleting Ikk2 in dermal fibroblasts. J Invest Dermatol 139, 1274-1283.
    Pubmed CrossRef
  5. Löwa A, Graff P, Kaessmeyer S and Hedtrich S (2020) Fibroblasts from atopic dermatitis patients trigger inflammatory processes and hyperproliferation in human skin equivalents. J Eur Acad Dermatol 34, e262-e265.
    Pubmed CrossRef
  6. He Y, Kim J and Tacconi C et al (2022) Mediators of capillary-to-venule convertsion in the chronic inflammatory skin disease psoriasis. J Invest Dermatol 142, 3313-3326.
    Pubmed CrossRef
  7. Rodrigues M, Kosaric N, Bonham CA and Gurtner GC (2019) Wound healing: a cellular perspective. Physiol Rev 99, 665-706.
    Pubmed KoreaMed CrossRef
  8. Lee H, Jeong AJ and Ye SK (2019) Highlighted STAT3 as a potential drug target for cancer therapy. BMB Rep 52, 415-423.
    Pubmed KoreaMed CrossRef
  9. Andreol F, Barbosa AJM, Parenti MD and Rio AD (2013) Modulation of epigenetic targets for anticancer therapy: clinicopathological relevance, structural data and drug discovery perspectives. Curr Pharm Design 19, 578-613.
    Pubmed KoreaMed CrossRef
  10. Fardi M, Solali S and Hagh MF (2018) Epigenetic mechanisms as a new approach in cancer treatment: an updated review. Genes Dis 5, 304-311.
    Pubmed KoreaMed CrossRef
  11. Akar RO, Selvİ S, Ulukaya E and Aztopal N (2019) Key actors in cancer therapy: epigenetic modifiers. Turk J Biol 43, 155-170.
    Pubmed KoreaMed CrossRef
  12. Shuto T, Furuta T and Oba M et al (2006) Promoter hypomethylation of Toll-like receptor-2 gene is associated with increased proinflammatory response toward bacterial peptidoglycan in cystic fibrosis bronchial epithelial cells. FASEB J: Off Publ Fed Am Soc Exp Biol 20, 782-784.
    Pubmed CrossRef
  13. Zhang S, Meng Y and Zhou L et al (2022) Targeting epigenetic regulators for inflammation: mechanisms and intervention therapy. MedComm 3, e173.
    Pubmed KoreaMed CrossRef
  14. Kim J, He Y and Tormen S et al (2022) The p300/CBP inhibitor A485 normalizes psoriatic fibroblast gene expression in vitro and reduces psoriasis-like skin inflammation in vivo. J Invest Dermatol 143, 431-443.
    Pubmed CrossRef
  15. Reyes-Palomares A, Gu M and Grubert F et al (2020) Remodeling of active endothelial enhancers is associated with aberrant gene-regulatory networks in pulmonary arterial hypertension. Nat Commun 11, 1673.
    Pubmed KoreaMed CrossRef DOAJ
  16. Ciesielski O, Biesiekierska M and Panthu B et al (2020) The epigenetic profile of tumor endothelial cells. Effects of combined therapy with antiangiogenic and epigenetic drugs on cancer progression. Int J Mol Sci 21, 2606.
    Pubmed KoreaMed CrossRef
  17. Shimizu H (2016) Structure and functions of the skin Shimizu's Dermatology 2nd, 1-42.
    Self
  18. Buckley CD, Barone F, Nayar S, Bénézech C and Caamaño J (2015) Stromal cells in chronic inflammation and tertiary lymphoid organ formation. Annu Rev Immunol 33, 715-745.
    Pubmed CrossRef
  19. Villa A, Trachsel E and Kaspar M et al (2008) A high-affinity human monoclonal antibody specific to the alternatively spliced EDA domain of fibronectin efficiently targets tumor neo-vasculature in vivo. Int J Cancer 122, 2405-2413.
    Pubmed CrossRef
  20. Kelsh-Lasher RM, Ambesi A, Bertram C and McKeown-Longo PJ (2017) Integrin α4β1 and TLR4 cooperate to induce fibrotic gene expression in response to fibronectin's EDA domain. J Invest Dermatol 137, 2505-2512.
    Pubmed KoreaMed CrossRef
  21. Kinchen J, Chen HH and Parikh K et al (2018) Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell 175, 372-386.
    Pubmed KoreaMed CrossRef
  22. Bhattacharjee O, Ayyangar U, Kurbet AS, Ashok D and Raghavan S (2019) Unraveling the ECM-immune cell crosstalk in skin diseases. Front Cell Dev Biol 7, 68.
