Next Article in Journal
Palmoplantar Psoriasis: A Clinico-Pathologic Study on a Series of 21 Cases with Emphasis on Differential Diagnosis
Next Article in Special Issue
The Philosophy of Dermatopathology
Previous Article in Journal
Potential Value of Visfatin, Omentin-1, Nesfatin-1 and Apelin in Renal Cell Carcinoma (RCC): A Systematic Review and Meta-Analysis
Previous Article in Special Issue
Secondary Malignant Tumors Arising in Nevus Sebaceus: Two Case Reports
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lichen Sclerosus: A Current Landscape of Autoimmune and Genetic Interplay

Department of Dermatology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui 910-1193, Japan
*
Author to whom correspondence should be addressed.
Diagnostics 2022, 12(12), 3070; https://doi.org/10.3390/diagnostics12123070
Submission received: 25 September 2022 / Revised: 24 November 2022 / Accepted: 30 November 2022 / Published: 6 December 2022

Abstract

:
Lichen sclerosus (LS) is an acquired chronic inflammatory dermatosis predominantly affecting the anogenital area with recalcitrant itching and soreness. Progressive or persistent LS may cause urinary and sexual disturbances and an increased risk of local skin malignancy with a prevalence of up to 11%. Investigations on lipoid proteinosis, an autosomal recessive genodermatosis caused by loss-of-function mutations in the extracellular matrix protein 1 (ECM1) gene, led to the discovery of a humoral autoimmune response to the identical molecule in LS, providing evidence for an autoimmune and genetic counterpart targeting ECM1. This paper provides an overview of the fundamental importance and current issue of better understanding the immunopathology attributed to ECM1 in LS. Furthermore, we highlight the pleiotropic action of ECM1 in homeostatic and structural maintenance of skin biology as well as in a variety of human disorders possibly associated with impaired or gained ECM1 function, including the inflammatory bowel disease ulcerative colitis, Th2 cell-dependent airway allergies, T-cell and B-cell activation, and the demyelinating central nervous system disease multiple sclerosis, to facilitate sharing the concept as a plausible therapeutic target of this attractive molecule.

1. Introduction

Lichen sclerosus (LS), also known as ‘lichen sclerosus et atrophicus’, ‘balanitis xerotica obliterans’, ’kraurosis vulvae’, or ‘hypoplastic dystrophy’, is an acquired chronic inflammatory disease that primarily affects the skin and mucous membranes, with a high occurrence in the anogenital area [1,2,3]. LS represents one of the most common referrals for pruritis and structural alteration in the vulva [1,4]. The predilection sites of the disease often cause serious urinary and sexual dysfunction, including dyspareunia and psychological impairments. In addition, LS has been associated with an increased risk of malignancy, mostly squamous cell carcinoma, in long-standing lesions in both sexes [5,6,7,8]. For therapeutic remedies, the topical application of potent corticosteroids is a primary mainstay [4,9,10]. Treatment options for refractories to the standard treatment regimens include systemic or local immunosuppressants (e.g., oral or topical calcineurin inhibitors), retinoids, phototherapy, and photodynamic therapy [11,12,13,14,15]. The mechanisms of action of these agents imply the possible involvement of an immunological disturbance in LS.
Although the pathogenesis of LS has yet to be elucidated, a series of etiological and epidemiological studies have suggested a possible genetic susceptibility and an autoimmune basis for the disease. For example, LS has a higher familial predisposition [16,17]; of 1052 individual cases with LS, 126 (~12%) had family histories. In addition, there have been increasing reports of monozygotic and dizygotic twins with LS. Genetic assessment of reliable numbers of LS cohorts identified a high association with the presence of particular human leukocyte antigens (HLAs) and haplotypes [18,19,20], such as DQ7, DR12, DRB1*12, and DRB1*13. Clinically, LS tends to coincide with various promiscuous autoimmune diseases with serum autoantibodies, such as morphea, Hashimoto’s thyroiditis, rheumatoid arthritis, pernicious anemia, type I diabetes mellitus, alopecia areata, vitiligo, morphea, and immuno-bullous mucocutaneous diseases [21,22,23,24,25,26,27]. Of these, serum anti-thyroid antibodies were highly detectable in female LS (11–12%), compared to normal females. A series of clinical evidence has gradually suggested a scenario for the possible involvement of autoimmune imbalance, particularly autoimmune response to skin antigen(s), in LS.
More critically, loss-of-function mutations in the extracellular matrix protein 1 (ECM1) gene were demonstrated in an autosomal recessive genodermatosis referred to as lipoid proteinosis (LiP), a counterpart disease that is a similar skin pathology to LS [28]. This clinical rationale has led to the identification of humoral autoimmunity to ECM1 protein in patients with female anogenital LS [29], which was also applicable in the immunopathogenesis of male penile LS [2]. Furthermore, the similar clinicopathology between LS and LiP has allowed for considerable progress in addressing the in vivo biological function of this attractive molecule in animal model studies using a passive transfer of anti-ECM1 antibodies, and gene knockout and transgenic systems targeted for ECM1 in mice and zebrafish [30,31,32,33]. In addition, recent studies have demonstrated the role of ECM1 in the genetic predisposition to the inflammatory bowel disease (IBD) ulcerative colitis [34,35], the acquisition of immune tolerance and allergic responses via particular T-cell subsets such as CD4+CD25+ regulatory T cells and Th2 cells [31,36], and activation of abundant B-cell biology [33]. Thus, the biology of ECM1 positions it as an attractive core candidate to interconnect disease-specific pathophysiology.

2. Clinical Characteristics of LS

2.1. Clinical Features

LS mostly affects the anogenital skin and mucous membranes in the vast majority of cases (85–98%), typically female [1,2,3]. In male patients, the site-specific predisposition has been associated with a high occurrence rate of LS on normal penile skin grafts whose original diseases were unrelated to LS [37], but this is not a fate of the original skin sites. Without regard to gender, the typical clinical picture includes erosions as well as whitish-pale, indurative polygonal (porcelain-like) papules and plaques, which later change to atrophic scarring (Figure 1). An itching sensation, among various symptoms, is primary and inevitable [38]. The lesion may account for the bullous and hemorrhagic appearance, but it causes a potential difficulty in arriving at an accurate diagnosis [39,40]. Several reports for extragenital LS cases have suggested a variety of affected skin sites including the head, neck, scalp, palms and soles, periorbital area, tongue, lip, and peristoma skin [41] (Figure 2). Interestingly, extragenital LS, unlike genital disease, seldom develops local skin malignancies, giving way to the assumption that genital circumstances can be a primary extrinsic factor in the development of LS. The proposed candidate may include occasional herpes virus infection, urine and/or fecal irritation, constitutive colonization of gram-negative bacteria, or any combination of these.

2.2. Epidemiology and Etiology

Results from epidemiological studies indicated that LS was probably underreported and may have had a prevalence of approximately 0.1–0.3%, with a male-to-female ratio of 1:10 [42,43]. LS can develop at any age; however, a bimodal peak has been found in prepubertal girls and postmenopausal women as well as in middle-aged men [2,42,44,45,46]. A retrospective analysis using a large number (n = 411) of uncharacterized penile dermatoses revealed that ~10% of the foreskin biopsy samples showed the typical LS pathology [47]. Therefore, LS may occur far more frequently than previously expected, although the gender bias remains unchanged (~3% in females and >0.07% in males) [46,48]. The significant sex ratio and age bimodality indicates a possible association with hormonal imbalance, particularly estrogen deficiency, but hormone replacement therapy neither improves existing disease nor provides any protective effects against the disease. Persistent LS may result in severe scarring, which can lead to impairments in micturition and sexual activity, urethral stricture, and associated physical morbidities [49,50], and more importantly an increased risk of malignancy, particularly non-differentiated squamous cell carcinoma (4.7–10.7%) [51,52] (Figure 1).

