Next Article in Journal
Differentiation and Growth-Arrest-Related lncRNA (DAGAR): Initial Characterization in Human Smooth Muscle and Fibroblast Cells
Previous Article in Journal
Thanatin: A Promising Antimicrobial Peptide Targeting the Achilles’ Heel of Multidrug-Resistant Bacteria
Previous Article in Special Issue
shRNA-Targeting Caspase-3 Inhibits Cell Detachment Induced by Pemphigus Vulgaris Autoantibodies in HaCaT Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathological Mechanisms Involved in Epidermolysis Bullosa Simplex: Current Knowledge and Therapeutic Perspectives

by
Mbarka Bchetnia
1,2,
Julie Powell
3,
Catherine McCuaig
3,
Anne-Marie Boucher-Lafleur
1,2,
Charles Morin
4,
Audrey Dupéré
4 and
Catherine Laprise
1,2,*
1
Département des Sciences Fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada
2
Centre Intersectoriel en Santé Durable, Saguenay, QC G7H 2B1, Canada
3
CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
4
Centre Intégré Universitaire de Santé et de Services Sociaux du Saguenay-Lac-Saint-Jean, Hôpital Universitaire de Chicoutimi, Saguenay, QC G7H 7K9, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(17), 9495; https://doi.org/10.3390/ijms25179495 (registering DOI)
Submission received: 15 July 2024 / Revised: 22 August 2024 / Accepted: 29 August 2024 / Published: 31 August 2024
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Skin Diseases)

Abstract

:
Epidermolysis bullosa (EB) is a clinically and genetically heterogeneous group of mechanobullous diseases characterized by non-scarring blisters and erosions on the skin and mucous membranes upon mechanical trauma. The simplex form (EBS) is characterized by recurrent blister formation within the basal layer of the epidermis. It most often results from dominant mutations in the genes coding for keratin (K) 5 or 14 proteins (KRT5 and KRT14). A disruptive mutation in KRT5 or KRT14 will not only structurally impair the cytoskeleton, but it will also activate a cascade of biochemical mechanisms contributing to EBS. Skin lesions are painful and disfiguring and have a significant impact on life quality. Several gene expression studies were accomplished on mouse model and human keratinocytes to define the gene expression signature of EBS. Several key genes associated with EBS were identified as specific immunological mediators, keratins, and cell junction components. These data deepened the understanding of the EBS pathophysiology and revealed important functional biological processes, particularly inflammation. This review emphasizes the three EBS subtypes caused by dominant mutations on either KRT5 or KRT14 (localized, intermediate, and severe). It aims to summarize current knowledge about the EBS expression profiling pattern and predicted molecular mechanisms involved and to outline progress in therapy.

1. Introduction

Epidermolysis bullosa simplex (EBS) affects the skin, a vital organ with a dynamic and complex organization protecting the body from stress conditions [1,2]. Most cases of EBS result from autosomal dominant mutations affecting either keratin 5 (KRT5) or KRT14 [3]. Under normal conditions, the corresponding proteins, keratin 5 (K5) and 14 (K14), form heterodimers assembling together into intermediate filaments (IFs) [3]. In EBS, causal mutations can impact the structure of these proteins, causing keratin aggregates in keratinocytes, triggering endoplasmic reticulum stress and leading to the release of pro-inflammatory molecules [4]. Intraepidermal ruptures occur and lead to loss of tissue integrity and severe blistering of the epidermis upon mechanical trauma or traction [5,6,7,8]. A schematic representation of the structural impairments leading to EBS symptoms can be visualized by Li et al. [9]. The keratin defects in EBS also trigger itching, causing further discomfort while also yielding new lesions by scratching the skin and, thus, promoting inflammation [10,11]. The severity of EBS is highly variable with three main clinical subtypes: localized EBS (EBS-loc), intermediate EBS (EBS-intermed), and severe EBS (EBS-sev) [12,13]. Figure 1 represents the characteristics of these different subtypes. Rare subtypes of EBS exist, for which other causal mutations were identified in several genes (plectin (PLEC), Kelch-like member 24 (KLHL24), dystonin (DST), exophilin-5 (EXPH5), and CD151 molecule (CD151)) [11,12].
To deepen the characterization of the molecular mechanisms underlying the EBS phenotype, several gene expression profiling studies were performed and reported varying and sometimes conflictual findings [14,15,16,17,18,19,20,21,22,23,24], highlighting the need to discuss these results in a single paper. Expression profiles are tissue-specific, thus allowing an accurate representation of pathway dysregulation in the infected tissue of the EBS patients—the skin [25]. Inflammation mediators and other components critical for immune response and skin barrier homeostasis, including keratinocyte proliferation, differentiation, and migration, are highly upregulated in many expression profile studies [14]. The common agreement that surfaced is the potential implication of inflammation in EBS pathogenesis [19,26,27] as well as the possibilities of efficient treatment development.
This review gathers and discusses the findings of EBS transcriptomic studies regarding three main clinical subtypes (EBS-loc, EBS-intermed, EBS-sev). Expression studies performed under normal conditions were selected, with special attention to those also proposing therapeutic avenues. The following keywords were used to search the Pubmed database: epidermolysis bullosa, gene expression, transcriptomic analysis, gene therapy, and EBS treatment.

2. EBS Gene Expression Profile Studies

2.1. Increase of Inflammation Components

Inflammatory processes are triggered by an immune system disturbance generally caused by tissue injury, infection, or other stresses. A variety of immune cell types migrate, proliferate, and produce cytokines, interleukins (ILs), and interferons (IFNs) in an orchestrated manner at the inflammation site [28,29]. Aside from the keratin defaults triggering EBS, it becomes clear that this disorder is associated with inflammation as reported by transcriptomic analysis. Various signaling inflammatory pathways were shown to be involved in the EBS pathomechanisms.

2.1.1. c-Jun N-Terminal Kinase (JNK) Stress Pathway

In the skin, keratinocytes produce pro-inflammatory cytokines IL-1α and IL-1β and their receptors [30]. These ILs activate keratinocytes in various pathological conditions and upon tissue damage [14]. IL-1β precursor is present in the cytoplasm and, after injury, is released and activates signal transduction pathways in surrounding cells [4]. IL-6 stimulates keratinocyte growth and migration in vitro mediate local inflammation and modulate immune response upon wounding along with IL-1β [31].
Transcriptome analysis revealed that IL-1β and IL-6 mRNA levels were upregulated in K5−/− mouse skin [14]. Increased expression of IL-1β was also shown in keratinocytes derived from one patient with EBS-sev [23,30]. Upregulation of IL-6, IL-8, and IL-10 was also observed in the epidermis of patients with either KRT5 or KRT14 mutations [24]. This is consistent with local inflammation that can accompany EBS lesions. The c-Jun N-terminal kinase (JNK) stress pathway is activated through IL-1β in an autocrine manner [30]. This activates the transcription of AP-1 responsive genes such as matrix metalloproteinase 1 (MMP1), kallikrein 7 (KLK7), rho guanine nucleotide exchange factors (ARHGEF family), and KRT14 and IL-1β themselves by positive feedback [20]. When EBS is caused by KRT14 mutations, expression of the dominantly interfering mutated allele would increase through this mechanism and potentially aggravate the phenotype. Increased expression of MMPs (1, 7, 9, 13, 19) in EBS-sev keratinocytes was reported, as well as KLKs (5–8, 10–11, 13–14), indicating that various genes encoding matrix-degrading proteins are targets of the IL-1β pathway [21]. KLK5 and KLK7 differential expression was validated on the protein level by Western blot [21]. This highlights the role of inflammation in exacerbating the EBS phenotype, particularly in EBS-sev.

2.1.2. IFN-γ Inflammatory Signaling Pathway

IFNs are major mediators of inflammation [32]. They are pleiotropic cytokines with antiviral, antitumor, and immunomodulatory functions and act as major effectors of the immune response [33]. Patients with EBS display higher systemic levels of inflammatory mediators such as IFN-γ, primarily secreted by NK cells and activated T lymphocytes [4]. Badowski et al. showed that the addition of IFN-γ to EBS-sev keratinocytes promotes the formation of protein aggregates, decreases cell–cell junctions, delays wound closure, and reduces cell proliferation as part of an immune antiviral response [34]. IFN-γ increases the phospho-Tyr701 signal transducer and activator of transcription (STAT) 1, which lies downstream in the IFN-γ inflammatory signaling pathway [35], as well as K17 as a stress response to reduce the dominant negative effect of mutated K14 possibly by copolymerizing with K5 in vivo [21,36].

2.1.3. Phosphatidylinositol 3-Kinase (PI3K)-Protein Kinase B (Akt)-mTOR Pathway

The mechanistic target of rapamycin (mTOR) is a member of the phosphatidylinositol 3-kinase (PI3K)-related kinase family. It is stimulated by the protein kinase B (Akt), immediately downstream of PI3K. The PI3K/Akt/mTOR pathway regulates cell growth, proliferation, motility, and survival [37]. A recent study carried out transcriptomic analysis in EBS patients to characterize the molecular pathways activated in the EBS-blistered epidermis. Their findings revealed that IL-6, IL-8, and IL-10 were upregulated in the epidermis of EBS patients, and predicted upstream regulators included TNF-a, IL-1b, IL-2, IL-6, PI3K, and mTOR. They hypothesized that this signaling process is one of the major activated pathways in EBS [24].