    Pubmed KoreaMed CrossRef
  23. Smith RS, Smith TJ, Blieden TM and Phipps RP (1997) Fibroblasts as sentinel cells. Synthesis of chemokines and regulation of inflammation. Am J Pathol 151, 317-322.
    Pubmed
  24. Georganas C, Liu H and Perlman H et al (2000) Regulation of IL-6 and IL-8 expression in rheumatoid arthritis synovial fibroblasts: the dominant role for NF-κB but not C/EBPβ or c-Jun. J Immunol 165, 7199-7206.
    Pubmed CrossRef
  25. Distler JH, Jüngel A and Huber LC et al (2005) The induction of matrix metalloproteinase and cytokine expression in synovial fibroblasts stimulated with immune cell microparticles. Proc Natl Acad Sci U S A 102, 2892-2897.
    Pubmed KoreaMed CrossRef
  26. Abonnenc M, Nabeebaccus AA and Mayr U et al (2013) Extracellular matrix secretion by cardiac fibroblasts. Circ Res 113, 1138-1147.
    Pubmed CrossRef
  27. Gubán B, Vas K and Balog Z et al (2016) Abnormal regulation of fibronectin production by fibroblasts in psoriasis. Brit J Dermatol 174, 533-541.
    Pubmed CrossRef
  28. Arasa J, Terencio MC and Andrés RM et al (2019) Defective induction of COX-2 expression by psoriatic fibroblasts promotes pro-inflammatory activation of macrophages. Front Immunol 10, 536.
    Pubmed KoreaMed CrossRef
  29. Jiao D, Wong CK and Qiu HN et al (2016) NOD2 and TLR2 ligands trigger the activation of basophils and eosinophils by interacting with dermal fibroblasts in atopic dermatitis-like skin inflammation. Cell Mol Immunol 13, 535-550.
    Pubmed KoreaMed CrossRef
  30. He H, Suryawanshi H and Morozov P et al (2020) Single-cell transcriptome analysis of human skin identifies novel fibroblast subpopulation and enrichment of immune subsets in atopic dermatitis. J Allergy Clin Immun 145, 1615-1628.
    Pubmed CrossRef
  31. Linthout SV, Miteva K and Tschope C (2014) Crosstalk between fibroblasts and inflammatory cells. Cardiovasc Res 102, 258-269.
    Pubmed CrossRef
  32. Coentro JQ, Pugliese E, Hanley G, Raghunath M and Zeugolis DI (2019) Current and upcoming therapies to modulate skin scarring and fibrosis. Adv Drug Deliver Rev 146, 37-59.
    Pubmed CrossRef
  33. Cappellari GG, Barazzoni R, Cattin L, Muro AF and Zanetti M (2016) Lack of fibronectin extra domain A alternative splicing exacerbates endothelial dysfunction in diabetes. Sci Rep-uk 6, 37965.
    Pubmed KoreaMed CrossRef
  34. Sprague AH and Khalil RA (2009) Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem Pharmacol 78, 539-552.
    Pubmed KoreaMed CrossRef
  35. von Stebut E, Boehncke WH and Ghoreschi K et al (2020) IL-17A in psoriasis and beyond: cardiovascular and metabolic implications. Front Immunol 10, 3096.
    Pubmed KoreaMed CrossRef
  36. Anyfanti P, Margouta A and Goulas K et al (2022) Endothelial dysfunction in psoriasis: an updated review. Front Med 9, 864185.
    Pubmed KoreaMed CrossRef
  37. Steinhoff M, Steinhoff A, Homey B, Luger TA and Schneider SW (2006) Role of vasculature in atopic dermatitis. J Allergy Clin Immunol 118, 190-197.
    Pubmed CrossRef
  38. Ma F, Plazyo O and Billi AC et al (2023) Single cell and spatial sequencing define processes by which keratinocytes and fibroblasts amplify inflammatory responses in psoriasis. Nat Commun 14, 3455.
    Pubmed KoreaMed CrossRef
  39. Hanna CW, Demond H and Kelsey G (2018) Epigenetic regulation in development: is the mouse a good model for the human?. Hum Reprod Update 24, 556-576.
    Pubmed KoreaMed CrossRef
  40. Pinborg A, Loft A and Romundstad LB et al (2016) Epigenetics and assisted reproductive technologies. Acta Obstet Gyn Scan 95, 10-15.
    Pubmed CrossRef
  41. Jabbari K and Bernardi G (2004) Cytosine methylation and CpG, TpG (CpA) and TpA frequencies. Gene 333, 143-149.
    Pubmed CrossRef
  42. Liu H, Li S and Wang X et al (2016) DNA methylation dynamics: identification and functional annotation. Brief Funct Genomics 15, 470-484.