2.3. Histopathological Features

Typical LS pathology displays a thickening epidermis with hyperkeratosis and follicular plugging, particularly in the early clinical stage, consequently followed by atrophic flattening of the epidermal rete ridges (Figure 3). The dermis underneath undergoes zonal hyalinization, intermingled with amorphous eosinophilic materials, homogeneous collagen bundles, and telangiectasia. A band-like infiltration of inflammatory cells may be present along with and/or separate from the hyalinizing dermis, which becomes sparser and more focal during the clinical course. However, each of these pathological findings often coexists at differing frequencies and degrees. Skin biopsies may therefore provide inconsistent pictures and result in difficulty differentiating other mimicking dermatoses affected in the anogenital area, resulting in the dilemma of diagnostic inaccuracy and delay [53,54].

3. Molecular Characteristics of ECM1: A Secretory Glycoprotein

3.1. Historical Background for the Discovery of the ECM1 Gene: What It Means

The human ECM1 gene was first isolated in 1997 and was mapped to chromosome 1q21.2, located centromerically to the gene cluster termed epidermal differentiation complex (EDC) [55,56]. Comparing the plane structure to the previously discovered mouse Ecm1 gene in 1994 [57], the human counterpart represents one exon fewer than the mouse gene; the sequence is homologous to the sixth shortest mouse exon [55]. The upstream regulatory sequences of the human gene contain putative binding sites for various major transcription factors, such as GATA, Sp1, AP-1, and ETS family members, all of which, except for the potential GATA-binding motifs, are highly conserved with the equivalent portion of the mouse Ecm1 gene [58]. The ECM gene is highly conserved and expressed in most eukaryotes and their various cell types [59], supporting the concept of potential significance in the evolutionary process, as was the discovery of a genetic disease caused by mutations in this gene, lipoid proteinosis [28].

3.2. Gene Structure and Variants

The human ECM1 gene is located on chromosome 1q21.2 and encodes four splice variants, ECM1a–d [55,56]. ECM1a (1.8-kb, 540 amino acids) comprises 10 exons, whereas ECM1b (1.4-kb, 415 amino acids) only lacks exon 7. ECM1c (1.85-kb, 559 amino acids) contains an additional exon (5a) within intron 5 of ECM1a. These three major variants show widespread and differential expression patterns in human tissues [58,60]. For example, ECM1a is ubiquitously expressed in major organs including the skin, liver, intestine, lung, ovary, prostate, testis, skeletal muscle, pancreas, and kidney, with the greatest expression levels observed in the placenta and heart. ECM1b expression seems to be restricted to the tonsils and epidermal keratinocytes, whereas the tissue/cell type-specific expression of ECM1c remains to be identified [61]. ECM1d comprises a minimum splicing variant, with an out-of-frame insertion of 71 nucleotides at the 5′ end of exon 2, resulting in a truncated protein of 57 amino acids [62] and an enigmatic biological significance. In skin, ECM1a was expressed in the epidermal basal layer, dermal blood vessels, outer root sheath of hair follicles, sebaceous lobules, and sweat gland epithelia, whereas ECM1b was localized to the suprabasal layers of the epidermis [60,61,63]. Thus, in vivo associations between each of the ECM1 splicing variants and skin biology are gradually forming.

3.3. Protein Structure and Function

ECM1 is an 85-kDa-secreted glycoprotein known to play pivotal roles in the structural and homeostatic organization of various skin components through direct binding with various extracellular molecules, such as perlecan, matrix metalloproteinase family members, fibulins, fibrillins, fibronectin, laminin-332, type IV and type VII collagens, cartilage-derived oligomeric matrix protein (COMP), proteoglycans, glycosaminoglycans, phospholipids (particularly phospholipid scramblase 1), and progranulin chondrogenic growth factor (PGRN) [61,64,65,66,67,68,69]. Of note, these molecules co-localize immunohistologically with in vivo ECM1 in human skin. The multifocal interaction between skin structural molecules contributes to the biological significance of ECM1 in epidermal growth and differentiation, basement membrane integrity, angiogenesis, endochondral development, and certain malignancies, as well as in the structural maintenance of the dermis (Figure 4). Most of these biological activities are associated with positive regulation of cell proliferation, migration, and differentiation, resulting in tissue formation and organization; however, negative effects on chondrocyte hypertrophy, matrix mineralization, and endochondral bone formation have been described as well [70,71,72]. ECM1 thus penetrates into the fundamental skin biology via complex organization with surrounding microstructural molecules, and mutations of their corresponding genes are responsible for a hereditary genodermatosis LiP [28].

4. Autoimmune Response in LS

4.1. Etiological Scenario for Autoimmunity to ECM1 in LS

Although no plausible evidence regarding the local and systemic autoimmune reactions characteristic of LS has been put forth, recent progress on the screening of disease-specific serum autoantibodies in LS is extrapolated from bipolar evidence for possible genetic susceptibility and a humoral autoimmune basis for the disease. Specifically, study findings have identified variable intra-familial cases with LS [16,73], an association with particular HLA class II antigens (DQ7-9, DR11, DR12, and DQ17) [18,20], and a high coexistence of various autoimmune diseases, such as morphea, Hashimoto’s thyroiditis, rheumatoid arthritis, pernicious anemia, type I diabetes mellitus, alopecia areata, vitiligo, bullous pemphigoid, and mucous membrane pemphigoid [21,22,23,24,25,27]. Of these autoantibody- and/or T-cell-driven diseases, anti-thyroid antibodies were highly detectable in female LS patients (11.1–40%) compared with other organ- and tissue-specific autoantibodies. However, the autoantibodies mostly do not correlate with either severity or duration of the corresponding diseases, proving irrelevant as a consequence of LS [74].
A century ago, evidence implicating a humoral autoimmune response in LS was demonstrated in a case where probable LS was induced through the injection of an autologous serum from an affected individual into non-lesional skin [75]. More critically, the skin pathology of LS shares considerable overlap with that of LiP (OMIM 247100), an ECM1-deficient genetic skin disease [28], for example, trauma-induced inflammation (also known as Koebner’s phenomenon) and lesional skin microscopy showing the atrophic epidermis with hyperkeratosis, disruption and duplication of the basement membrane, and hyaline (glassy-like) collagen changes and telangiectasia in the upper dermis (Figure 3). Altogether, this clinicopathological evidence straightforwardly implicates a counterpart disease concept targeting ECM1 in both LS and LiP [76].

4.2. Identification of IgG Autoantibodies Reactive with ECM1 in LS

Primary screening using immunoblotting with cultured normal human keratinocyte substrates identified detectable levels of serum IgG-class antibodies to three of the four ECM1 isoforms, namely ECM1a–c in ~70% of female patients with genital LS [29]. The fidelity of the seroreactivity was confirmed using a bacterially generated full-length recombinant ECM1a protein. Thereafter, antigen-specific enzyme-linked immunosorbent assays (ELISAs) utilizing a highly antigenic portion of the recombinant ECM1 protein (359–559 amino acids) optimized the immunoreactivity of serum anti-ECM1 antibodies in 74–80% of female patients with genital LS, with 94% specificity in discriminating LS from other autoimmune diseases and healthy controls [30]. In another cohort study, ECM1 seroreactivity was also detectable in male patients with male penile LS [2]. The update series has further accelerated the serological diagnostic accuracy in individual cases suspicious of LS [39,75]. Irrespective of gender, humoral autoimmunity to ECM1 needs to be considered on the basis of its pathogenic significance in LS.