2.1.4. Wnt-Receptor Signaling Pathway

Enrichment clustering of dysregulated genes in whole-transcriptome microarray analysis of an EBS-sev cell line highlighted the upregulation at the RNA and protein levels of WNT5a, encoding for one of the so-called noncanonical Wnt ligands [22]. During normal development, WNT5a is secreted and directs the migration of target cells along concentration gradients. Dysregulation of WNT5a signaling facilitates invasion by multiple tumor types into contiguous tissues [38]. It is known that patients with EBS-sev have a slightly higher risk of developing basal cell carcinoma (BCC) [4], and dysregulation in WNT5a signaling could contribute to this outcome. The roles of WNT5a in this context have not been fully investigated yet nor its impact on keratinocytes migration.

2.1.5. Bone Morphogenetic Proteins Signaling (BMPs)

Bone morphogenetic proteins (BMPs) are signaling polypeptides of the transforming growth factor-β (TGF-β) superfamily acting as important multifactor players in the development of vertebrate skin and appendages. BMP signaling acts as a potent tumor suppressor in the skin, inhibiting mainly epidermal-derived and hair follicule-derived tumor formation [39]. Consequently, dysregulation of this pathway can lead to abnormal skin development and tumor formation [40]. A downregulation of three genes—GPC3 (Glypican 3), MSX2 (Msh homeobox 2), and ILK (Integrin-linked kinase)—involved in BMP signaling was observed in a whole-transcriptome microarray analysis of EBS-sev immortalized cells [22].

2.1.6. T Helper Type 17 (Th17) Immune Response

T helper type 17 (Th17) cells play a role in adaptive immunity by protecting the body against pathogens by producing antimicrobial peptides, recruiting immunocytes via induction of chemokines, such as Chemokine (C-C motif) ligand 20 (CCL20), and repairing tissue by enhancing epithelial proliferation. The Th17 signaling pathway is tightly controlled and is terminated after infection is ablated and tissue repair is completed [41,42]. Higher levels of Th17 immune response markers such as IL-17, IL-21, and IL-22 were observed in the blister roof of patients with EBS-sev compared with controls by analysis of cytokine mRNA expression. Higher expression of TNFα, CCL20, IL-5, and IL-22 was also observed in the blister fluid. Moreover, blister roofs and fluid from patients with EBS-sev showed an increase in other cytokines inducing the differentiation of T cells toward Th17 cells, such as TGF-β, IL-6, and IL-21 [26]. Altogether, these results strongly suggest the involvement of the Th17 immune response in the EBS pathogenesis.

2.2. Keratins and Cell-Junction Components

Microarray experiments identified 129 differentially expressed probes/genes linked to cell–substrate or cell–cell junctions and/or other components of the cytoskeleton dysregulated in EBS keratinocytes. About 20 of these genes, encoding for cell junction components, including desmosomes, hemidesmosomes, adherent junctions, tight junctions, and gap junctions, are downregulated in the EBS-sev cell line compared with controls, with four of them also validated on the protein level [16]. Another study reported an upregulation of the two most abundant desmosomes components, desmoglein 3 (DSG3) and desmoplakin (DSP), in EBS-sev cells and interpreted this finding as a cellular response aiming at increasing the number and internal integrity of desmosomes [18]. Desmosomes are crucial for the integrity of the skin as they ensure mechanical strength by providing strong adhesion between cells. Several type 1 (K14, K16, K17) and type 2 (K5, K6a, K6b) keratins were upregulated in EBS-sev cells [18]. Another EBS expression microarray analysis on the epidermis of patients with EBS-sev identified several upregulated genes involved in epidermal keratinization, such as SPRR4 and KRT79 [20]. KRT19 was significantly downregulated in one microarray analysis of EBS-sev keratinocytes versus controls indicating that KRT19 could constitute another useful biomarker in EBS [22].
The expression profile of EBS represents a lengthy list of dysregulated genes. Table 1 summarizes the results obtained from all studies cited above.

3. Strategies to Improve and Alleviate EBS Manifestations

EBS remains one of the most well-understood inherited bullous diseases. However, it represents a challenge for treatment because of the dominant impact of keratin mutations and the broad number of mutations affecting either KRT5 or KRT14 (more than 200) [43]. The knowledge generated regarding the potential of inflammation components as therapeutic targets and the explosion of omics data combined with new genome editing approaches provided powerful insights for EBS. Added to symptom relief therapies, approaches targeting causative genetic defects are in development more than ever with the aim of curing or treating this disease. Table 2 regroups information regarding the therapeutic molecules tested in the context of EBS.

3.1. Therapeutic Molecules: Anti-Inflammatory

The skin abnormalities in EBS are associated with inflammation and immune reaction via the production of chemokines and cytokines. This indicates possible therapeutic perspectives for decreasing chronic inflammation. Figure 2 offers an overview of key inflammatory pathways in EBS, identified in Section 2 of the present review, and the associated therapeutic approaches discussed below.

3.1.1. Tetracycline Antibiotics

Tetracyclines are broad-spectrum bacteriostatic antibiotics used in dermatology for treating acne and are well known for inhibiting bacterial protein synthesis [44,45,46]. Tetracyclines are pluripotent drugs affecting various cellular functions, such as prion infectivity through direct interaction with proteinase-resistant forms of the prion protein [47]. One specific tetracycline, doxycycline, is an inhibitor of cell proliferation that modulates smooth muscle cell growth and matrix remodeling after arterial injury [48].
One clinical study suggests that oral treatment with tetracycline on patients with EBS-sev led to a significant reduction of blistering and less fragile epidermis [14,49]. Another trial on the K5−/− mouse model observed downregulation of matrix MMP13 and IL-1β expression and extended the survival of mice from less than one to up to eight hours when doxycycline was applied [14,50]. This shows the potential effect of doxycycline on transcription and its function as a potent MMP inhibitor, which could also be observed in a human model.

3.1.2. Diacerein

Diacerein, is reported to block the release of active IL-1β by inhibiting plasma membrane-bound IL-1 converting enzyme (ICE) [51]. Treatment of keratinocytes from patients with EBS-sev with diacerein reduced the expression levels of K14 and IL-1β and the phosphorylation levels of JNK and, also, stabilized the intermediate IFs network upon heat shock [30]. One percent diacerein cream was tested for the topical treatment of EBS-sev in a pilot [52] and a phase II/III clinical trial and produced promising results with a reduction of more than 40% in blisters despite its transient effect [53]. A recent study reported no significant differences between the efficiency of diacerein 1% and placebo for patients with EBS, even when stratifying by severity [54]. These discordant results highlight the need for further investigations before stating on its efficiency as a reliable treatment for EBS clinical manifestations.

3.1.3. Humanized IFN-γ Blocking Antibody

Therapeutic approaches aiming to block IFN-γ function might be a strategy for reducing skin fragility as it is usually increased in skin lesions. IFN-γ blocking antibodies were tested for the treatment of skin disorders, including dystrophic EB [55]. One mouse antibody targeting IFN-γ was successfully humanized without loss of affinity and applied to a cell model of EBS-sev. Results showed restoration of cell proliferation, increased cell–cell adhesion, accelerated wound closure in the presence of IFN-γ, and reduced IFN-γ mediated keratin aggregation [34,56]. This antibody represents a potentially valuable therapeutic agent for clinical trials as it is already humanized and can be directly administered in patients with EBS in effective doses, preventing side effects.

3.1.4. mTOR Inhibitors

Using transcriptomic data for drug repositioning yielded novel treatment avenues for other skin conditions such as hidradenitis suppurativa [57], basal cell carcinoma [58], and dermatomyositis [59]. This strategy of selecting the best potential drugs whose effects on transcription would oppose the EBS signature led to the identification of the mTOR inhibitors as the best candidates. An independent pilot study of two patients with EBS treated with topical sirolimus for painful plantar keratoderma due to chronic blistering was conducted. Marked clinical improvement and a notable reduction of keratoderma were observed in EBS patients [24]. Sirolimus is known for its immunosuppressive properties. It forms a complex with FK binding proteins (FKBPs), ultimately binding with mTOR and inhibiting its function [60].

3.1.5. 4-Phenyl Butyric Acid (4-PBA)

4-phenyl butyric acid (4-PBA) is a chemical chaperone facilitating protein folding which has been shown to reduce keratin aggregation in immortalized EBS-loc and EBS-sev primary keratinocytes after heat shock [61,62]. It was also reported that 4-PBA treatment led to a reduction of about 40% of the number of EBS-sev keratinocytes containing cytoplasmic keratin aggregates, highlighting the role of endogenous chaperone molecules in the degradation of mutated keratin molecules. Monitoring of ERK activation by western-blot analysis indicated that 4-PBA has a positive effect on ERK activation and reduces IL-1β expression [23,63].

3.1.6. Apremilast (Anti-IL-17 Agent)

The presence of an immune infiltrate characterized by a Th17 response in the skin of EBS patients suggested that inhibitors of this pathway may be a promising new treatment [26]. Apremilast is a small molecule approved for the treatment of psoriasis, specifically inhibiting cyclic AMP phosphodiesterase-4 and Th1/Th17 activation [64,65]. Castela et al. 2019 tested the potential therapeutic benefits of apremilast in a small pilot study of three individuals with EBS-sev. They reported a rapid and sustained decrease in the number of blisters in all patients. Clinical trials should be the next step toward using this agent as a standard treatment for EBS patients.