    Pubmed CrossRef
  43. Zhang P, Su Y, Chen H, Zhao M and Lu Q (2010) Abnormal DNA methylation in skin lesions and PBMCs of patients with psoriasis vulgaris. J Dermatol Sci 60, 40-42.
    Pubmed CrossRef
  44. Zhou F, Wang W and Shen C et al (2016) Epigenome-wide association analysis identified nine skin dna methylation loci for psoriasis. J Investig Dermatol 136, 779-787.
    Pubmed CrossRef
  45. Gu X, Nylander E, Coates PJ, Fahraeus R and Nylander K (2015) Correlation between reversal of DNA methylation and clinical symptoms in psoriatic epidermis following narrow-band UVB phototherapy. J Invest Dermatol 135, 2077-2083.
    Pubmed KoreaMed CrossRef
  46. Ovejero-Benito MC, Reolid A and Sánchez-Jiménez P et al (2018) Histone modifications associated with biological drug response in moderate-to-severe psoriasis. Exp Dermatol 27, 1361-1371.
    Pubmed CrossRef
  47. Ma S and Zhang Y (2020) Profiling chromatin regulatory landscape: insights into the development of ChIP-seq and ATAC-seq. Mol Biomed 1, 9.
    Pubmed KoreaMed CrossRef
  48. Li J, Jiang TX and Hughes MW et al (2012) Progressive alopecia reveals decreasing stem cell activation probability during aging of mice with epidermal deletion of DNA methyltransferase 1. J Investig Dermatol 132, 2681-2690.
    Pubmed KoreaMed CrossRef
  49. Sen GL, Reuter JA, Webster DE, Zhu L and Khavari PA (2010) DNMT1 maintains progenitor function in self-renewing somatic tissue. Nature 463, 563-567.
    Pubmed KoreaMed CrossRef
  50. Ezhkova E, Pasolli HA and Parker JS et al (2009) Ezh2 orchestrates gene expression for the stepwise differentiation of tissue-specific stem cells. Cell 136, 1122-1135.
    Pubmed KoreaMed CrossRef
  51. Sada A, Jacob F and Leung E et al (2016) Defining the cellular lineage hierarchy in the interfollicular epidermis of adult skin. Nat Cell Biol 18, 619-631.
    Pubmed KoreaMed CrossRef
  52. Paquet-Fifield S, Schlüter H and Li A et al (2009) A role for pericytes as microenvironmental regulators of human skin tissue regeneration. J Clin Investig 119, 2795-2806.
    Pubmed KoreaMed CrossRef
  53. Akar RO, Selvi S, Ulukaya E and Aztopal N (2019) Key actors in cancer therapy: epigenetic modifiers. Turk J Biol 43, 155-170.
    Pubmed KoreaMed CrossRef
  54. Kim TH, Jung JA and Kim GD et al (2010) The histone deacetylase inhibitor, trichostatin A, inhibits the development of 2,4-dinitrofluorobenzene-induced dermatitis in NC/Nga mice. Int Immunopharmacol 10, 1310-1315.
    Pubmed CrossRef
  55. Kim HJ and Kim YJ (2016) Age-related epigenetic regulation in the brain and its role in neuronal diseases. BMB Rep 49, 671-680.
    Pubmed KoreaMed CrossRef
  56. Lee JH and Ryu H (2010) Epigenetic modification is linked to Alzheimer's disease: is it a maker or a marker?. BMB Rep 43, 649-655.
    Pubmed CrossRef
  57. Han J, Park SG and Bae JB et al (2012) The characteristics of genome-wide DNA methylation in naïve CD4+ T cells of patients with psoriasis or atopic dermatitis. Biochem Biophys Res Commun 422, 157-163.
    Pubmed CrossRef
  58. Chen M, Chen ZQ and Cui PG et al (2008) The methylation pattern of p16INK4a gene promoter in psoriatic epidermis and its clinical significance. Br J Dermatol 158, 987-993.
    Pubmed CrossRef
  59. Kim TH, Jung JA and Kim GD et al (2010) The histone deacetylase inhibitor, trichostatin A, inhibits the development of 2,4-dinitrofluorobenzene-induced dermatitis in NC/Nga mice. Int Immunopharmacol 10, 1310-1315.
    Pubmed CrossRef
  60. Ming M, Zhao B and Shea CR et al (2015) Loss of sirtuin 1 (SIRT1) disrupts skin barrier integrity and sensitizes mice to epicutaneous allergen challenge. J Allergy Clin Immunol 135, 936-945.
    Pubmed KoreaMed CrossRef
  61. Tovar-Castillo LE, Cancino-Díaz JC and García-Vázquez F et al (2007) Under-expression of VHL and over-expression of HDAC-1, HIF-1alpha, LL-37, and IAP-2 in affected skin biopsies of patients with psoriasis. Int J Dermatol 46, 239-246.