5. Issues that Need to be Addressed to Better Understand ECM1 Autoimmunity in LS

5.1. Lack of In Vivo-Bound Anti-ECM1 IgG in the LS Skin

Debate continues regarding the difficulties in detecting the anti-ECM1 autoantibody in LS lesional skin. For example, direct immunofluorescence studies of LS skin have shown no signals [44,77]. In addition, indirect immunofluorescence studies using LS patients’ sera on normal human skin sections also showed negative (with standard dilutions of the sera) or only faintly positive signals along with the lower epidermis and basement membrane. These controversial observations may be attributable to differences in affinity and/or avidity of antigen-specific serum IgG to the in vivo-native ECM1 antigen, because the IgG fraction of affinity purified from LS sera exhibits intense immuno-reactivity in the lower epidermis, and similar observations were made from immuno-labeling with rabbit anti-ECM1 polyclonal antibodies on normal human skin [29] (Figure 4). Based on our recognition, ECM1 is considered a secretory glycoprotein that acts as a biological ‘glue’ to stabilize robust structural proteins [78], such as BPAG I/II, laminin-332, and collagens. One may consider that the flowability and turnover of in vivo ECM1, in part, affect the accessibility and reactivity of the antibody. Considering these technical limitations, the standard immunohistochemical approach using patients’ skin or sera remains less valuable in a routine laboratory workup, and the ELISA system specific for ECM1—or at least immunoblotting using recombinant ECM1 protein—may currently be a preferable tool for a noninvasive and objective serodiagnosis in LS [30].

5.2. Difficulty in the Establishment of Mouse Models for LS and LiP

The pathogenic relevance of autoimmunity to ECM1 in LS has been reevaluated by mouse passive-transfer experiments using intra-cutaneous injections of either a rabbit anti-ECM1 polyclonal antibody or an affinity-purified IgG from ECM1 ELISA-positive LS patients’ sera [30]. Both antibodies recognize the regions of the ECM1 protein that are highly homologous between humans and mice, and when injected into the skin, both IgGs were indeed accessible to native mouse ECM1 in vivo, as was identical immunoreactivity to human skin. The mouse skin sites injected either with a rabbit anti-ECM1 polyclonal antibody or an affinity-purified IgG from LS sera exhibited a clinicopathology compatible with the early clinical stage of LS, including erythematous swelling (dermal inflammation) and dilated blood vessels (telangiectasia) for up to two weeks after the initial injection. However, this approach failed to reproduce dermal hyalinosis or scarring, both of which are histological hallmarks of the well-established (late) stage of LS. This incomplete observation raises possible interpretations for how hyalinosis and/or sclerotic events in LS skin are indeed consequences of complex and sensitive events. For example, the sensitivity may depend on the particular genetic background(s) of the mouse strain(s) or the HLA class II haplotypes of the patients [18,20]. In addition, it may be affected by the constitutive impairment of in vivo ECM1 function, e.g., more prolonged exposure to anti-ECM1 antibodies. Considering the clinical aspect that (i) the vast majority of LS affects anogenital skin and (ii) extragenital LS sometimes develops around utero-abdominal fistula [41,79], urinary irritation and/or local infection with gram-negative bacteria may be (a) confounding factor(s).
Additional experiments have explored the fact that conventional/targeted disruptions of the Ecm1 gene in mice (ECM1−/−) render it lethal (surviving for no more than 6–8 weeks) [31,80], strongly suggesting that ECM1 is indispensable for at least early embryonic development. In contrast, LiP patients represent an ECM1-knockout condition in humans who can survive and show no evidence of short life spans or disease-related mortality. In addition to this discrepancy, a systematic disease translational study comparing gene transcriptional responses to inflammatory insults in mice and humans detected disparities in the gene expression profiles between the mouse models and their human counterparts [80]. Species-specific gene regulation may thus represent a challenge to reproducing typical LS pathology in mice.

5.3. Establishment of ECM1-Knockdown Human Dermal Fibroblasts

A series of our failures for recapitulating LS and LiP phenotypes in mouse models led us to re-recognize what happens to the impaired ECM1 function in vitro. ECM1 siRNA knockdown in human dermal fibroblasts showed a significant delay of growth and migratory activities, as well as collagen gel contraction, compared to control fibroblasts [69]. Also, the ECM1 knockdown upregulated genes related to structural, fibrogenic, and carcinogenic properties, some of which shared the skin structural molecules that are major binding partners for ECM1, such as laminin-332, and type IV and VII collagens [64,67,69]. All of the binders displayed altered immunolabeling at the basement membrane zone and dermal vessels in the LS lesional skin (Figure 4), where ECM1 is highly expressed. Combining these in vitro data with the ECM1 autoantibody scenario, one may speculate that the antibody-dependent impairment of ECM1 function disrupts a biological interconnection with the in vivo binding partners, resulting in their functional behavior to maintain the structural integrity responsible for the LS pathology (Figure 5).

6. Lessens from Novel ECM1 Function in Other Animal Models

6.1. Th2 Cell-Dependent Allergic Response in the Airway

DNA microarray assays have disclosed the expression of the Ecm1 gene in mouse hematopoietic cells, particularly in T cells [81,82], although the transcription levels considerably differ in the T-cell differentiation- and lineage-dependent manner; it was much higher in CD4+ helper T cells and CD4+CD25+ T cells (Tregs) but relatively lower in CD8+ cytotoxic T cells and CD3-negative naïve T cells. Of the CD4+ helper T-cell lineage, the ECM1 expression was almost prone to Th2 cells [31], indicating the possible association between ECM1 and allergic reactions. This scenario was also compounded through the finding that chimeric BALB/c mice transplanted with ECM1-deficient bone marrow cells showed a decrease in inflammatory response in experimentally induced airway allergy. Functional analysis for several T-cell lineages from ECM1-knockout mice exhibited no substantial differences in the proliferation activity, cytokine/chemokine profiles, and polarization of their differentiation, suggesting the direct action of ECM1 in Th2 cell trafficking from lymph nodes into circulation.
In Th2 cells, ECM1 mRNA and protein expression were detectable three days after antigen-dependent engagement of the T-cell receptor. Subsequently, ECM1 can bind with an IL-2 receptor subunit (CD122), but with neither CD25 nor CD132, to inhibit the phosphorylation and activation of the downstream-signaling molecules, such as STAT5, KLF2, and S1P1 [31], resulting in the downregulation of Th2 cell trafficking to the local inflammatory sites.
On the other hand, freshly isolated and activated CD4+ CD25+ Tregs highly express ECM1 transcription [83]. CD4+ CD25+Tregs are well-known to regulate innate and adaptive immune responses, tumor immunity, and a potent anti-inflammatory capacity in autoimmune and chronic inflammatory diseases, such as autoimmune encephalitis, diabetes, thyroiditis, IBDs, and contact skin hypersensitivity [84,85,86,87,88]. Naturally occurring Tregs, the other Treg phenotype that comprises up to 5% of the peripheral CD4+ T-cell pool, have also been shown to express ECM1 [87]. More critically, the ECM1 transcription was significantly increased in naïve T cells by transient transduction of forkhead box P3 (FOXP3), a transcription factor that acts as a master control molecule for the development and function of CD4+CD25+ Tregs in the thymus and periphery [88].

6.2. Macrophage Polarization in Inflammatory Bowel Diseases (IBDs)

Genotyping using a reliable number of ulcerative colitis cohorts (n = 905) determined a strong disease susceptibility locus at the ECM1 gene [34]. The foothold identification further accelerated the research activity concerning ECM1-targeted culprit cells in the disease. Mice transplanted with ECM1 knocked-down macrophages, a phenotype unable to polarize towards M1 macrophage, decreased pathological inflammation of colitis in experimental IBD mice [34], raising a direct interpretation of ECM1 function to regulate the IBD-dependent macrophage lineage. However, data from several trials determining ECM1 gene mutation and/or polymorphism in IBD cohorts remains unstable as the genetic signature [88,89], which may be impacted by racial difference. Also, there has been no available evidence of a relationship between ECM1 and the intestinal epithelial barrier, as well as permeability balance and luminal antigen absorption in the intestine interface.

6.3. Miscellaneous

6.3.1. Multiple Sclerosis

Multiple sclerosis is a chronic inflammatory condition in the central nervous system characterized by demyelination and axonal damage through Th17-dependent innate immunity. Administration of recombinant ECM1 ameliorated the severity of encephalomyelitis with cerebral demyelination and inflammation, accompanied by a decrease in the Th17 response, in an experimental model for multiple sclerosis [90]. Inversely, in vivo overexpression of ECM1 successfully inhibited encephalomyelitis with Th17 cell activation. The protective action of ECM1 is mediated in part by its direct interaction with av-integrin on dendritic cells, blocking the integrin-mediated activation of TGF-β. Data support a possible engagement in a replacement therapy targeting ECM1.