3.1.7. Afatinib (Epidermal Growth Factor Receptor (EGFR) Inhibitor)

Several pro-inflammatory cytokines, indicators of stress and wound-like activation of EBS keratinocytes, were found to be elevated in blister fluid from patients with EBS [66]. These include the epidermal growth factor (EGF) and its receptor EGFR, acting in a signaling pathway that is essential for keratinocytes migration and wound closure [67,68]. Treatment of EBS-sev cells in vitro with the FDA-approved EGFR inhibitor afatinib reduced keratin aggregates and favored stable filament networks. Afatinib treatment reverted the wound activation and healing migration state of EBS-sev disease model cells to a more quiescent state associated with improved keratin filament networks and cell–cell connections. This should improve the mechanical resilience of the epidermis and provide stability and resistance to mechanical stress [69].

3.2. Therapeutic Molecules: Beyond Anti-Inflammatory Mechanisms

Direct targeting of cellular structural abnormalities or lesions is another treatment strategy employed in EBS. Some of these molecules have the ability to directly influence post-translational mechanisms acting on pathways leading to impaired cellular integrity.

3.2.1. Aluminium Chloride Hexahydrate

Aluminum chloride hexahydrate efficiency was first tested in two different case reports. These studies reported it as an effective treatment to prevent blisters in patients with EBS-loc [70,71]. A double-blind placebo-controlled randomized study on the administration of aluminum chloride hexahydrate in the same manner (20% solution in anhydrous ethyl alcohol) yielded no significant results regarding blister numbers in patients with EBS-loc [72]. The mechanisms underlying the possible success in two cases remain unclear and no further positive results were obtained for this molecule in EBS.

3.2.2. Botulinum Toxins (BoNT)

Botulinum toxins (BoNT) are produced by the bacteria of the genra Clostridium. Their action inhibits the release of acetylcholine (ACh) and stops the signal for motion sent to muscle fibers [73]. BoNT, particularly type A and B (BTX-A and BTX-B), were used for treating the feet of patients with EBS-loc and EBS-sev in two different studies [74,75]. These studies showed improvement in blistering and alleviated pedal pain. One case report on a pediatric patient with EBS also showed pedal improvement for three parameters (blistering, pain, and odor) [76]. It is known that BoNT can have an effect on sensory nerves and, thus, affect the tenderness of the feet of patients with EBS, which seemingly helps diminish blister formation.

3.2.3. Parthenolide (PN)

One plant derivate, parthenolide (PN), is known as a sesquiterpene lactone found in feverfew. PN possesses anti-inflammatory, redox-modulating, and epigenetic properties as well as selective toxicity for specific cancer cells [77]. A study by Sun et al. identified PN by high-throughput drug screening of molecules stabilizing IFs. They showed that PN normalized acetylation on keratins through augmented binding with NAD-dependent sirtuin 2 (SIRT2) [78]. This linked the acetylation pathways to the IF organization in a mice cellular model of EBS where disruption of keratin filaments is triggered by a mutation on KRT14. This resulted in increased cell adhesion and resistance to mechanical stress [78]. It is possible to assume that some clinical manifestations of EBS, such as blistering, could be alleviated with PN. Clinical trials are still needed before the effect on the cells and skin of EBS patients is proven.

3.2.4. PKC412 Kinase Inhibition

This kinase is also known as midostaurin, a targeted therapy used in combination with chemotherapy for the treatment of acute myeloid leukemia [79]. This molecule has also been identified as normalizing the level of disruption of the keratin filament caused by a mutation in KRT18 in mice [80]. Following this rationale, Rietscher et al. used this molecule to reduce keratin aggregates in keratinocytes derived from patients with a causal mutation on KRT14. Results showed a lowering of 40% in keratin aggregates as well as lesser phosphorylation on keratin, both leading to strengthened cellular cohesion [81]. This kinase directly affects the phosphorylation level of keratins because of the high accessibility of the non-α-helical domains in the head and tail of keratins [81,82]. This lowered phosphorylation, in turn, allows for a better organization of intracellular keratin filaments and indirectly affects DSP, a protein useful in cell–cell contact sites [81]. This modification of a post-translational mechanism holds promising outcomes for EBS treatment.
Table 2. Molecules tested in epidermolysis bullosa simplex (EBS).
Table 2. Molecules tested in epidermolysis bullosa simplex (EBS).
Molecule ReferenceType of StudyPatientsVehicleTargetOutcomesPerspectives
4-PBA[62]In vitroEBS-loc, EBS-sev1 mM solutionImmortalized keratinocytesReduction of keratin aggregates upon heat shockReversing protein aggregates and reducing tissue fragility
[63]In vitroEBS-sev1 mM solutionImmortalized keratinocytesReduction of keratin aggregates and amelioration of inflammatory phenotypeAssay with low doses to avoid toxicity
[23]In vitroEBS-sev1 μM solutionKeratinocytesReduction of around 40% of keratin aggregatesReversing protein aggregates and reducing tissue fragility
Afatinib[69]In vitroEBS-sev1 μM solutionImmortalized keratinocytesReduction of keratin aggregates, induction of quiescent state to cellsOptimize afatinib for therapy (reduce adverse side effects when used in cancer therapy)
Aluminium chloride hexahydrate[70]Case reportEBS-loc20% in alcoholFeetPrevent new blistersStudy other cases to establish a pattern of efficiency
[71]Case reportEBS-loc20% in alcoholHands and feetPrevent new blistersStudy other cases to establish a pattern of efficiency
[72]Therapeutic assayEBS-loc20% in alcoholFeetNo significant reduction of blistersStudy the effect on more severe subtypes
Apremilast[26]Therapeutic assayEBS-sevOral treatment (10 mg/day to 30 mg twice/day)SkinRapid and sustained (7–10 months) improvement in skin lesionsTrial with a higher number of patients (placebo–control)
Botulinum toxins[74]Case reportEBS-loc100 U BTX-A (Botox) in salineFeet65% reduction in blister surface area, decrease in pain and perspirationTrial with a higher number of patients (placebo–control), testing higher concentration of the molecule
[75]Therapeutic assayEBS-loc, EBS-sev170–700 U BTX-A (Dysport) in saline, 2500 U BTX-B (Neurobloc) in salineFeetImprovement in blistering and pedal painTrial with a higher number of patients (placebo–control)
[76]Case reportEBS100 U BTX-A (Botox) in salineFeetReduction of blisters, smaller blisters, decreased pedal pain and odorTreatment for pain management and improved quality of life
Diacerein[30]In vitroEBS-sev10 μg mL−1 solutionKeratinocytesStabilization of the IF network and reduction of inflammatory componentsTrial with EBS-sev patients
[52]Therapeutic assayEBS-sev1% creamArmpitsReduction of blistersTrial with a higher number of patients
[53]Phase 2/3 clinical trialEBS-sev1% creamSkinMore than 40% reduction in blister numberTrial with a higher number of patients and more invasive data acquisition
[54]Therapeutic assayEBS-sev, EBS-intermed1% creamSkinNo significant improvement compared with vehicule cream aloneFocus on EBS-sev individuals with a bigger sample size
Doxycycline[14]In vivoK5−/− mice50 μg/mL solution with 5% sucroseSkinDownregulation of MMPP13 and IL-1βScreening similar compounds for additional targets
INF-γ blocking antibodies[34]In vitroCell model of EBS-sevHumanized monoclonal antibodyKeratinocytesReversing effect of IFN-γ (less keratin aggregates, restored cell proliferation, increased cell–cell adhesion, accelerated wound closure)Promising therapy for patients with EBS or other skin disease (in vivo assays)
mTOR inhibitors[24]Therapeutic assayEBS 2% sirolimus ointmentFeetReduction of blisters and keratodermaDrug repositioning based on transcriptomic signature in EBS
Parthenolide[78]In vitroMice cell model of EBS5 µM solutionKeratinocytesIncreasing cell adhesion and resistance to mechanical stressTrials with keratinocytes of patients to assess efficiency
PKC412 kinase inhibitor[81]In vitroEBS-sev1 µM solutionImmortalized keratinocytesReduction of 40% of keratin aggregates and strengthening of cellular cohesionPretesting on animal models and skin explants before oral or topic administration in EBS patients
Tetracycline[49]Therapeutic assayEBS-sevOral treatment (1500 mg/day)SkinReduction of blisters and less fragile epidermis (dose-dependant response)Trial with a higher number of patients
Abbreviations—BTX-A: botulinum toxin type-A; BTX-B: botulinum toxin type-B; EBS-intermed: intermediate EBS; EBS-loc: localized EBS; EBS-sev: severe EBS; IL-1β: interleukin-1 β; MMPP13: matrix metallopeptidase 1.

3.3. Genome Editing Approaches

3.3.1. RNA Trans-Splicing

Studies using spliceosome-mediated mRNA trans-splicing (SMaRT) have proved its feasibility to correct mutations at the mRNA level by replacing up to 4.5 kb of a sequence [83]. The basis of SMaRT is the engineering of an RNA trans-splicing molecule (RTM) containing the coding region to be replaced. This method was used to correct a mutation in exon 1 of KRT14 by using a previously reported 5′-trans-splicing module replacing exons 1–7 of the KRT14 mRNA [84,85], Accordingly, the RNA trans-splicing module was transduced into an EBS-sev patient-derived keratinocyte cell line. Subsequently, skin equivalents were generated with these corrected cells and grafted onto the backs of mice. These skin equivalents revealed a stable, well-differentiated epidermis, thus showing the potential of trans-splicing as a therapeutic approach for EBS. The major advantage of trans-splicing is that one RNA trans-splicing module can be employed to correct multiple variants in several exons, eliminating the need to design specific RTM for each mutation.