    Pubmed CrossRef
  62. Kwon Y, Choi Y and Kim M et al (2021) HDAC6 and CXCL13 mediate atopic dermatitis by regulating cellular interactions and expression levels of miR-9 and SIRT1. Front Pharmacol 12, 691279.
    Pubmed KoreaMed CrossRef
  63. Reynolds G, Vegh P and Fletcher J et al (2021) Developmental cell programs are co-opted in inflammatory skin disease. Science 371, eaba6500.
    Pubmed KoreaMed CrossRef
  64. Rodda LB, Lu E and Bennett ML et al (2018) Single-cell RNA sequencing of lymph node stromal cells reveals niche-associated heterogeneity. Immunity 48, 1014-1028.
    Pubmed KoreaMed CrossRef
  65. Guerrero-Juarez CF, Dedhia PH and Jin S et al (2019) Single-cell analysis reveals fibroblast heterogeneity and myeloid-derived adipocyte progenitors in murine skin wounds. Nat Commun 10, 650.
    Pubmed KoreaMed CrossRef
  66. Efremova M, Vento-Tormo M, Teichmann SA and Vento-Tormo R (2020) CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat Protoc 15, 1484-1506.
    Pubmed CrossRef
  67. Tung KY, Liu FT, Lai YJ, Lee CH and Hsia YP (2015) Chapter 11-Epigenetics in psoriasis A2-Lu, Qianjin; in In Epigenetics and Dermatology, Chang C.C. and Richardson B.C. (eds.), 227-24, Academic Press, Boston, MA, USA.
    CrossRef
  68. Yi JZ and McGee JS (2021) Epigenetic‐modifying therapies: an emerging avenue for the treatment of inflammatory skin diseases. Exp Dermatol 30, 1167-1176.
    Pubmed CrossRef
  69. Zhang L, Chai R and Tai Z et al (2024) Noval advance of histone modification in inflammatory skin diseases and related treatment methods. Front Immunol 14, 1286776.
    Pubmed KoreaMed CrossRef
  70. Chen C, Yi X and Liu P et al (2023) CD147 Facilitates the pathogenesis of psoriasis through glycolysis and H3K9me3 modification in keratinocytes. Research (Wash D C) 6, 0167.
    Pubmed KoreaMed CrossRef
  71. Huang S, Zhen Y and Yin X et al (2023) KMT2C induced by FABP5P3 aggravates keratinocyte hyperproliferation and psoriasiform skin inflammation by upregulating the transcription of PIK3R3. J Invest Dermatol 143, 37-47.
    Pubmed CrossRef
  72. Zhang T, Yang L and Ke Y et al (2020) EZH2-dependent epigenetic modulation of histone H3 lysine-27 contributes to psoriasis by promoting keratinocyte proliferation. Cell Death Dis 11, 826.
    Pubmed KoreaMed CrossRef
  73. Masalha M, Ben-Dov IZ and Ram O et al (2021) H3K27Ac modification and gene expression in psoriasis. J Dermatol Sci 103, 93-100.
    Pubmed CrossRef
  74. Xia X, Cao G and Sun G et al (2020) GLS1-mediated glutaminolysis unbridled by MALT1 protease promotes psoriasis pathogenesis. J Clin Invest 130, 5180-5196.
    Pubmed KoreaMed CrossRef
  75. Traisaeng S, Herr DR, Kao HJ, Chuang TH and Huang CM (2019) A derivative of butyric acid, the fermentation metabolite of staphylococcus epidermidis, inhibits the growth of a staphylococcus aureus strain isolated from atopic dermatitis patients. Toxins (Basel) 11, 311.
    Pubmed KoreaMed CrossRef
  76. Zhao M, Feng D and Hu L et al (2023) 3D genome alterations in T cells associated with disease activity of systemic lupus erythematosus. Ann Rheum Dis 82, 226-234.
    Pubmed KoreaMed CrossRef
  77. Zhao M, Wu X and Zhang Q et al (2010) RFX1 regulates CD70 and CD11a expression in lupus T cells by recruiting the histone methyltransferase SUV39H1. Arthritis Res Ther 12, R227.
    Pubmed KoreaMed CrossRef
  78. Tsou PS, Coit P, Kilian NC and Sawalha AH (2018) EZH2 modulates the DNA methylome and controls T cell adhesion through junctional adhesion molecule A in lupus patients. Arthritis Rheumatol 70, 98-108.
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles
  • CrossRef (0)

Funding Information
  • Hannam University
      10.13039/501100002488

Collections

Services
Social Network Service

e-submission

Archives