6.3.2. B Cell Function

Follicular helper T cells (TFH) are a subset of the CD4+ helper T-cell lineage that enables a variety of B-cell responses, i.e., the formation of germinal centers (GCs), affinity maturation of GC B cells, differentiation of high-affinity antibody-producing plasma cells, and production of memory B cells. All the B cell-specific reactions were impaired by an ECM1 knockout in antigen-immunized mice [33]. Exogenously injected ECM1 into mice infected with influenza virus exhibited a protective immune response via enhancing differentiation towards TFH cells and production of virus-neutralizing antibodies. ECM1 can therefore promote TFH cell differentiation and antibody production, both of which are indispensable for humoral autoimmunity.

7. Conclusions

Considerable progress has recently been made in both clinical and animal studies designed to elucidate the in vivo function of ECM1. Novel insights regarding this molecule in the restricted T-cell repertoire, B-cell activation, organ-specific allergic reaction, and genetic susceptibility to ulcerative colitis have been condensed during the last decade. In addition, much attention has been paid to the role of ECM1 in tumor biology, particularly its microenvironment as an alternative to tumor-directed therapy [72,91,92]. Notwithstanding these updates, genetic ablation of ECM1 and passive transfer of ECM1-specific antibodies in mice has yet to fully explain the characteristic pathophysiology in human diseases LiP and LS, respectively. Future studies concerning the establishment of animal models for LS now await sophistication of the overall technical processes.
Clinical observation shows that LiP patients are viable without an inherent susceptibility to any type of cancer, unlike LS patients. ECM1 can therefore be dispensable or at least compensable spatially and temporally for the development of certain organs, including the skin. Future studies will encourage research on the tissue and organ developmental stage-specific significance of ECM1 action.

Author Contributions

Conceptualization, structure, and writing, N.O.; conceptualization and correction, M.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding

Institutional Review Board Statement

Not available.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by JSPS KAKENHI Grant Numbers JP21K08321 (N.O.) and JP22K08426 (M.H.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Goldstein, A.T.; Marinoff, S.C.; Christopher, K.; Srodon, M. Prevalence of Vulvar Lichen Sclerosus in a General Gynecology Practice. J. Reprod. Med. 2005, 50, 477–480. [Google Scholar] [PubMed]
  2. Edmonds, E.V.J.; Hunt, S.; Hawkins, D.; Dinneen, M.; Francis, N.; Bunker, C.B. Clinical Parameters in Male Genital Lichen Sclerosus: A Case Series of 329 Patients. J. Eur. Acad. Dermatology Venereol. 2012, 26, 730–737. [Google Scholar] [CrossRef] [PubMed]
  3. Hagedorn, M.; Buxmeyer, B.; Schmitt, Y.; Bauknecht, T. Survey of Genital Lichen Sclerosus in Women and Men. Arch. Gynecol. Obstet. 2002, 266, 86–91. [Google Scholar] [CrossRef] [PubMed]
  4. Kirtschig, G.; Becker, K.; Günthert, A.; Jasaitiene, D.; Cooper, S.; Chi, C.C.; Kreuter, A.; Rall, K.K.; Aberer, W.; Riechardt, S.; et al. Evidence-Based (S3) Guideline on (Anogenital) Lichen Sclerosus. J. Eur. Acad. Dermatol. Venereol. 2015, 29, e1–e43. [Google Scholar] [PubMed]
  5. Bleeker, M.C.G.; Visser, P.J.; Overbeek, L.I.H.; Van Beurden, M.; Berkhof, J. Lichen Sclerosus: Incidence and Risk of Vulvar Squamous Cell Carcinoma. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1224–1230. [Google Scholar] [CrossRef] [Green Version]
  6. Davick, J.; Samuelson, M.; Krone, J.; Stockdale, C. The Prevalence of Lichen Sclerosus in Patients with Vulvar Squamous Cell Carcinoma. Int. J. Gynecol. Pathol. 2017, 36, 305–309. [Google Scholar] [CrossRef]
  7. Micheletti, L.; Preti, M.; Radici, G.; Boveri, S.; Di Pumpo, O.; Privitera, S.S.; Ghiringhello, B.; Benedetto, C. Vulvar Lichen Sclerosus and Neoplastic Transformation: A Retrospective Study of 976 Cases. J. Low. Genit. Tract. Dis. 2016, 20, 180–183. [Google Scholar] [CrossRef] [Green Version]
  8. Leis, M.; Singh, A.; Li, C.; Ahluwalia, R.; Fleming, P.; Lynde, C.W. Risk of Vulvar Squamous Cell Carcinoma in Lichen Sclerosus and Lichen Planus: A Systematic Review. J. Obstet. Gynaecol. Canada 2022, 44, 182–192. [Google Scholar] [CrossRef]
  9. Hasegawa, M.; Ishikawa, O.; Asano, Y.; Sato, S.; Jinnin, M.; Takehara, K.; Fujimoto, M.; Yamamoto, T.; Ihn, H. Diagnostic Criteria, Severity Classification and Guidelines of Lichen Sclerosus et Atrophicus. J. Dermatol. 2018, 45, 891–897. [Google Scholar] [CrossRef] [Green Version]
  10. Neill, S.M.; Lewis, F.M.; Tatnall, F.M.; Cox, N.H. British Association of Dermatologists’ Guidelines for the Management of Lichen Sclerosus 2010. Br. J. Dermatol. 2010, 163, 672–682. [Google Scholar] [CrossRef]
  11. Funaro, D.; Lovett, A.; Leroux, N.; Powell, J. A Double-Blind, Randomized Prospective Study Evaluating Topical Clobetasol Propionate 0.05% versus Topical Tacrolimus 0.1% in Patients with Vulvar Lichen Sclerosus. J. Am. Acad. Dermatol. 2014, 71, 84–91. [Google Scholar] [CrossRef] [PubMed]
  12. Bulbul Baskan, E.; Turan, H.; Tunali, S.; Toker, S.C.; Saricaoglu, H. Open-Label Trial of Cyclosporine for Vulvar Lichen Sclerosus. J. Am. Acad. Dermatol. 2007, 57, 276–278. [Google Scholar] [CrossRef] [PubMed]
  13. Virgili, A.; Corazza, M.; Bianchi, A.; Mollica, G.; Califano, A. Open Study of Topical 0.025% Tretinoin in the Treatment of Vulvar Lichen Sclerosus: One Year of Therapy. J. Reprod. Med. Obstet. Gynecol. 1995, 40, 614–618. [Google Scholar]
  14. Terras, S.; Gambichler, T.; Moritz, R.K.C.; Stücker, M.; Kreuter, A. UV-A1 Phototherapy vs Clobetasol Propionate, 0.05%, in the Treatment of Vulvar Lichen Sclerosus: A Randomized Clinical Trial. JAMA Dermatol. 2014, 150, 621–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Sotiriou, E.; Panagiotidou, D.; Ioannidis, D. An Open Trial of 5-Aminolevulinic Acid Photodynamic Therapy for Vulvar Lichen Sclerosus. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008, 141, 187–188. [Google Scholar] [CrossRef]
  16. Thomas, R.H.M.; Kennedy, C.T.C. The Development of Lichen Sclerosus et Atrophicus in Monozygotic Twin Girls. Br. J. Dermatol. 1986, 114, 377–379. [Google Scholar] [CrossRef]
  17. Sahn, E.E.; Bluestein, E.L.; Oliva, S. Familial Lichen Sclerosus et Atrophicus in Childhood. Pediatr. Dermatol. 1994, 11, 160–163. [Google Scholar] [CrossRef]
  18. Azurdia, R.M.; Luzzi, G.A.; Byren, I.; Welsh, K.; Wojnarowska, F.; Marren, P.; Edwards, A. Lichen Sclerosus in Adult Men: A Study of HLA Associations and Susceptibility to Autoimmune Disease. Br. J. Dermatol. 1999, 140, 79–83. [Google Scholar] [CrossRef]
  19. Marren, P.; Jell, J.; Charnock, F.M.; Bunce, M.; Welsh, K.; Wojnarowska, F. The Association between Lichen Sclerosus and Antigens of the HLA System. Br. J. Dermatol. 1995, 132, 197–203. [Google Scholar] [CrossRef]
  20. Gao, X.H.; Barnardo, M.C.M.N.; Winsey, S.; Ahmad, T.; Cook, J.; Agudelo, J.D.; Zhai, N.; Powell, J.J.; Fuggle, S.V.; Wojnarowska, F. The Association between HLA DR, DQ Antigens, and Vulval Lichen Sclerosus in the UK: HLA DRB112 and Its Associated DRB112/DQB10301/04/09/010 Haplotype Confers Susceptibility to Vulval Lichen Sclerosus, and HLA DRB10301/04 and Its Associated DRB10301/04/DQB10201/02/03 Haplotype Protects from Vulval Lichen Sclerosus. J. Investig. Dermatol. 2005, 125, 895–899. [Google Scholar]
  21. Khan Mohammad Beigi, P. The Immunogenetics of Morphea and Lichen Sclerosus. Adv. Exp. Med. Biol. 2022, 1367, 155–172. [Google Scholar] [PubMed]
  22. Guarneri, F.; Giuffrida, R.; Di Bari, F.; Cannavò, S.P.; Benvenga, S. Thyroid Autoimmunity and Lichen. Front. Endocrinol. 2017, 8, 146. [Google Scholar] [CrossRef] [PubMed]
  23. García-Bravo, B.; Sánchez-Pedreño, P.; Rodríguez-Pichardo, A.; Camacho, F. Lichen Sclerosus et Atrophicus. A Study of 76 Cases and Their Relation to Diabetes. J. Am. Acad. Dermatol. 1988, 19, 482–485. [Google Scholar] [CrossRef] [PubMed]
  24. Harrington, C.I.; Dunsmore, I.R. An Investigation into the Incidence of Auto-immune Disorders in Patients with Lichen Sclerosus and Atrophicus. Br. J. Dermatol. 1981, 104, 563–566. [Google Scholar] [CrossRef] [PubMed]
  25. Thomas, R.H.M.; Ridley, C.M.; McGibbon, D.H.; Black, M.M. Lichen Sclerosus et Atrophicus and Autoimmunity—a Study of 350 Women. Br. J. Dermatol. 1988, 118, 41–46. [Google Scholar] [CrossRef]
  26. Baldo, M.; Bailey, A.; Bhogal, B.; Groves, R.W.; Ogg, G.; Wojnarowska, F. T Cells Reactive with the NC16A Domain of BP180 Are Present in Vulval Lichen Sclerosus and Lichen Planus. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 186–190. [Google Scholar] [CrossRef]
  27. Walsh, M.L.; Leonard, N.; Shawki, H.; Bell, H.K. Lichen Sclerosus and Immunobullous Disease. J. Low. Genit. Tract Dis. 2012, 16, 468–470. [Google Scholar] [CrossRef]
  28. Hamada, T.; McLean, W.H.I.; Ramsay, M.; Ashton, G.H.S.; Nanda, A.; Jenkins, T.; Edelstein, I.; South, A.P.; Bleck, O.; Wessagowit, V.; et al. Lipoid Proteinosis Maps to 1q21 and Is Caused by Mutations in the Extracellular Matrix Protein 1 Gene (ECM1). Hum. Mol. Genet. 2002, 11, 833–840. [Google Scholar] [CrossRef] [Green Version]
  29. Oyama, N.; Chan, I.; Neill, S.M.; Hamada, T.; South, A.P.; Wessagowit, V.; Wojnarowska, F.; D’Cruz, D.; Hughes, G.J.; Black, M.M.; et al. Autoantibodies to Extracellular Matrix Protein 1 in Lichen Sclerosus. Lancet 2003, 362, 118–123. [Google Scholar] [CrossRef]
  30. Oyama, N.; Chan, I.; Neill, S.M.; South, A.P.; Wojnarowska, F.; Kawakami, Y.; D’Cruz, D.; Mepani, K.; Hughes, G.J.; Bhogal, B.S.; et al. Development of Antigen-Specific ELISA for Circulating Autoantibodies to Extracellular Matrix Protein 1 in Lichen Sclerosus. J. Clin. Investig. 2004, 113, 1550–1559. [Google Scholar] [CrossRef]