3.3.2. Transcription Activator-like Effector Nucleases (TALENS)

Programmable nucleases, named transcription activator-like effector nucleases (TALENs), are an approach consisting of the induction of DNA double-strand breaks (DSBs). This activates the endogenous repair machinery, resulting in gene disruption via nonhomologous end joining (NHEJ). Aushev et al. showed that TALENs efficiently disrupted the KRT5 locus in immortalized EBS-intermed and EBS-sev human epidermal keratinocytes, with 25% and 29% of clones showing NHEJ, respectively [66]. Inactivation of the mutant KRT5 allele resulted in the elimination of structural abnormalities in IFs and the reduction of cytoplasmic keratin aggregates upon stress.

3.3.3. CRISPR-Cas9

CRISPR/Cas9 gene editing allows for the introduction of precise changes to specific sequences of genes. It has become a widely used tool in medical research for the correction of disease-associated mutations [86,87,88]. Kocher et al. corrected a causal hotspot mutation in exon 6 of KRT14 resulting in EBS-sev phenotype in vitro [89]. A double-nicking strategy targeting intron 7 was used, followed by homology-directed repair (HDR). Co-delivery into keratinocytes of a Cas9 D10A nickase (Cas9n), a predicted single guide RNA pair specific for intron 7, and a minicircle donor vector harboring the homology donor template resulted in a recombination efficiency of more than 30% and correction of the mutant KRT14 allele without off-target activity.
Bchetnia et al. succeeded in eliminating the mutant protein K5 by disrupting the mutant allele while leaving the wild-type functional [90]. The mutant allele was disrupted for one heterozygous EBS-sev pathogenic variation within KRT5 by introducing a DNA cleavage in the patient’s keratinocytes using a stringent CRISPR-Cas9 system. Results showed successful stringent mutant allele-specific silencing at the DNA and RNA level, indicating permanent mutant allele-specific inactivation. Edited EBS-sev keratinocytes produced a lower amount of K5 and K14 proteins compared with non-edited EBS cells with no disturbance of cellular properties observed.

4. Why Therapeutic Approaches Are Difficult to Find in Epidermolysis Bullosa?

Genetic variations in EBS are caused by several mutations, leading to subtypes with variable severity [12,13]. This genetic variability is challenging for the development of universal therapeutic approaches. Another important challenge is that specific molecular targets across all subtypes of EBS remain elusive due to the diversity of genetic mutations involved (as well as the difficulty of identifying a readout). In addition, the constant presence of wounds in EBS patients can trigger immune responses and inflammation, complicating treatment efforts. Anti-inflammatory approaches might exacerbate these issues, and finding ways to modulate the immune response without causing further damage is complex. Finally, as for other rare disorders, the scarcity of EBS patients means that clinical trials for the development and validation of potential therapies are difficult to establish [91]. The results obtained from existing trials are encouraging but difficult to interpret for all the phenotypes observed. Despite these challenges, new technologies in gene editing, regenerative medicine, high-throughput molecule screening, drug repurposing, and innovative wound care strategies are promising potential therapeutic avenues.

5. Conclusions

This review summarized the differentially expressed mRNAs in EBS and predicted the biological processes and signaling pathways involved in this skin disorder. The cellular pathology of most EBS subtypes is associated with the fragility of the IFs network, cytolysis of the basal layer of the epidermis, or attenuation of hemidesmosomal/desmosomal components. Despite the limited overlap of genes identified, major evidence in the literature supports the implication of inflammation in EBS. Such review will undoubtedly have a bearing on the development of new therapeutic strategies and accelerate the translation of these approaches to the clinic. Although no single therapy has achieved complete success, it seems probable that combining different therapeutic principles will yield the best outcomes and improve the quality of life of patients with EBS.

Author Contributions

Conceptualization, M.B. and C.L.; methodology, M.B. and C.L.; investigation, M.B., A.-M.B.-L. and C.L.; resources, C.L.; writing—original draft preparation, M.B.; writing—review and editing, J.P., C.M. (Catherine McCuaig), C.M. (Charles Morin), A.D., A.-M.B.-L. and C.L.; visualization, M.B. and C.L.; supervision, C.L.; project administration, C.L.; funding acquisition, C.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Canada Research Chair in Genomics of Asthma and Allergic Diseases, grant number CRC-2021-00108.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Catherine Laprise is part of the Quebec AIRS Health Network (RHN; http://rsr-qc.ca/en/, accessed on 29 August 2024), investigator in the CHILD Study, the director of the Centre Intersectoriel en Santé Durable (CISD) of the Université du Québec à Chicoutimi (UQAC) and the chairholder of the Canada Research Chair in the Genomics of Asthma and Allergic Diseases (http://www.chairs.gc.ca, accessed on 29 August 2024) as well as the co-chairholder of the Chaire de Recherche en Santé Durable du Québec.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Simpson, C.L.; Patel, D.M.; Green, K.J. Deconstructing the skin: Cytoarchitectural determinants of epidermal morphogenesis. Nat. Rev. Mol. Cell Biol. 2011, 12, 565–580. [Google Scholar] [CrossRef]
  2. Chambers, E.S.; Vukmanovic-Stejic, M. Skin barrier immunity and ageing. Immunology 2020, 160, 116–125. [Google Scholar] [CrossRef] [PubMed]
  3. Coulombe, P.A.; Kerns, M.L.; Fuchs, E. Epidermolysis bullosa simplex: A paradigm for disorders of tissue fragility. J. Clin. Investig. 2009, 119, 1784–1793. [Google Scholar] [CrossRef] [PubMed]
  4. Evtushenko, N.A.; Beilin, A.K.; Kosykh, A.V.; Vorotelyak, E.A.; Gurskaya, N.G. Keratins as an Inflammation Trigger Point in Epidermolysis Bullosa Simplex. Int. J. Mol. Sci. 2021, 22, 12446. [Google Scholar] [CrossRef] [PubMed]
  5. Beriault, D.R.; Haddad, O.; McCuaig, J.V.; Robinson, Z.J.; Russell, D.; Lane, E.B.; Fudge, D.S. The mechanical behavior of mutant K14-R125P keratin bundles and networks in NEB-1 keratinocytes. PLoS ONE 2012, 7, e31320. [Google Scholar] [CrossRef] [PubMed]
  6. Fuchs, E.; Coulombe, P.; Cheng, J.; Chan, Y.M.; Hutton, E.; Syder, A.; Degenstein, L.; Yu, Q.C.; Letai, A.; Vassar, R. Genetic bases of epidermolysis bullosa simplex and epidermolytic hyperkeratosis. J. Investig. Dermatol. 1994, 103, 25S–30S. [Google Scholar] [CrossRef] [PubMed]
  7. Coulombe, P.A.; Lee, C.H. Defining keratin protein function in skin epithelia: Epidermolysis bullosa simplex and its aftermath. J. Investig. Dermatol. 2012, 132, 763–775. [Google Scholar] [CrossRef] [PubMed]
  8. Bar, J.; Kumar, V.; Roth, W.; Schwarz, N.; Richter, M.; Leube, R.E.; Magin, T.M. Skin fragility and impaired desmosomal adhesion in mice lacking all keratins. J. Investig. Dermatol. 2014, 134, 1012–1022. [Google Scholar] [CrossRef]
  9. Li, P.; Rietscher, K.; Jopp, H.; Magin, T.M.; Omary, M.B. Posttranslational modifications of keratins and their associated proteins as therapeutic targets in keratin diseases. Curr. Opin. Cell Biol. 2023, 85, 102264. [Google Scholar] [CrossRef]
  10. Snauwaert, J.J.; Yuen, W.Y.; Jonkman, M.F.; Moons, P.; Naulaers, G.; Morren, M.A. Burden of itch in epidermolysis bullosa. Br. J. Dermatol. 2014, 171, 73–78. [Google Scholar] [CrossRef] [PubMed]
  11. Scheffschick, A.; Kiritsi, D.; Magin, T.M. Keratin defects trigger the itch-inducing cytokine thymic stromal lymphopoietin through amphiregulin-epidermal growth factor receptor signaling. J. Allergy Clin. Immunol. 2019, 144, 1719–1722. [Google Scholar] [CrossRef]
  12. Fine, J.D.; Bruckner-Tuderman, L.; Eady, R.A.; Bauer, E.A.; Bauer, J.W.; Has, C.; Heagerty, A.; Hintner, H.; Hovnanian, A.; Jonkman, M.F.; et al. Inherited epidermolysis bullosa: Updated recommendations on diagnosis and classification. J. Am. Acad. Dermatol. 2014, 70, 1103–1126. [Google Scholar] [CrossRef] [PubMed]
  13. Has, C.; Bauer, J.W.; Bodemer, C.; Bolling, M.C.; Bruckner-Tuderman, L.; Diem, A.; Fine, J.D.; Heagerty, A.; Hovnanian, A.; Marinkovich, M.P.; et al. Consensus reclassification of inherited epidermolysis bullosa and other disorders with skin fragility. Br. J. Dermatol. 2020, 183, 614–627. [Google Scholar] [CrossRef]
  14. Lu, H.; Chen, J.; Planko, L.; Zigrino, P.; Klein-Hitpass, L.; Magin, T.M. Induction of inflammatory cytokines by a keratin mutation and their repression by a small molecule in a mouse model for EBS. J. Investig. Dermatol. 2007, 127, 2781–2789. [Google Scholar] [CrossRef] [PubMed]
  15. Liovic, M.; Lee, B.; Tomic-Canic, M.; D’Alessandro, M.; Bolshakov, V.N.; Lane, E.B. Dual-specificity phosphatases in the hypo-osmotic stress response of keratin-defective epithelial cell lines. Exp. Cell Res. 2008, 314, 2066–2075. [Google Scholar] [CrossRef] [PubMed]
  16. Liovic, M.; D’Alessandro, M.; Tomic-Canic, M.; Bolshakov, V.N.; Coats, S.E.; Lane, E.B. Severe keratin 5 and 14 mutations induce down-regulation of junction proteins in keratinocytes. Exp. Cell Res. 2009, 315, 2995–3003. [Google Scholar] [CrossRef] [PubMed]
  17. Roth, W.; Reuter, U.; Wohlenberg, C.; Bruckner-Tuderman, L.; Magin, T.M. Cytokines as genetic modifiers in K5-/- mice and in human epidermolysis bullosa simplex. Hum. Mutat. 2009, 30, 832–841. [Google Scholar] [CrossRef]
  18. Wagner, M.; Hintner, H.; Bauer, J.W.; Onder, K. Gene expression analysis of an epidermolysis bullosa simplex Dowling-Meara cell line by subtractive hybridization: Recapitulation of cellular differentiation, migration and wound healing. Exp. Dermatol. 2012, 21, 111–117. [Google Scholar] [CrossRef]
  19. Wagner, M.; Trost, A.; Hintner, H.; Bauer, J.W.; Onder, K. Imbalance of intermediate filament component keratin 14 contributes to increased stress signalling in epidermolysis bullosa simplex. Exp. Dermatol. 2013, 22, 292–294. [Google Scholar] [CrossRef]
  20. Bchetnia, M.; Tremblay, M.L.; Leclerc, G.; Duperee, A.; Powell, J.; McCuaig, C.; Morin, C.; Legendre-Guillemin, V.; Laprise, C. Expression signature of epidermolysis bullosa simplex. Hum. Genet. 2012, 131, 393–406. [Google Scholar] [CrossRef]
  21. Lettner, T.; Lang, R.; Klausegger, A.; Hainzl, S.; Bauer, J.W.; Wally, V. MMP-9 and CXCL8/IL-8 are potential therapeutic targets in epidermolysis bullosa simplex. PLoS ONE 2013, 8, e70123. [Google Scholar] [CrossRef] [PubMed]
  22. Herzog, J.; Rid, R.; Wagner, M.; Hundsberger, H.; Eger, A.; Bauer, J.; Onder, K. Whole-transcriptome gene expression profiling in an epidermolysis bullosa simplex Dowling-Meara model keratinocyte cell line uncovered novel, potential therapeutic targets and affected pathways. BMC Res. Notes 2015, 8, 785. [Google Scholar] [CrossRef] [PubMed]
  23. Coutier, J.; Bonnette, M.; Martineau, S.; Mercadier, A.; Domingues, S.; Saidani, M.; Jarrige, M.; Polveche, H.; Darle, A.; Holic, N.; et al. Human-Induced Pluripotent Stem Cell–Derived Keratinocytes, a Useful Model to Identify and Explore the Pathological Phenotype of Epidermolysis Bullosa Simplex. J. Investig. Dermatol. 2022, 142, 2695–2705 e2611. [Google Scholar] [CrossRef]
  24. Lee, G.H.; Lekwuttikarn, R.; Tafoya, E.; Martin, M.; Sarin, K.Y.; Teng, J.M. Transcriptomic Repositioning Analysis Identifies mTOR Inhibitor as Potential Therapy for Epidermolysis Bullosa Simplex. J. Investig. Dermatol. 2022, 142, 382–389. [Google Scholar] [CrossRef]
  25. Sonawane, A.R.; Platig, J.; Fagny, M.; Chen, C.Y.; Paulson, J.N.; Lopes-Ramos, C.M.; DeMeo, D.L.; Quackenbush, J.; Glass, K.; Kuijjer, M.L. Understanding Tissue-Specific Gene Regulation. Cell Rep. 2017, 21, 1077–1088. [Google Scholar] [CrossRef] [PubMed]
  26. Castela, E.; Tulic, M.K.; Rozieres, A.; Bourrat, E.; Nicolas, J.F.; Kanitakis, J.; Vabres, P.; Bessis, D.; Mazereeuw, J.; Morice-Picard, F.; et al. Epidermolysis bullosa simplex generalized severe induces a T helper 17 response and is improved by apremilast treatment. Br. J. Dermatol. 2019, 180, 357–364. [Google Scholar] [CrossRef]