  31. Li, Z.; Zhang, Y.; Liu, Z.; Wu, X.; Zheng, Y.; Tao, Z.; Mao, K.; Wang, J.; Lin, G.; Tian, L.; et al. ECM1 Controls T(H)2 Cell Egress from Lymph Nodes through Re-Expression of S1P(1). Nat. Immunol. 2011, 12, 178–185. [Google Scholar] [CrossRef] [PubMed]
  32. Li, Q.; Donahue, A.; Smith, S.; McGrath, J.A.; Oyama, N.; Uitto, J. Zebrafish Model of Lipoid Proteinosis. J. Investig. Dermatol. 2011, 131, S70. [Google Scholar]
  33. He, L.; Gu, W.; Wang, M.; Chang, X.; Sun, X.; Zhang, Y.; Lin, X.; Yan, C.; Fan, W.; Su, P.; et al. Extracellular Matrix Protein 1 Promotes Follicular Helper T Cell Differentiation and Antibody Production. Proc. Natl. Acad. Sci. USA 2018, 115, 8621–8626. [Google Scholar] [CrossRef] [PubMed]
  34. Fisher, S.A.; Tremelling, M.; Anderson, C.A.; Gwilliam, R.; Bumpstead, S.; Prescott, N.J.; Nimmo, E.R.; Massey, D.; Berzuini, C.; Johnson, C.; et al. Genetic Determinants of Ulcerative Colitis Include the ECM1 Locus and Five Loci Implicated in Crohn’s Disease. Nat. Genet. 2008, 40, 710–712. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, Y.; Li, X.; Luo, Z.; Ma, L.; Zhu, S.; Wang, Z.; Wen, J.; Cheng, S.; Gu, W.; Lian, Q.; et al. ECM1 Is an Essential Factor for the Determination of M1 Macrophage Polarization in IBD in Response to LPS Stimulation. Proc. Natl. Acad. Sci. USA 2020, 117, 3083–3092. [Google Scholar] [CrossRef]
  36. Sugimoto, N.; Oida, T.; Hirota, K.; Nakamura, K.; Nomura, T.; Uchiyama, T.; Sakaguchi, S. Foxp3-Dependent and -Independent Molecules Specific for CD25+CD4+ Natural Regulatory T Cells Revealed by DNA Microarray Analysis. Int. Immunol. 2006, 18, 1197–1209. [Google Scholar] [CrossRef] [Green Version]
  37. Abdelbaky, A.M.; Aluru, P.; Keegan, P.; Greene, D.R. Development of Male Genital Lichen Sclerosus in Penile Reconstruction Skin Grafts after Cancer Surgery: An Unreported Complication. BJU Int. 2012, 109, 776–779. [Google Scholar] [CrossRef]
  38. Virgili, A.; Borghi, A.; Toni, G.; Minghetti, S.; Corazza, M. Prospective Clinical and Epidemiologic Study of Vulvar Lichen Sclerosus: Analysis of Prevalence and Severity of Clinical Features, Together with Historical and Demographic Associations. Dermatology 2014, 228, 145–151. [Google Scholar] [CrossRef]
  39. Kawakami, Y.; Oyama, N.; Hanami, Y.; Kimura, T.; Kishimoto, K.; Yamamoto, T. A Case of Lichen Sclerosus of the Scalp Associated with Autoantibodies to Extracellular Matrix Protein 1. Arch. Dermatol. 2009, 145, 1458–1460. [Google Scholar] [CrossRef]
  40. Khatib, J.; Wargo, J.J.; Krishnamurthy, S.; Travers, J.B. Hemorrhagic Bullous Lichen Sclerosus: A Case Report. Am. J. Case Rep. 2020, 21, e919353-1. [Google Scholar] [CrossRef]
  41. Al-Niaimi, F.; Lyon, C. Peristomal Lichen Sclerosus: The Role of Occlusion and Urine Exposure? Br. J. Dermatol. 2013, 168, 643–646. [Google Scholar] [CrossRef] [PubMed]
  42. Lansdorp, C.A.; Van Den Hondel, K.E.; Korfage, I.J.; Van Gestel, M.J.; Van Der Meijden, W.I. Quality of Life in Dutch Women with Lichen Sclerosus. Br. J. Dermatol. 2013, 168, 787–793. [Google Scholar] [CrossRef] [PubMed]
  43. Wallace, H.J. Lichen Sclerosus et Atrophicus. Trans. St. Johns. Hosp. Dermatol. Soc. 1971, 57, 9–30. [Google Scholar] [PubMed]
  44. Powell, J.; Wojnarowska, F. Childhood Vulvar Lichen Sclerosus: An Increasingly Common Problem. J. Am. Acad. Dermatol. 2001, 44, 803–806. [Google Scholar] [CrossRef] [PubMed]
  45. Nelson, D.M.; Peterson, A.C. Lichen Sclerosus: Epidemiological Distribution in an Equal Access Health Care System. J. Urol. 2011, 185, 522–525. [Google Scholar] [CrossRef]
  46. Kizer, W.S.; Prarie, T.; Morey, A.F. Balanitis Xerotica Obliterans: Epidemiologic Distribution in an Equal Access Health Care System. South. Med. J. 2003, 96, 9–11. [Google Scholar] [CrossRef]
  47. West, D.S.; Papalas, J.A.; Selim, M.A.; Vollmer, R.T. Dermatopathology of the Foreskin: An Institutional Experience of over 400 Cases. J. Cutan. Pathol. 2013, 40, 11–18. [Google Scholar] [CrossRef]
  48. Leibovitz, A.; Kaplun, V.; Saposhnicov, N.; Habot, B. Vulvovaginal Examinations in Elderly Nursing Home Women Residents. Arch. Gerontol. Geriatr. 2000, 31, 1–4. [Google Scholar] [CrossRef]
  49. Vittrup, G.; Westmark, S.; Riis, J.; Mørup, L.; Heilesen, T.; Jensen, D.; Melgaard, D. The Impact of Psychosexual Counseling in Women With Lichen Sclerosus: A Randomized Controlled Trial. J. Low. Genit. Tract Dis. 2022, 26, 258–264. [Google Scholar] [CrossRef]
  50. Haefner, H.K.; Aldrich, N.Z.; Dalton, V.K.; Gagné, H.M.; Marcus, S.B.; Patel, D.A.; Berger, M.B. The Impact of Vulvar Lichen Sclerosus on Sexual Dysfunction. J. Women’s Health 2014, 23, 765–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Halonen, P.; Jakobsson, M.; Heikinheimo, O.; Riska, A.; Gissler, M.; Pukkala, E. Lichen Sclerosus and Risk of Cancer. Int. J. Cancer 2017, 140, 1998–2002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Lee, A.; Bradford, J.; Fischer, G. Long-Term Management of Adult Vulvar Lichen Sclerosus: A Prospective Cohort Study of 507 Women. JAMA Dermatol. 2015, 151, 1061–1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Regauer, S.; Liegl, B.; Reich, O. Early Vulvar Lichen Sclerosus: A Histopathological Challenge. Histopathology 2005, 47, 340–347. [Google Scholar] [CrossRef] [PubMed]
  54. Niamh, L.; Naveen, S.; Hazel, B. Diagnosis of Vulval Inflammatory Dermatoses: A Pathological Study with Clinical Correlation. Int. J. Gynecol. Pathol. 2009, 28, 554–558. [Google Scholar] [CrossRef]
  55. Smits, P.; Ni, J.; Feng, P.; Wauters, J.; Van Hul, W.; El Boutaibi, M.; Dillon, P.J.; Merregaert, J. The Human Extracellular Matrix Gene 1 (ECM1): Genomic Structure, CDNA Cloning, Expression Pattern, and Chromosomal Localization. Genomics 1997, 45, 487–495. [Google Scholar] [CrossRef] [PubMed]
  56. Johnson, M.R.; Wilkin, D.J.; Vos, H.L.; Ortiz De Luna, R.I.; Dehejia, A.M.; Polymeropoulos, M.H.; Francomano, C.A. Characterization of the Human Extracellular Matrix Protein 1 Gene on Chromosome 1q21. Matrix Biol. 1997, 16, 289–292. [Google Scholar] [CrossRef] [Green Version]
  57. Mathieu, E.; Meheus, L.; Raymackers, J.; Merregaert, J. Characterization of the Osteogenic Stromal Cell Line MN7: Identification of Secreted MN7 Proteins Using Two-Dimensional Polyacrylamide Gel Electrophoresis, Western Blotting, and Microsequencing. J. Bone Miner. Res. 1994, 9, 903–913. [Google Scholar] [CrossRef]
  58. Smits, P.; Bhalerao, J.; Merregaert, J. Molecular Cloning and Characterization of the Mouse Ecm1 Gene and Its 5’ Regulatory Sequences. Gene 1999, 226, 253–261. [Google Scholar] [CrossRef]
  59. Sercu, S.; Oyama, N.; Merregaert, J. Importance of Extracellular Matrix Protein 1 (ECM1) in Maintaining the Functional Integrity of the Human Skin. Undefined 2009, 3, 44–51. [Google Scholar] [CrossRef]
  60. Sander, C.S.; Sercu, S.; Ziemer, M.; Hipler, U.C.; Elsner, P.; Thiele, J.; Merregaert, J. Expression of Extracellular Matrix Protein 1 (ECM1) in Human Skin Is Decreased by Age and Increased upon Ultraviolet Exposure. Br. J. Dermatol. 2006, 154, 218–224. [Google Scholar] [CrossRef]
  61. Mongiat, M.; Fu, J.; Oldershaw, R.; Greenhalgh, R.; Gown, A.M.; Iozzo, R.V. Perlecan Protein Core Interacts with Extracellular Matrix Protein 1 (ECM1), a Glycoprotein Involved in Bone Formation and Angiogenesis. J. Biol. Chem. 2003, 278, 17491–17499. [Google Scholar] [CrossRef] [Green Version]