  27. Mellerio, J.E. Potential therapeutic targeting of inflammation in epidermolysis bullosa simplex. Br. J. Dermatol. 2019, 180, 258–260. [Google Scholar] [CrossRef]
  28. Schroder, K.; Hertzog, P.J.; Ravasi, T.; Hume, D.A. Interferon-gamma: An overview of signals, mechanisms and functions. J. Leukoc. Biol. 2004, 75, 163–189. [Google Scholar] [CrossRef]
  29. Richmond, J.M.; Harris, J.E. Immunology and skin in health and disease. Cold Spring Harb. Perspect. Med. 2014, 4, a015339. [Google Scholar] [CrossRef]
  30. Wally, V.; Lettner, T.; Peking, P.; Peckl-Schmid, D.; Murauer, E.M.; Hainzl, S.; Hintner, H.; Bauer, J.W. The pathogenetic role of IL-1beta in severe epidermolysis bullosa simplex. J. Investig. Dermatol. 2013, 133, 1901–1903. [Google Scholar] [CrossRef] [PubMed]
  31. Gallucci, R.M.; Sloan, D.K.; Heck, J.M.; Murray, A.R.; O’Dell, S.J. Interleukin 6 indirectly induces keratinocyte migration. J. Investigig Dermatol. 2004, 122, 764–772. [Google Scholar] [CrossRef] [PubMed]
  32. Castro, F.; Cardoso, A.P.; Goncalves, R.M.; Serre, K.; Oliveira, M.J. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front. Immunol. 2018, 9, 847. [Google Scholar] [CrossRef]
  33. Gresser, I. Biologic effects of interferons. J. Investig. Dermatol. 1990, 95, 66S–71S. [Google Scholar] [CrossRef]
  34. Badowski, C.; Tan, T.S.; Aliev, T.; Trudil, D.; Larina, M.; Argentova, V.; Firdaus, M.J.; Benny, P.; Woo, V.S.T.; Lane, E.B. Detrimental Effects of IFN-gamma on an Epidermolysis Bullosa Simplex Cell Model and Protection by a Humanized Anti-IFN-gamma Monoclonal Antibody. JID Innov. 2022, 2, 100096. [Google Scholar] [CrossRef] [PubMed]
  35. Platanias, L.C. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 2005, 5, 375–386. [Google Scholar] [CrossRef] [PubMed]
  36. Kerns, M.L.; DePianto, D.; Dinkova-Kostova, A.T.; Talalay, P.; Coulombe, P.A. Reprogramming of keratin biosynthesis by sulforaphane restores skin integrity in epidermolysis bullosa simplex. Proc. Natl. Acad. Sci. USA 2007, 104, 14460–14465. [Google Scholar] [CrossRef]
  37. Ma, X.M.; Blenis, J. Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell Biol. 2009, 10, 307–318. [Google Scholar] [CrossRef] [PubMed]
  38. Galluzzi, L.; Spranger, S.; Fuchs, E.; López-Soto, A. WNT Signaling in Cancer Immunosurveillance. Trends Cell Biol. 2019, 29, 44–65. [Google Scholar] [CrossRef]
  39. Sneddon, J.B.; Zhen, H.H.; Montgomery, K.; van de Rijn, M.; Tward, A.D.; West, R.; Gladstone, H.; Chang, H.Y.; Morganroth, G.S.; Oro, A.E.; et al. Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer-associated stromal cells and can promote tumor cell proliferation. Proc. Natl. Acad. Sci. USA 2006, 103, 14842–14847. [Google Scholar] [CrossRef]
  40. Sharov, A.A.; Mardaryev, A.N.; Sharova, T.Y.; Grachtchouk, M.; Atoyan, R.; Byers, H.R.; Seykora, J.T.; Overbeek, P.; Dlugosz, A.; Botchkarev, V.A. Bone morphogenetic protein antagonist noggin promotes skin tumorigenesis via stimulation of the Wnt and Shh signaling pathways. Am. J. Pathol. 2009, 175, 1303–1314. [Google Scholar] [CrossRef]
  41. Kim, C.H. Migration and function of Th17 cells. Inflamm. Allergy Drug Targets 2009, 8, 221–228. [Google Scholar] [CrossRef] [PubMed]
  42. Harper, E.G.; Guo, C.; Rizzo, H.; Lillis, J.V.; Kurtz, S.E.; Skorcheva, I.; Purdy, D.; Fitch, E.; Iordanov, M.; Blauvelt, A. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: Implications for psoriasis pathogenesis. J. Investig. Dermatol. 2009, 129, 2175–2183. [Google Scholar] [CrossRef] [PubMed]
  43. Szeverenyi, I.; Cassidy, A.J.; Chung, C.W.; Lee, B.T.; Common, J.E.; Ogg, S.C.; Chen, H.; Sim, S.Y.; Goh, W.L.; Ng, K.W.; et al. The Human Intermediate Filament Database: Comprehensive information on a gene family involved in many human diseases. Hum. Mutat. 2008, 29, 351–360. [Google Scholar] [CrossRef]
  44. Autio, P.; Keski-Oja, J. Tetracyclines as anti-inflammatory treatment in skin diseases. Nord. Med. 1996, 111, 348–351. [Google Scholar]
  45. Khanna, N. Follow up of patients of acne vulgaris treated with tetracyclines. Indian. J. Dermatol. Venereol. Leprol. 1996, 62, 301–303. [Google Scholar]
  46. Yang, Y.L.; Wang, L.X.; Fei, X.M.; Lei, F.; Lu, W.P.; Yu, X.Q.; Zhang, S. The Effect of Doxycycline on the Expression of MMP-2 and MMP-9 in Multiple Myeloma. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2022, 30, 487–492. [Google Scholar] [CrossRef]
  47. Forloni, G.; Iussich, S.; Awan, T.; Colombo, L.; Angeretti, N.; Girola, L.; Bertani, I.; Poli, G.; Caramelli, M.; Grazia Bruzzone, M.; et al. Tetracyclines affect prion infectivity. Proc. Natl. Acad. Sci. USA 2002, 99, 10849–10854. [Google Scholar] [CrossRef] [PubMed]
  48. Bendeck, M.P.; Conte, M.; Zhang, M.; Nili, N.; Strauss, B.H.; Farwell, S.M. Doxycycline modulates smooth muscle cell growth, migration, and matrix remodeling after arterial injury. Am. J. Pathol. 2002, 160, 1089–1095. [Google Scholar] [CrossRef]
  49. Retief, C.R.; Malkinson, F.D.; Pearson, R.W. Two familial cases of epidermolysis bullosa simplex successfully treated with tetracycline. Arch. Dermatol. 1999, 135, 997–998. [Google Scholar] [CrossRef]
  50. Sapadin, A.N.; Fleischmajer, R. Tetracyclines: Nonantibiotic properties and their clinical implications. J. Am. Acad. Dermatol. 2006, 54, 258–265. [Google Scholar] [CrossRef]
  51. Moldovan, F.; Pelletier, J.P.; Jolicoeur, F.C.; Cloutier, J.M.; Martel-Pelletier, J. Diacerhein and rhein reduce the ICE-induced IL-1beta and IL-18 activation in human osteoarthritic cartilage. Osteoarthr. Cartil. 2000, 8, 186–196. [Google Scholar] [CrossRef] [PubMed]
  52. Wally, V.; Kitzmueller, S.; Lagler, F.; Moder, A.; Hitzl, W.; Wolkersdorfer, M.; Hofbauer, P.; Felder, T.K.; Dornauer, M.; Diem, A.; et al. Topical diacerein for epidermolysis bullosa: A randomized controlled pilot study. Orphanet J. Rare Dis. 2013, 8, 69. [Google Scholar] [CrossRef] [PubMed]
  53. Wally, V.; Hovnanian, A.; Ly, J.; Buckova, H.; Brunner, V.; Lettner, T.; Ablinger, M.; Felder, T.K.; Hofbauer, P.; Wolkersdorfer, M.; et al. Diacerein orphan drug development for epidermolysis bullosa simplex: A phase 2/3 randomized, placebo-controlled, double-blind clinical trial. J. Am. Acad. Dermatol. 2018, 78, 892–901 e897. [Google Scholar] [CrossRef]
  54. Teng, J.; Paller, A.S.; Bruckner, A.L. Diacerein 1% Ointment for the Treatment of Epidermolysis Bullosa Simplex: A Randomized, Controlled Trial. J. Drugs Dermatol. 2023, 22, 599–604. [Google Scholar] [CrossRef]
  55. Skurkovich, B.; Skurkovich, S. Autoimmune diseases are connected with disturbances in cytokine synthesis, and therapy with IFN-gamma blockers is their main pathogenetic treatment. Ann. N. Y. Acad. Sci. 2007, 1109, 167–177. [Google Scholar] [CrossRef]
  56. Larina, M.V.; Aliev, T.K.; Solopova, O.N.; Pozdnyakova, L.P.; Korobova, S.V.; Yakimov, S.A.; Sveshnikov, P.G.; Dolgikh, D.A.; Kirpichnikov, M.P. Neutralizing Monoclonal and Chimeric Antibodies to Human IFN-gamma. Bioorg. Khim. 2015, 41, 316–326. [Google Scholar] [CrossRef]
  57. Kuo, K.Y.; Cho, H.G.; Sarin, K.Y. Identification of Atorvastatin for Moderate to Severe Hidradenitis through Drug Repositioning Using Public Gene Expression Datasets. J. Investig. Dermatol. 2018, 138, 1209–1212. [Google Scholar] [CrossRef] [PubMed]
  58. Mirza, A.N.; Fry, M.A.; Urman, N.M.; Atwood, S.X.; Roffey, J.; Ott, G.R.; Chen, B.; Lee, A.; Brown, A.S.; Aasi, S.Z.; et al. Combined inhibition of atypical PKC and histone deacetylase 1 is cooperative in basal cell carcinoma treatment. JCI Insight 2017, 2, e97071. [Google Scholar] [CrossRef]
  59. Cho, H.G.; Fiorentino, D.; Lewis, M.; Sirota, M.; Sarin, K.Y. Identification of Alpha-Adrenergic Agonists as Potential Therapeutic Agents for Dermatomyositis through Drug-Repurposing Using Public Expression Datasets. J. Investig. Dermatol. 2016, 136, 1517–1520. [Google Scholar] [CrossRef]
  60. Brown, E.J.; Albers, M.W.; Shin, T.B.; Ichikawa, K.; Keith, C.T.; Lane, W.S.; Schreiber, S.L. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 1994, 369, 756–758. [Google Scholar] [CrossRef]
  61. Bchetnia, M.; Lacroix, J.; Farez, T.; Larouche, M.; Powell, J.; McCuaig, C.; Dupere, A.; Morin, C.; Legendre-Guillemin, V.; Laprise, C. Reduction in keratin aggregates in epidermolysis bullosa simplex keratinocytes after pretreatment with trimethylamine N-oxide. Exp. Dermatol. 2016, 25, 229–230. [Google Scholar] [CrossRef]
  62. Chamcheu, J.C.; Navsaria, H.; Pihl-Lundin, I.; Liovic, M.; Vahlquist, A.; Torma, H. Chemical chaperones protect epidermolysis bullosa simplex keratinocytes from heat stress-induced keratin aggregation: Involvement of heat shock proteins and MAP kinases. J. Investig. Dermatol. 2011, 131, 1684–1691. [Google Scholar] [CrossRef] [PubMed]
  63. Sporrer, M.; Prochnicki, A.; Tolle, R.C.; Nystrom, A.; Esser, P.R.; Homberg, M.; Athanasiou, I.; Zingkou, E.; Schilling, A.; Gerum, R.; et al. Treatment of keratinocytes with 4-phenylbutyrate in epidermolysis bullosa: Lessons for therapies in keratin disorders. EBioMedicine 2019, 44, 502–515. [Google Scholar] [CrossRef]
  64. Papp, K.; Reich, K.; Leonardi, C.L.; Kircik, L.; Chimenti, S.; Langley, R.G.; Hu, C.; Stevens, R.M.; Day, R.M.; Gordon, K.B.; et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: Results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J. Am. Acad. Dermatol. 2015, 73, 37–49. [Google Scholar] [CrossRef] [PubMed]
  65. Sakkas, L.I.; Bogdanos, D.P. Are psoriasis and psoriatic arthritis the same disease? The IL-23/IL-17 axis data. Autoimmun. Rev. 2017, 16, 10–15. [Google Scholar] [CrossRef]
  66. Aushev, M.; Koller, U.; Mussolino, C.; Cathomen, T.; Reichelt, J. Traceless Targeting and Isolation of Gene-Edited Immortalized Keratinocytes from Epidermolysis Bullosa Simplex Patients. Mol. Ther. Methods Clin. Dev. 2017, 6, 112–123. [Google Scholar] [CrossRef]
  67. Kirfel, G.; Herzog, V. Migration of epidermal keratinocytes: Mechanisms, regulation, and biological significance. Protoplasma 2004, 223, 67–78. [Google Scholar] [CrossRef] [PubMed]
  68. Hudson, L.G.; McCawley, L.J. Contributions of the epidermal growth factor receptor to keratinocyte motility. Microsc. Res. Tech. 1998, 43, 444–455. [Google Scholar] [CrossRef]
  69. Tan, T.S.; Common, J.E.A.; Lim, J.S.Y.; Badowski, C.; Firdaus, M.J.; Leonardi, S.S.; Lane, E.B. A cell-based drug discovery assay identifies inhibition of cell stress responses as a new approach to treatment of epidermolysis bullosa simplex. J. Cell Sci. 2021, 134, jcs258409. [Google Scholar] [CrossRef]
  70. Tkach, J.R. Treatment of recurrent bullous eruption of the hand and feet (Weber-Cockayne disease) with topical aluminum chloride. J. Am. Acad. Dermatol. 1982, 6, 1095–1096. [Google Scholar] [CrossRef]
  71. Jennings, J.L. Aluminum chloride hexahydrate treatment of localized epidermolysis bullosa. Arch. Dermatol. 1984, 120, 1382. [Google Scholar] [CrossRef] [PubMed]
  72. Younger, I.R.; Priestley, G.C.; Tidman, M.J. Aluminum chloride hexahydrate and blistering in epidermolysis bullosa simplex. J. Am. Acad. Dermatol. 1990, 23, 930–931. [Google Scholar] [CrossRef]
  73. Choudhury, S.; Baker, M.R.; Chatterjee, S.; Kumar, H. Botulinum Toxin: An Update on Pharmacology and Newer Products in Development. Toxins 2021, 13, 58. [Google Scholar] [CrossRef] [PubMed]
  74. Abitbol, R.J.; Zhou, L.H. Treatment of Epidermolysis Bullosa Simplex, Weber-Cockayne Type, With Botulinum Toxin Type A. Arch. Dermatol. 2009, 145, 13–15. [Google Scholar] [CrossRef] [PubMed]
  75. Swartling, C.; Karlqvist, M.; Hymnelius, K.; Weis, J.; Vahlquist, A. Botulinum toxin in the treatment of sweat-worsened foot problems in patients with epidermolysis bullosa simplex and pachyonychia congenita. Br. J. Dermatol. 2010, 163, 1072–1076. [Google Scholar] [CrossRef] [PubMed]
  76. Holahan, H.M.; Farah, R.S.; Ferguson, N.N.; Paller, A.S.; Legler, A.A. Treatment of symptomatic epidermolysis bullosa simplex with botulinum toxin in a pediatric patient. JAAD Case Rep. 2016, 2, 259–260. [Google Scholar] [CrossRef]
  77. Freund, R.R.A.; Gobrecht, P.; Fischer, D.; Arndt, H.D. Advances in chemistry and bioactivity of parthenolide. Nat. Prod. Rep. 2020, 37, 541–565. [Google Scholar] [CrossRef]
  78. Sun, J.; Li, P.; Gui, H.; Rittié, L.; Lombard, D.B.; Rietscher, K.; Magin, T.M.; Xie, Q.; Liu, L.; Omary, M.B. Deacetylation via SIRT2 prevents keratin-mutation-associated injury and keratin aggregation. JCI Insight 2023, 8, e166314. [Google Scholar] [CrossRef]
  79. Luskin, M.R.; DeAngelo, D.J. Midostaurin/PKC412 for the treatment of newly diagnosed FLT3 mutation-positive acute myeloid leukemia. Expert. Rev. Hematol. 2017, 10, 1033–1045. [Google Scholar] [CrossRef]
  80. Kwan, R.; Chen, L.; Looi, K.; Tao, G.Z.; Weerasinghe, S.V.; Snider, N.T.; Conti, M.A.; Adelstein, R.S.; Xie, Q.; Omary, M.B. PKC412 normalizes mutation-related keratin filament disruption and hepatic injury in mice by promoting keratin-myosin binding. Hepatology 2015, 62, 1858–1869. [Google Scholar] [CrossRef]
  81. Rietscher, K.; Jahnke, H.G.; Rübsam, M.; Lin, E.W.; Has, C.; Omary, M.B.; Niessen, C.M.; Magin, T.M. Kinase Inhibition by PKC412 Prevents Epithelial Sheet Damage in Autosomal Dominant Epidermolysis Bullosa Simplex through Keratin and Cell Contact Stabilization. J. Investig. Dermatol. 2022, 142, 3282–3293. [Google Scholar] [CrossRef]
  82. Omary, M.B.; Ku, N.O.; Tao, G.Z.; Toivola, D.M.; Liao, J. “Heads and tails” of intermediate filament phosphorylation: Multiple sites and functional insights. Trends Biochem. Sci. 2006, 31, 383–394. [Google Scholar] [CrossRef] [PubMed]
  83. Tockner, B.; Kocher, T.; Hainzl, S.; Reichelt, J.; Bauer, J.W.; Koller, U.; Murauer, E.M. Construction and validation of an RNA trans-splicing molecule suitable to repair a large number of COL7A1 mutations. Gene Ther. 2016, 23, 775–784. [Google Scholar] [CrossRef]
  84. Wally, V.; Brunner, M.; Lettner, T.; Wagner, M.; Koller, U.; Trost, A.; Murauer, E.M.; Hainzl, S.; Hintner, H.; Bauer, J.W. K14 mRNA reprogramming for dominant epidermolysis bullosa simplex. Hum. Mol. Genet. 2010, 19, 4715–4725. [Google Scholar] [CrossRef] [PubMed]
  85. Peking, P.; Breitenbach, J.S.; Ablinger, M.; Muss, W.H.; Poetschke, F.J.; Kocher, T.; Koller, U.; Hainzl, S.; Kitzmueller, S.; Bauer, J.W.; et al. An ex vivo RNA trans-splicing strategy to correct human generalized severe epidermolysis bullosa simplex. Br. J. Dermatol. 2019, 180, 141–148. [Google Scholar] [CrossRef]
  86. Hainzl, S.; Peking, P.; Kocher, T.; Murauer, E.M.; Larcher, F.; Del Rio, M.; Duarte, B.; Steiner, M.; Klausegger, A.; Bauer, J.W.; et al. COL7A1 Editing via CRISPR/Cas9 in Recessive Dystrophic Epidermolysis Bullosa. Mol. Ther. 2017, 25, 2573–2584. [Google Scholar] [CrossRef]
  87. Li, P.; Kleinstiver, B.P.; Leon, M.Y.; Prew, M.S.; Navarro-Gomez, D.; Greenwald, S.H.; Pierce, E.A.; Joung, J.K.; Liu, Q. Allele-Specific CRISPR-Cas9 Genome Editing of the Single-Base P23H Mutation for Rhodopsin-Associated Dominant Retinitis Pigmentosa. CRISPR J. 2018, 1, 55–64. [Google Scholar] [CrossRef] [PubMed]
  88. Giannelli, S.G.; Luoni, M.; Castoldi, V.; Massimino, L.; Cabassi, T.; Angeloni, D.; Demontis, G.C.; Leocani, L.; Andreazzoli, M.; Broccoli, V. Cas9/sgRNA selective targeting of the P23H Rhodopsin mutant allele for treating retinitis pigmentosa by intravitreal AAV9.PHP.B-based delivery. Hum. Mol. Genet. 2018, 27, 761–779. [Google Scholar] [CrossRef]
  89. Kocher, T.; Peking, P.; Klausegger, A.; Murauer, E.M.; Hofbauer, J.P.; Wally, V.; Lettner, T.; Hainzl, S.; Ablinger, M.; Bauer, J.W.; et al. Cut and Paste: Efficient Homology-Directed Repair of a Dominant Negative KRT14 Mutation via CRISPR/Cas9 Nickases. Mol. Ther. 2017, 25, 2585–2598. [Google Scholar] [CrossRef]