  62. Horev, L.; Potikha, T.; Ayalon, S.; Molho-Pessach, V.; Ingber, A.; Gany, M.A.; Edin, B.S.; Glaser, B.; Zlotogorski, A. A Novel Splice-Site Mutation in ECM-1 Gene in a Consanguineous Family with Lipoid Proteinosis. Exp. Dermatol. 2005, 14, 891–897. [Google Scholar] [CrossRef] [PubMed]
  63. Smits, P.; Poumay, Y.; Karperien, M.; Tylzanowski, P.; Wauters, J.; Huylebroeck, D.; Ponec, M.; Merregaert, J. Differentiation-Dependent Alternative Splicing and Expression of the Extracellular Matrix Protein 1 Gene in Human Keratinocytes. J. Investig. Dermatol. 2000, 114, 718–724. [Google Scholar] [CrossRef] [PubMed]
  64. Sercu, S.; Zhang, M.; Oyama, N.; Hansen, U.; Ghalbzouri, A.E.L.; Jun, G.; Geentjens, K.; Zhang, L.; Merregaert, J.H. Interaction of Extracellular Matrix Protein 1 with Extracellular Matrix Components: ECM1 Is a Basement Membrane Protein of the Skin. J. Investig. Dermatol. 2008, 128, 1397–1408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kong, L.; Tian, Q.; Guo, F.; Mucignat, M.T.; Perris, R.; Sercu, S.; Merregaert, J.; Di Cesare, P.E.; Liu, C.-J. Interaction between Cartilage Oligomeric Matrix Protein and Extracellular Matrix Protein 1 Mediates Endochondral Bone Growth. Matrix Biol. 2010, 29, 276–286. [Google Scholar] [CrossRef] [Green Version]
  66. Merregaert, J.; Van Langen, J.; Hansen, U.; Ponsaerts, P.; El Ghalbzouri, A.; Steenackers, E.; Van Ostade, X.; Sercu, S. Phospholipid Scramblase 1 Is Secreted by a Lipid Raft-Dependent Pathway and Interacts with the Extracellular Matrix Protein 1 in the Dermal Epidermal Junction Zone of Human Skin. J. Biol. Chem. 2010, 285, 37823–37837. [Google Scholar] [CrossRef] [Green Version]
  67. Sercu, S.; Lambeir, A.M.; Steenackers, E.; El Ghalbzouri, A.; Geentjens, K.; Sasaki, T.; Oyama, N.; Merregaert, J. ECM1 Interacts with Fibulin-3 and the Beta 3 Chain of Laminin 332 through Its Serum Albumin Subdomain-like 2 Domain. Matrix Biol. 2009, 28, 160–169. [Google Scholar] [CrossRef]
  68. Fujimoto, N.; Terlizzi, J.; Brittingham, R.; Fertala, A.; McGrath, J.A.; Uitto, J. Extracellular Matrix Protein 1 Interacts with the Domain III of Fibulin-1C and 1D Variants through Its Central Tandem Repeat 2. Biochem. Biophys. Res. Commun. 2005, 333, 1327–1333. [Google Scholar] [CrossRef]
  69. Utsunomiya, N.; Utsunomiya, A.; Chino, T.; Hasegawa, M.; Oyama, N. Gene Silencing of Extracellular Matrix Protein 1 (ECM1) Results in Phenotypic Alterations of Dermal Fibroblasts Reminiscent of Clinical Features of Lichen Sclerosus. J. Dermatol. Sci. 2020, 100, 99–109. [Google Scholar] [CrossRef]
  70. Han, Z.; Ni, J.; Smits, P.; Underhill, C.B.; Xie, B.; Chen, Y.; Liu, N.; Tylzanowski, P.; Parmelee, D.; Feng, P.; et al. Extracellular Matrix Protein 1 (ECM1) Has Angiogenic Properties and Is Expressed by Breast Tumor Cells. FASEB J. 2001, 15, 988–994. [Google Scholar] [CrossRef] [Green Version]
  71. Deckers, M.M.L.; Smits, P.; Karperien, M.; Ni, J.; Tylzanowski, P.; Feng, P.; Parmelee, D.; Zhang, J.; Bouffard, E.; Gentz, R.; et al. Recombinant Human Extracellular Matrix Protein 1 Inhibits Alkaline Phosphatase Activity and Mineralization of Mouse Embryonic Metatarsals in Vitro. Bone 2001, 28, 14–20. [Google Scholar] [CrossRef] [PubMed]
  72. Sercu, S.; Zhang, L.; Merregaert, J. The Extracellular Matrix Protein 1: Its Molecular Interaction and Implication in Tumor Progression. Cancer Investig. 2008, 26, 375–384. [Google Scholar] [CrossRef] [PubMed]
  73. Sherman, V.; McPherson, T.; Baldo, M.; Salim, A.; Gao, X.; Wojnarowska, F. The High Rate of Familial Lichen Sclerosus Suggests a Genetic Contribution: An Observational Cohort Study. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 1031–1034. [Google Scholar] [CrossRef] [PubMed]
  74. Goolamali, S.K.; Shuster, S.; Barnes, E.W.; Irvine, W.J. Organ-Specific Antibodies in Patients with Lichen Sclerosus. Br. Med. J. 1974, 4, 78–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Irvine, H. Idiopathic Atrophy of the Skin. JAMA 1913, 61, 396–400. [Google Scholar] [CrossRef]
  76. Chan, I. The Role of Extracellular Matrix Protein 1 in Human Skin. Clin. Exp. Dermatol. 2004, 29, 52–56. [Google Scholar] [CrossRef]
  77. Bushkell, L.L.; Friedrich, E.G.; Jordon, R.E. An Appraisal of Routine Direct Immunofluorescence in Vulvar Disorders. Acta Derm. Venereol. 1981, 61, 157–161. [Google Scholar]
  78. Chan, I.; Liu, L.; Hamada, T.; Sethuraman, G.; Mcgrath, J.A. The Molecular Basis of Lipoid Proteinosis: Mutations in Extracellular Matrix Protein 1. Exp. Dermatol. 2007, 16, 881–890. [Google Scholar] [CrossRef]
  79. Kirby, L.; Gran, S.; Kreuser-Genis, I.; Owen, C.; Simpson, R. Is Urinary Incontinence Associated with Lichen Sclerosus in Females? A Systematic Review and Meta-Analysis. Ski. Health Dis. 2021, 1, e13. [Google Scholar] [CrossRef]
  80. Sercu, S.; Liekens, J.; Umans, L.; Van De Putte, T.; Beek, L.; Huylebroeck, D.; Zwijsen, A.; Merregaert, J. The Extracellular Matrix 1 Gene Is Essential for Early Mouse Development. FEBS J. 2005, 272, 571. [Google Scholar]
  81. Liu, Z.; Kim, J.H.; Falo, L.D.; You, Z. Tumor Regulatory T Cells Potently Abrogate Antitumor Immunity. J. Immunol. 2009, 182, 6160–6167. [Google Scholar] [CrossRef] [Green Version]
  82. Maul, J.; Loddenkemper, C.; Mundt, P.; Berg, E.; Giese, T.; Stallmach, A.; Zeitz, M.; Duchmann, R. Peripheral and Intestinal Regulatory CD4+CD25high T Cells in Inflammatory Bowel Disease. Gastroenterology 2005, 128, 1868–1878. [Google Scholar] [CrossRef] [PubMed]
  83. Marazuela, M.; García-López, M.A.; Figueroa-Vega, N.; De La Fuente, H.; Alvarado-Sánchez, B.; Monsiváis-Urenda, A.; Sánchez-Madrid, F.; Gonzalez-Amaro, R. Regulatory T Cells in Human Autoimmune Thyroid Disease. J. Clin. Endocrinol. Metab. 2006, 91, 3639–3646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Vocanson, M.; Hennino, A.; Cluzel-Tailhardat, M.; Saint-Mezard, P.; Benetiere, J.; Chavagnac, C.; Berard, F.; Kaiserlian, D.; Nicolas, J.F. CD8+ T Cells Are Effector Cells of Contact Dermatitis to Common Skin Allergens in Mice. J. Investig. Dermatol. 2006, 126, 815–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Shapira, E.; Brodsky, B.; Proscura, E.; Nyska, A.; Erlanger-Rosengarten, A.; Wormser, U. Amelioration of Experimental Autoimmune Encephalitis by Novel Peptides: Involvement of T Regulatory Cells. J. Autoimmun. 2010, 35, 98–106. [Google Scholar] [CrossRef] [PubMed]
  86. Zhang, Y.; Bandala-Sanchez, E.; Harrison, L.C. Revisiting Regulatory T Cells in Type 1 Diabetes. Curr. Opin. Endocrinol. Diabetes. Obes. 2012, 19, 271–278. [Google Scholar] [CrossRef] [PubMed]
  87. Fontenot, J.D.; Gavin, M.A.; Rudensky, A.Y. Foxp3 Programs the Development and Function of CD4+CD25+ Regulatory T Cells. Nat. Immunol. 2003, 4, 986–992. [Google Scholar] [CrossRef]
  88. Prager, M.; Buettner, J.; Buening, C. Genes Involved in the Regulation of Intestinal Permeability and Their Role in Ulcerative Colitis. J. Dig. Dis. 2015, 16, 713–722. [Google Scholar] [CrossRef]
  89. Adali, G.; Tunali, N.E.; Yorulmaz, E.; Tiryakioǧlu, N.O.; Mungan, S.G.; Ulaşoǧlu, C.; Enç, F.Y.; Tuncer, I. Extracellular Matrix Protein 1 Gene Rs3737240 Single Nucleotide Polymorphism Is Associated with Ulcerative Colitis in Turkish Patients. Turk. J. Gastroenterol. 2017, 28, 254–259. [Google Scholar] [CrossRef] [Green Version]
  90. Su, P.; Chen, S.; Zheng, Y.H.; Zhou, H.Y.; Yan, C.H.; Yu, F.; Zhang, Y.G.; He, L.; Zhang, Y.; Wang, Y.; et al. Novel Function of Extracellular Matrix Protein 1 in Suppressing Th17 Cell Development in Experimental Autoimmune Encephalomyelitis. J. Immunol. 2016, 197, 1054–1064. [Google Scholar] [CrossRef] [Green Version]