  90. Bchetnia, M.; Dionne Gagné, R.; Powell, J.; Morin, C.; McCuaig, C.; Dupérée, A.; Germain, L.; Tremblay, J.P.; Laprise, C. Allele-Specific Inactivation of an Autosomal Dominant Epidermolysis Bullosa Simplex Mutation Using CRISPR-Cas9. CRISPR J. 2022, 5, 586–597. [Google Scholar] [CrossRef]
  91. Pfendner, E.; Uitto, J.; Fine, J.D. Epidermolysis bullosa carrier frequencies in the US population. J. Investig. Dermatol. 2001, 116, 483–484. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Epidermolysis bullosa simplex (EBS) main subtypes.
Figure 1. Epidermolysis bullosa simplex (EBS) main subtypes.
Ijms 25 09495 g001
Figure 2. Inflammatory key pathways involved in blister formation in epidermolysis bullosa simplex (EBS) and their regulation by various therapeutic approaches.
Figure 2. Inflammatory key pathways involved in blister formation in epidermolysis bullosa simplex (EBS) and their regulation by various therapeutic approaches.
Ijms 25 09495 g002
Table 1. Epidermolysis bullosa simplex (EBS) gene expression profiling studies under normal growth conditions.
Table 1. Epidermolysis bullosa simplex (EBS) gene expression profiling studies under normal growth conditions.
ReferenceModel/Cell LinesUpregulated GenesDownregulated Genes
[14]WT mouse skin vs.
K5-−/− mouse skin
Gene symbolMethodGene symbolMethod
CXCL1, CXCL2, HPRT, KRT6aMA
IL6, IL1-βMA,
RTq-PCR
[16]WT vs.
EBS-sev KEB-7
ACTN1, AJAP1, CDH2, CDKN2A,
CST6, DBN1, FLNB, FN1, GNA11, IGSF4, KIAA0992, KRT18, KRT8, KRTHB1, LMNB2, MYL9, PODXL, PRSS2, RHOA, TGM2, TNC, TPM2, TUBB2, VIM
MAABLIM1, AMOTL2, ANXA1, ARHGDIB, CLDN4, COL17A1, CSTA, CTGF, CXADR, DDR1, DST, EGFR, ENC1, FRMD4B, GABARAPL1, GPNMB, HSPB1, IL18, IL8, KRT13, KRT15, KRT16, KRT19, KRT5, LPXN, LY6D, MTSS1, OLR1, PCDH7, PDLIM1, PDZK3, PFN2, RND3, SAA1, SCEL, SLC9A3R1, SNCG, PRR1B, STEAP1, TSPAN1MA
CDH1, GJB5, ITGA6, LAMA5, LAMB3, PPLMA,
RTq-PCR
CLDN3, CLDN7, DSC2, PKP1RTq-PCR
DSG3, GJA1, JUPMA,
RTq-PCR, WB, IF
DSPMA, WB, IF
WT vs. EBS-sev KEB-1 GJA1, DSG3, DSP, JUPWB
WT vs. EBS-loc KEB-4 ACTA2, AJAP1, ANK3, CLDN3, CLDN7, GSN, KRT14, KRT15, KRT16, KRT17, KRT4, LGALS7, HLA-B, HLA-C, NEFHMAPCDH7, BASP1, CTGF, CYR61, DST, ENC1, FRMD4B, IL18, KRT19, LPXN, OLR1, RDX, RND3, STK6MA
[17]WT mouse skin vs. K5−/− mouse skinMCP-1, CCL2, MIP-3a/CCL20, MIP-3b/CCL19, CXCL16
[18]WT vs. EBS-sev KEB-7ACSL3, ANXA2, ARPC3, CAV1, CCT5, EEF1G, EIF1, ENO1, GLUD1, H2AFZ, HNRPDL, SP90AB1, MAN2A1, PDCD4, RAB27B, RPL13, RPL19, RPL23A, RPL28, RPL31, RPL4, RPL5, RPL7A, RPLP0, RPS18, RPS2, PS25, RPS3, RPS4X, RPS6, RPS8, SFN, TAX1BP1, TMBIM6, TPI1, TPT1, EEF1A1, GSN, HNRNPA1, KRT5, KRT6ASSH
ANXA8, DDIT4, DSG3, DSP, F3, KRT14, KRT16, KRT17, KRT6B, MALAT1, PERP, TXNIP, UBE2K, YWHAZSSH,
RTq-PCR
[20]WT human epidermis tissue vs.
EBS-sev and EBS-loc human epidermis tissue
SPRR2B, AREG, BDP1, NR4A2, GAL, LYVE1, PSG4, H19, PDE6A, C5orf27, THRSP, FAR2, AADACL3, CRAT, AGR2, IGFL4, KLK6, PM20D1, ATP12A, FSIP2MA
AWAT2, DGAT2L6, FADS2, ACSBG1, SPRR4, KRT79MA,
RTq-PCR
ELOVL3MA, RTq-PCR, WB
WT human epidermis vs. EBS-sev human epidermisFADS1, CYP4F8, AWAT1, ALOX15B, ACSM3, SOAT1, SLC27A2, HAO2, INSIG1, KRTAP5-8, KRT25, KRT71, KRT74, KRT27, TCHH, CHI3L1, MUC1, SLCO4C1, TGIF2LX, IGLJ3, IGJ, IGHA1, TMEM56, LRCC37A2, THRSP, FAR2, AADACL3, CRAT, AGR2, IGFL4, KLK6, PM20D1, ATP12A, FSIP2
AWAT2, DGAT2L6, FADS2, ACSBG1, SPRR4, KRT79MA,
RTq-PCR
ELOVL3MA, RTq-PCR, WB
[30]WT immortalized keratinocytes vs. immortalized EBS-sev cell lines (KEB-7, EBDM-1) KRT14,IL-1b, KRT6ASqRT-PCR
[21]WT immortalized keratinocytes vs. immortalized EBS-sev cell lines (KEB-7, EBDM-1) KLK6, KLK8, KLK10, KLK11, KLK13, KLK14, MMP1, MMP13, WIPF1, ARHGEF4, ARHGEF37, CDC42BPG, KRT6BMA
KLK5, KLK7, KRT14, KRT15, KRT16 KRT17, KRT5MA, sqRT-PCR, WB
MMP7, MMP9, MMP19, ARHGEF9, DSC1, DSC2, DSC3, DSG1, DSG3, DSG4, GJA1, GJB2, GJB6, CXCL1, CXCL8/IL-8, CXCL14MA,
sqRT-PCR
[22]WT vs. EBS-sev KEB-7 KGFLP1, ANKRD2, PIK3R3, PTPN20A, FAM21A, APBB2, DUOX1, ZNF627, MCOLN2, APOB, AJAP1, BNC2, LOC100652860, FKTN, TMEM204, BMS1P1, MAN1A1, STOX1, RPL10, CDC144C, NOX5EVC2, PTPN20C, PTPN20B, NID1, ASAH2, FLJ20444, TP53INP1, FRG1B, MSLN, DENND1B, IFFO2, STRBP, DSEL, AOX1, LRP12, ADHFE1, FAM21D, SNORD64, FAM21A, EFEMP1, TSPYL5, RNF212, DDX43, ZNF136, CCDC144A, CCDC144A, ZNF700, BGN, H2AFY2, SNORD116-21, HOXA9, RPS23MAMIR492, H19, EYA4, TMPRSS15, ITGBL1, EDIL3, CDR1, NEFL, SMOC2, GHR, TFPI2, ARHGAP28, NNMT, SOX2, HIST1H, PCCA, ZNF570, CDK14, MEST, CYP7B1, GALNTL4, CRIP2, IPO7, SAAL1, KRTCAP3, FAM159A, EYA1, CDC25B, NKX2-6, HTATIP2, ILK, ACSF2, PDZD2, CENPH, TOX, VSTM2L, SYT17, SLC7A2, IKZF3MA
TDRD12, NEFH, NLRP2, KLK5, ENPP1, ZFP42, DKK1, CYYR1, C10orf99, SYCP2, PRICKLE1,
SLC44A5, PLA2G7, MOXD1,
WNT5A, WISP3, ARHGEF9, HSD17B11, ADAMTSL3, FAM102B SGMS1, ARHGAP29, SLC15A2, ROBO1, ERCC6, NREP, KIAA1324L, ROR1, ZFAND4, SELENBP1, NF334
PTPN20C, IRX4, PTPN20A, PNMAL1, NID1, ZNF32, AHI1, ELAVL2, SLC16A9, FAM25A,
MAPK8, CDC14B, GABPB2 TCHH, LMF1, CSTF2T, SGK1, UAP1, POPDC2, ZNF502
MA,
RTq-PCR
KRT19, KYNU, PDZK1, OLFM4, SLC38A4, BST2, PPARGC1A, GALNT5, FKBP10, GIPC2, AMOT, ZNF114, CLEC2B, FAM198B, SLC2A3, CAPNS2, TBX18, LRCH2, NEFM, CPT1C, ZNF43, LY75, GLDC, TMTC1, SLCO1B3, SLC6A14, SLC24A3, EPSTI1, SATB2, HSD17B2, AKR1B10, GPC3, IFITM3, HOXD10, MSX2, IL17RB, BLMH, SLC9A2, CPNE1, WDR17, RB1, DPYD, PRTFDC1, GLRX, PPP1R16B, GTF2H2D, REPS2, GPR143, GTF2H2, CYP7B1, BCL11A, MERTK, PRDM5, ACOXL, AHCY, ARMCX2, PAX6, HOXD11, SMARCA1, IFI44L, PITRM1, NAP1L5, PIGUMA,
RTq-PCR
[23]WT human keratinocytes vs. EBS-loc and EBS-intermed keratinocytesACOT1, ALDH1A3, ALOX15B, ALPL 6, ANGPTL4, ASPN 34, ATP6V0A4, BEX1, C6orf223, CARD17, CCDC9B, CCL2, CCL20, CD99L2, CNIH3, CNTN3, COL1A1, COL6A6, CRYBB2P1, CXCL10, CXCL11, CXCL5, CXCL6, CYP1A1, DAAM2, DLK1, ECHDC1, ELN, FIBIN, FMOD, FNDC1, G0S2, GALNT14, GALNT16, HLA-DRB1, HMGB3, HS3ST2, HSD17B2, IER3, IFI27, IFI44, IL1A, IL1B, IL7R, INHBA, INSYN2B, ITGA10, ITGBL1, KRT6B, LAMB3, LIF, LINC00520, LINC-PINT, LOC100996732, LOX14, LSP1P4, LSP1P5, MANCR, MFAP4, MT1L, MX2, NFIX, NOV, NPAS2, NPIPB12, NPIPB13, NPIPB3, NPIPB5, NUPR1, LFML2B, PCDHA6, PDGFRB, PLD5, POSTN, PTGS1, RTEL1-TNFRSF6B, SAA1, SAA2, SAA2-SAA4, SERPINB2, SFRP2, SLC9A7, SMOC1, SMOC2, ST6GAL2, SYNDIG1, TAC1, TAGLN, TFCP2L1, TMEM255B, TNFRSF6B, TNMD, TPST1, TRNP1, VNN1, WIPI1, XDH RNA seq
Abbreviations—EBDM-1: EBS severe disease phenotype cell line with K14 R125H mutation; EBS-loc: localized EBS; EBS-sev: severe EBS; IF: immunofluorescence; K5: keratin 5; KEB-1: cell line with K5 E475G mutation; KEB-4: cell line with K14 V270M mutation; KEB-7: cell line with K14 R125P mutation; MA: microarray; RTq-PCR: real-time quantitative PCR; sqRT-PCR: semi-quantitative real-time PCR; SSH: suppression subtractive hybridization; WB: Western blot; WT: wild-type. In bold—genes found to be regulated in multiple studies.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bchetnia, M.; Powell, J.; McCuaig, C.; Boucher-Lafleur, A.-M.; Morin, C.; Dupéré, A.; Laprise, C. Pathological Mechanisms Involved in Epidermolysis Bullosa Simplex: Current Knowledge and Therapeutic Perspectives. Int. J. Mol. Sci. 2024, 25, 9495. https://doi.org/10.3390/ijms25179495

AMA Style

Bchetnia M, Powell J, McCuaig C, Boucher-Lafleur A-M, Morin C, Dupéré A, Laprise C. Pathological Mechanisms Involved in Epidermolysis Bullosa Simplex: Current Knowledge and Therapeutic Perspectives. International Journal of Molecular Sciences. 2024; 25(17):9495. https://doi.org/10.3390/ijms25179495

Chicago/Turabian Style

Bchetnia, Mbarka, Julie Powell, Catherine McCuaig, Anne-Marie Boucher-Lafleur, Charles Morin, Audrey Dupéré, and Catherine Laprise. 2024. "Pathological Mechanisms Involved in Epidermolysis Bullosa Simplex: Current Knowledge and Therapeutic Perspectives" International Journal of Molecular Sciences 25, no. 17: 9495. https://doi.org/10.3390/ijms25179495

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