  91. Lee, K.M.; Nam, K.; Oh, S.; Lim, J.; Kim, R.K.; Shim, D.; Choi, J.H.; Lee, S.J.; Yu, J.H.; Lee, J.W.; et al. ECM1 Regulates Tumor Metastasis and CSC-like Property through Stabilization of β-Catenin. Oncogene 2015, 34, 6055–6065. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, L.; Yu, J.; Ni, J.; Xu, X.-M.; Wang, J.; Ning, H.; Pei, X.-F.; Chen, J.; Yang, S.; Underhill, C.B.; et al. Extracellular Matrix Protein 1 (ECM1) Is over-Expressed in Malignant Epithelial Tumors. Cancer Lett. 2003, 200, 57–67. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Stage-specific clinical and pathological features of female genital LS. In the early clinical stage of LS (left columns), it initiates as erythema and mild erosion, mostly covering the entire peri-vaginal and perianal area. Pathologically, early LS shows parakeratotic scales, irregular epidermal thickening, and intense inflammatory infiltrates in the upper dermis with faint homogenization of dermal collagen bundles. The condition fluctuates and gradually exacerbates into persistent erosions with focal blistering and induration, leading to a whitish-pale appearance in the middle clinical stage (middle columns). Note that dermal hyalinosis is more apparent with dilated blood vessels. The chain of these inflammatory events finally results in scarring and irreversible adhesion of external genital parts, and abruptly develops squamous cell carcinoma in the late clinical stage (right columns). The preexisting dermal hyalinosis in the skin pathology further extends diffusely, becoming more prone to eosinophilic staining and a thinner color. Dilated blood vessels in the upper dermis look atrophic, narrowing in size with thickening vessel walls.
Figure 1. Stage-specific clinical and pathological features of female genital LS. In the early clinical stage of LS (left columns), it initiates as erythema and mild erosion, mostly covering the entire peri-vaginal and perianal area. Pathologically, early LS shows parakeratotic scales, irregular epidermal thickening, and intense inflammatory infiltrates in the upper dermis with faint homogenization of dermal collagen bundles. The condition fluctuates and gradually exacerbates into persistent erosions with focal blistering and induration, leading to a whitish-pale appearance in the middle clinical stage (middle columns). Note that dermal hyalinosis is more apparent with dilated blood vessels. The chain of these inflammatory events finally results in scarring and irreversible adhesion of external genital parts, and abruptly develops squamous cell carcinoma in the late clinical stage (right columns). The preexisting dermal hyalinosis in the skin pathology further extends diffusely, becoming more prone to eosinophilic staining and a thinner color. Dilated blood vessels in the upper dermis look atrophic, narrowing in size with thickening vessel walls.
Diagnostics 12 03070 g001
Figure 2. Clinical features of LS developed on the extragenital skin. The extragenital LS occurs at any skin sites, including extremities (upper left and right), scalp (lower left), and trunk (lower right).
Figure 2. Clinical features of LS developed on the extragenital skin. The extragenital LS occurs at any skin sites, including extremities (upper left and right), scalp (lower left), and trunk (lower right).
Diagnostics 12 03070 g002
Figure 3. Clinicopathology of lipoid proteinosis (an ECM1-lacking genodermatosis: upper panels) and lichen sclerosus (an anti-ECM1 autoantibody-carrying condition in females and males; left and right in the lower panels, respectively). Irrespective of a predilection to different skin sites between the two diseases, their skin pathologies display similar features (right columns), including packed and parakeratotic hyperkeratosis, epidermal atrophy, and diffuse hyaline changes and dilated blood vessels in the upper dermis. Of these, dermal hyalinosis is a hallmark of both diseases.
Figure 3. Clinicopathology of lipoid proteinosis (an ECM1-lacking genodermatosis: upper panels) and lichen sclerosus (an anti-ECM1 autoantibody-carrying condition in females and males; left and right in the lower panels, respectively). Irrespective of a predilection to different skin sites between the two diseases, their skin pathologies display similar features (right columns), including packed and parakeratotic hyperkeratosis, epidermal atrophy, and diffuse hyaline changes and dilated blood vessels in the upper dermis. Of these, dermal hyalinosis is a hallmark of both diseases.
Diagnostics 12 03070 g003
Figure 4. Multifocal interaction of in vivo ECM1 with surrounding extracellular matrix and structural molecules in the skin. ECM1 is expressed in the major skin components (particularly the epidermal basal layer and basement membranes) and adjunct appendages (blood vessel walls and follicular epithelium), as immunostained in normal human skin (left panel), and regulates the different in vivo turnover and feedback productivity of binding partners as a ‘biological glue’, contributing to the integrity and maintenance of skin homeostasis (right panel).
Figure 4. Multifocal interaction of in vivo ECM1 with surrounding extracellular matrix and structural molecules in the skin. ECM1 is expressed in the major skin components (particularly the epidermal basal layer and basement membranes) and adjunct appendages (blood vessel walls and follicular epithelium), as immunostained in normal human skin (left panel), and regulates the different in vivo turnover and feedback productivity of binding partners as a ‘biological glue’, contributing to the integrity and maintenance of skin homeostasis (right panel).
Diagnostics 12 03070 g004
Figure 5. Schematic image for autoimmune and genetic impairment of ECM1 function in the skin. Genetic ablation and autoantibody targeting of the skin ECM1 cause dysregulation of its binding partners, as listed in Figure 3, contributing to the homeostatic imbalance or collapse in the epidermis (dyskeratosis and atrophy), dermis (collagen homogenization and sclerosis), and blood vessels (telangiectasia and impermeability). The chain of these statements finally establishes the pathological features seen in LiP and LS.
Figure 5. Schematic image for autoimmune and genetic impairment of ECM1 function in the skin. Genetic ablation and autoantibody targeting of the skin ECM1 cause dysregulation of its binding partners, as listed in Figure 3, contributing to the homeostatic imbalance or collapse in the epidermis (dyskeratosis and atrophy), dermis (collagen homogenization and sclerosis), and blood vessels (telangiectasia and impermeability). The chain of these statements finally establishes the pathological features seen in LiP and LS.
Diagnostics 12 03070 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Oyama, N.; Hasegawa, M. Lichen Sclerosus: A Current Landscape of Autoimmune and Genetic Interplay. Diagnostics 2022, 12, 3070. https://doi.org/10.3390/diagnostics12123070

AMA Style

Oyama N, Hasegawa M. Lichen Sclerosus: A Current Landscape of Autoimmune and Genetic Interplay. Diagnostics. 2022; 12(12):3070. https://doi.org/10.3390/diagnostics12123070

Chicago/Turabian Style

Oyama, Noritaka, and Minoru Hasegawa. 2022. "Lichen Sclerosus: A Current Landscape of Autoimmune and Genetic Interplay" Diagnostics 12, no. 12: 3070. https://doi.org/10.3390/diagnostics12123070

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop