Next Article in Journal
Navigating the Alzheimer’s Biomarker Landscape: A Comprehensive Analysis of Fluid-Based Diagnostics
Previous Article in Journal
A Novel Method to Profile Transcripts Encoding SH2 Domains in the Patiria miniata Mature Egg Transcriptome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Resolution of Chronic Inflammation, Restoration of Epigenetic Disturbances and Correction of Dysbiosis as an Adjunctive Approach to the Treatment of Atopic Dermatitis

by
Gregory Livshits
1,2,* and
Alexander Kalinkovich
2
1
Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel
2
Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv 6927846, Israel
*
Author to whom correspondence should be addressed.
Cells 2024, 13(22), 1899; https://doi.org/10.3390/cells13221899
Submission received: 8 October 2024 / Revised: 7 November 2024 / Accepted: 14 November 2024 / Published: 18 November 2024
(This article belongs to the Section Cellular Pathology)

Abstract

:
Atopic dermatitis (AD) is a chronic inflammatory skin disease with multifactorial and unclear pathogenesis. Its development is characterized by two key elements: epigenetic dysregulation of molecular pathways involved in AD pathogenesis and disrupted skin and gut microbiota (dysbiosis) that jointly trigger and maintain chronic inflammation, a core AD characteristic. Current data suggest that failed inflammation resolution is the main pathogenic mechanism underlying AD development. Inflammation resolution is provided by specialized pro-resolving mediators (SPMs) derived from dietary polyunsaturated fatty acids acting through cognate receptors. SPM levels are reduced in AD patients. Administration of SPMs or their stable, small-molecule mimetics and receptor agonists, as well as supplementation with probiotics/prebiotics, demonstrate beneficial effects in AD animal models. Epidrugs, compounds capable of restoring disrupted epigenetic mechanisms associated with the disease, improve impaired skin barrier function in AD models. Based on these findings, we propose a novel, multilevel AD treatment strategy aimed at resolving chronic inflammation by application of SPM mimetics and receptor agonists, probiotics/prebiotics, and epi-drugs. This approach can be used in conjunction with current AD therapy, resulting in AD alleviation.

1. Overview

Atopic dermatitis (AD) is the most common chronic inflammatory skin disease, affecting up to 20% of children and 10% of adults, depending on ethnicity [1,2,3]. It is a complex and heterogeneous immune-mediated skin condition characterized by distinct phenotypes, each exhibiting various molecular variants [4,5,6,7]. Noxious environmental stimuli, immune dysregulation, genetic and epigenetic factors, impaired epidermal barrier integrity, and skin and gut microbiota abnormalities (dysbiosis) are all considered to play an important role in initiating and maintaining the state of chronic inflammation in AD and contribute to the control of the disease phenotype [8,9].
Being an immune-mediated disease, AD requires the use of immune-targeted approaches for its treatment. AD emerged as the T helper 2 (Th2) disease, and this is supported by multiple observations, including increased levels of cytokines and chemokines secreted by Th2 cells, such as interleukin 4 (IL-4), IL-5, IL-13, CCL17, CCL18, and CCL22 [10,11,12]. AD is also characterized by the Th22-skewed phenotype with the overproduction of IL-22, while the contributions of the Th1 and Th17 axes vary depending on the AD endophenotype [13,14].
These observations lead to the development of targeted therapies, in particular, biological drugs (mainly monoclonal antibodies to cytokines and cytokine receptors) and Janus kinase inhibitors (JAKi) [15,16,17]. Although these treatments greatly improve AD management, they are not without serious side effects, mainly due to non-specific immune suppression [18,19,20]. Moreover, unlike psoriasis, where cure rates reach 80%, targeted cytokine inhibition in patients with AD has not demonstrated comparable efficacy [17]. This discrepancy may be due to the heterogeneous nature of AD, which includes a wide range of phenotypes and endotypes [21,22]. However, persistent inflammation remains a constant characteristic of any type of chronic AD [23,24,25]. The use of monoclonal antibodies against key cytokines and their receptors involved in AD-associated inflammation, such as IL-4 and IL-13, has shown therapeutic effects in severe AD [17,24,26].
However, as in the case of JAKi, they are not able to resolve chronic inflammation and also have serious side effects due to their nonspecific immunosuppressive activity [27,28,29]. In this regard, an application of the agents capable of resolving chronic inflammation may be an attractive therapeutic approach for AD. Inflammation resolution is an active process involving the activation of signaling pathways during inflammation initiation with the subsequent generation of specialized pro-resolution mediators (SPMs) derived from polyunsaturated fatty acids (PUFAs) that activate resolution mechanisms [30]. These mediators include resolvins, lipoxins, protectins, and maresins acting through a range of G protein-coupled receptors (GPCRs) [31]. Notably, PUFA derivative levels are decreased in patients with AD, and PUFA supplementation ameliorates the disease manifestations [32,33,34,35]. Moreover, recent Mendelian randomization analysis suggests that elevated levels of n-3 PUFA and docosahexaenoic acids (DHA), which is a derivative of n-3 PUFA, are associated with a lower risk of AD [36]. Several preclinical studies in AD models revealed clear beneficial effects of SPMs [37,38], supporting the validity of the idea of using agents that resolve chronic inflammation to treat AD.
The prevalence of AD has tripled in the past 30 years in industrial countries [39], raising the possibility that environmental factors, acting through epigenetic mechanisms, may be important. Indeed, epigenetic alterations (DNA methylation, histone modifications, and non-coding RNA-mediated regulation) were shown to contribute to key pathogenic events, such as immune activation, T-cell polarization and keratinocyte dysfunction in AD [40,41]. This suggests an application of drugs selectively influencing the epigenetic landscape (epidrugs) in AD. In support, some studies report on the efficacy of micro-RNA (miRNA) targeting in AD murine models [42,43,44]. The epigenetic regulators have been shown to modulate chromatin structure in epidermal keratinocytes by influencing the function of genes, mainly epidermal differentiation complex (EDC) genes, which are associated with proliferation and differentiation processes in keratinocytes and their precursors [45]. Thus, we can propose the epidrugs that are most suitable for inhibiting inflammation (and/or promoting its resolution) as well as supporting keratinocyte function as potential AD therapeutic agents.
AD is associated with marked changes in the skin microbiota composition (dysbiosis), specifically loss of commensal diversity and dominant colonization by pathogenic Staphylococcus bacteria, primarily S. aureus [46]. This dysbiosis is associated with more severe disease, more pronounced type 2 immune abnormality, and barrier impairment [47]. Conversely, restoration of commensal diversity may precede and indicate disease resolution [48,49,50]. The development of AD is also characterized by gut dysbiosis, where, as in the skin, the abundance and diversity of commensals are reduced while the abundance of pathobionts is increased [51,52]. AD-associated gut dysbiosis results in impaired intestinal barrier integrity (leaky gut syndrome) and reduced production of short-chain fatty acids (SCFA), mainly the anti-inflammatory butyrate and propionate. Notably, the absence of SCFA-producing microbes preceded AD symptoms [46] and was found to be positively correlated with disease severity in infants [53]. Moreover, butyrate has been shown to attenuate skin inflammation exacerbated by S. aureus [54], thereby supporting the existence of a gut-skin axis playing a detrimental role in the pathogenesis of AD [55,56].
Based on these findings, we suggest that the combination of SPM mimetics and receptor agonists with dysbiosis and epigenetic disturbance correctors has translational potential to alleviate the pathological manifestations of AD. The ultimate goal of this strategy is to effectively resolve chronic inflammation. Evidence supporting this new AD treatment approach prompted the present review.

2. The Major Role of Chronic Inflammation in the Pathogenesis of AD

Despite careful, long-term study [4,8,57], the mechanisms underlying the pathogenesis of AD remain unclear. However, recent research has shown notable progress. Major advances are related to a better understanding of genetic and epigenetic regulation [39,40,58], the role of innate immunity [59,60], the involvement of T-cell subsets and cytokines [61,62,63], the role of skin and gut dysbiosis [49,50,64], and mechanisms of epithelial barrier disruption in the pathogenesis of AD [65,66,67,68]. It is assumed that all these mechanisms underlie the development and maintenance of chronic inflammation, which is considered a key pathogenic factor in AD [61,69,70].
Over the last 3 decades, the prevailing hypothesis has been that acute AD is predominantly driven by Th2 cytokines, with a shift to a Th1-dominant milieu in the chronic phase [71,72,73]. However, a detailed study of the transcriptomic changes that characterize the transition from acute to chronic lesional skin has led to a paradigm shift in the immunopathology of AD [11,74]. Of particular significance are the results of the study by Tsoi et al. [74], in which RNA sequencing was used to characterize non-lesional, acute and chronic lesional skin from patients with AD and healthy skin. Genetic transcriptome analysis revealed that 74% of genes whose regulation was disrupted in acute AD were also observed in chronic AD, including genes involved in epithelial differentiation (IL-20, KRT16, KRT6B, S100A8, and S100A9), antimicrobial and immunomodulatory chemokines (CXCL1, CXCL6), T cell regulation (TNIP3, CLEC3A) and Th2 cell differentiation (IL-19). In contrast, only 34% of the expressed genes found in chronic skin lesions were also altered in acute lesions [74]. Moreover, acute skin lesions and cytokine profiles often exhibit a biphasic profile, indicating an ongoing transition to a chronic phase [74]. These findings indicate that acute lesions result in widespread activation of the immune system, while chronic lesions leave these genes intact.
Specifically, genes related to most Th-cell subsets, including Th1-, Th2-, Th17-, and Th22-related genes, were found to be dysregulated already in acute lesions, and these genes remained or are further dysregulated in chronic lesions [74]. In contrast to the paradigm suggesting a Th2 to Th1 switch, this study reported no association of the immunological switch with the transition from acute to chronic stage, but rather, progressive immune activation of all Th subsets was observed.
Another important observation reported in this study is that a relatively small number of genes are differentially regulated in chronic versus acute AD: 29 genes showed increased and 13 decreased expression levels, suggesting relatively small differences between acute and chronic lesions. Altogether, these findings suggest that the transition of acute-to-chronic AD is associated with quantitative rather than qualitative changes in cytokine responses.
In addition to the above-mentioned T-cell subsets, other immune cells, as well as keratinocytes, are deeply involved in skin inflammation, in which the coexistence of all these cells creates a complex, self-sustaining vicious cycle that underlies AD pathogenesis. As demonstrated in Figure 1, activated keratinocytes produce numerous chemokines, including thymus- and activation-regulated chemokine (TARC)/CCL17, regulated on activation, normal T-cell-expressed and secreted (RANTES)/CCL5, monocyte chemotactic protein (MCP)-4/CCL13, eotaxin-3/CCL26, and some others, which attract and activate Langerhans cells (LCs), dendritic cells (DCs), eosinophils, basophils, mast cells, type-2 innate lymphoid cells (ILC2), and Th17 cells. Keratinocytes also produce thymic stromal lymphopoietin (TSLP), which, in concert with all mentioned mediators, stimulates type 2 innate lymphoid (ILC2) cells to produce IL-5 and IL-13 and also Th2 cells to produce IL-1β, IL-4, IL-6, IL-8, IL-13, IL-18, IL-25, IL-31, and IL-33. In particular, IL-4 and IL-13 recruit eosinophils and stimulate B cells for the production of IgE and several chemokines, such as CCL5, CCL11, CCL17, CCL18, and CCL-22.
IL-31 stimulates keratinocytes, whereas IL-33 decreases the expression of filaggrin (FLG) by keratinocytes, a major epidermal protein, thereby promoting itch (pruritus) and skin barrier disruption. In addition, IL-4 and IL-13 suppress the production of antimicrobial peptides (AMPs), weakening the barrier function against microbes. IL-4, IL-5, and IL-13 further amplify the Th2 immune responses and exacerbate AD. Th17 and Th22 cells also increase in number, leading to enhanced secretion of pro-inflammatory mediators. In addition to the Th2 immune response, IL-17 from Th17 cells and IL-22 from Th22 cells induce acanthosis in the epidermis, which contributes to the formation of intractable lichenified skin lesions. Produced by activated DCs, IL-12, together with IL-18, activate Th1 cells to produce IFNγ, which, in turn, stimulates keratinocytes. This pro-inflammatory vicious cycle drives and maintains AD-associated chronic skin inflammation, thereby exacerbating the disease manifestations [61,62,75,76,77,78,79].

3. Epigenetic Profile of AD

Epigenetic changes (i.e., modifications of genetic expression information not associated with changes in DNA sequence) play an important role in normal development, cell-specific gene expression, and the pathogenesis of many complex diseases [80,81], including inflammatory skin disorders [4,82,83,84,85]. The major molecular epigenetic mechanisms include DNA methylation, histone modification, and regulation by non-coding RNAs (ncRNAs), mainly by miRNAs [86]. The key players governing these epigenetic modifications include three families of epigenetic enzymes, namely writers, readers, and erasers.
Writers are a group of enzymes that act on histones and add small covalent modifications such as methyl and acetyl groups. DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B) add methyl groups to the fifth carbon of cytosine residues that are linked by a phosphate to a guanine nucleotide (i.e., a CpG dinucleotide). Most of the CpG dinucleotides, which are distributed irregularly across the genome, are methylated. The exception is CpG dinucleotides within CpG islands that are usually unmethylated [87,88]. Histone acetyltransferases (HATs) add acetyl groups, and histone methyltransferases (HMTs) add methyl groups to histones.
Readers are a diverse group of proteins, such as the bromodomain and extra-terminal domain (BET) family of proteins. They identify histone modifications made by writers and mediate downstream biological events. Erasers remove epigenetic modifications. Histone demethylases (HDMs) remove methyl groups, and histone deacetylases (HDACs) remove acetyl groups from histones. In humans, 18 different mammalian HDACs have been identified, of which class III HDACs (Sirtuins, SIRTs) are involved in the development of asthma [89] and AD [90]. All these epigenetic modifications play a critical role in altering chromatin conformation, leading to either transcriptional repression or gene activation [91,92,93].
Significant differences in DNA methylation patterns were observed between lesional and intact epidermis in AD patients, suggesting that aberrant DNA methylation may play a role in the pathogenesis of AD [94]. In this study, substantial hypermethylation changes in the S100A proteins OAS2 and KRT6A, involved in the regulation of innate immunity [95], have been found to be related to their increased expression in lesional skin. In another study [96], global hypomethylation was observed in DCs and monocytes from AD patients compared to healthy controls along with locus-specific hypomethylation at the high-affinity receptor for IgE (FCER1G) promoter in correlation with its overexpression. Increased DNA methylation of the FLG gene, a major structural protein in the stratum corneum, was observed in the lesional epidermis of patients with severe AD compared to the non-lesional epidermis [97] and was also associated with an increased AD risk [98]. Interestingly, although the CD4+ T cells from AD patients do not show significant changes in global methylation patterns compared to healthy controls [99], CD4+ T cells expressing the cutaneous lymphocyte antigen (CLA) (CD4+CLA+ T cells) showed significant differences in DNA methylation in 40 genes, including IL-13 gene, compared with healthy controls [100]. These results suggest the involvement of epigenetic regulation in the functional activity of T cells homing to the skin in AD pathogenesis.
DMT1 mRNA expression level is significantly lower in peripheral blood mononuclear cells (PBMCs) from AD patients compared with healthy controls [101]. mRNA and protein expression of TSLP is upregulated in the skin lesions from patients with AD in association with promoter hypomethylation (detected by using bisulfite sequencing), suggesting that DNA demethylation (detected by using 5-aza, a DNA methyltransferase inhibitor) of a regulatory region of TSLP may contribute to its overexpression in AD skin lesions [102]. Bisulfite pyrosequencing of the promoter region of human AMP beta-defensin-1 (HBD-1) showed significantly higher methylation frequencies at the CpG 3 and CPG 4 sites in AD lesional samples than in non-lesional AD skin and healthy skin samples [103], suggesting that promoter DNA methylation contributes to HBD-1 deficiency in AD. Additionally, highly methylated SNPs in the gene encoding kinesin family member 3A protein (KIF3A) were found in the skin samples from AD patients compared with healthy controls [104]. In this study, KIF3A knockout mice demonstrated impaired junctional proteins and increased susceptibility to developing AD, suggesting an important role of KIF3A in maintaining skin barrier integrity.
Currently, only a few studies have focused on AD-specific histone modifications. They have found, for example, that the expression of the SIRT1 gene, involved in several cellular pathways and associated with the beneficial effects on skin aging [105], is down-regulated in the lesions of AD patients compared to normal individuals [90]. In a study of human and mouse keratinocytes, it was shown that physical interaction between HDAC1 and the FLG promoter [detected by chromatin immunoprecipitation (ChIP) assay] stimulates FLG promoter activity, which was significantly suppressed by TNFα and IFNγ [106].
The vast majority of the results linking epigenetics and AD have been obtained from the study of ncRNAs, primarily miRNAs [107,108,109]. As illustrated in Figure 2, miRNAs are involved in the regulation of key mechanisms in AD pathogenesis, such as T-cell imbalance, chronic inflammation, and skin barrier dysfunction. miR-155 is one of the most up-regulated miRNAs in AD patients [110], and its plasma levels positively correlate with the disease severity, the percentage of Th17 cells and IL-17 expression [111]. In this study, miR-155 was found to be predominantly expressed in activated skin infiltrating immune cells, presumably due to reduced expression of CTLA-4, an immune checkpoint receptor that has an inhibitory effect on T-cell responses, thereby promoting chronic skin inflammation. In addition, miR-155 stimulates the differentiation of T regulatory cells (Tregs) and Th17 cells by targeting suppressors of cytokine signaling 1 (SOCS1), the important negative regulator of JAK/signal transducer and activator of transcription (JAK/STAT) signaling pathway [112]. In a murine model of AD, silencing of miR-155 attenuated epidermal thickening, reduced inflammatory cell infiltration and Th2 cytokine secretion as well as increased the expression of protein kinase inhibitor α (PKIα) and tight junction proteins; reduced expression of TSLP and IL-33 was also found in these mice [113].
An increased expression of miR-146a in keratinocytes of AD patients appears to play an anti-inflammatory role. Indeed, miR-146a inhibits the expression of NF-κB upstream elements, such as caspase recruitment domain-containing protein 10 (CARD10), tumor necrosis factor receptor-associated factor 6 (TRAF6), and interleukin-1 receptor-associated kinase 1 (IRAK1), thereby reducing IFNγ- and NF-κB-activated chronic skin inflammation [114]. Elevated miRNA-146a expression in human primary keratinocytes stimulated with IFNγ, TNFα, or IL-1β is associated with reduced expression of numerous pro-inflammatory factors, including CCL5 and CCL8 [115]. In a murine model of AD, miR-146a-deficient mice developed skin inflammation, characterized by increased accumulation of infiltrating immune cells and increased expression of CCL5 and CCL8 in the skin [115].
Another example is miR-124. It inhibits the expression of p65, a member of the NF-κB family involved in the inflammatory reactions. The decreased expression of miR-124 was observed in lesional skin samples from chronic AD [116]. In this study, keratinocyte stimulation with IFNγ or TNFα resulted in upregulation of IL-8, CCL5 and CCL8 partially reversed by miR-124. These data indicate that miR-124 suppresses NF-κB-dependent inflammatory responses in keratinocytes and chronic skin inflammation in AD, suggesting elevation of miR-146a as a potential therapeutic approach in AD.
Increased expression of several miRs has been suggested to be associated with the worsening of AD, suggesting that their inhibition may result in attenuation of AD manifestations. For example, the viability of human keratinocytes and release of IL-1β and IL-6 after stimulation with TNFα and IFNγ are negatively associated with the expression of miR-375-3p through the activation of Yes-associated protein 1 (YAP1) and lymphoepithelial Kazal type inhibitor (LEKTI) inflammatory pathways [117]. The negative role of miR-375-3p in AD pathogenesis is shown in a clinical study in which the severity of AD is positively associated with miR-375-3p levels in saliva samples [118]. Increased expression of miR-10a-5p in keratinocytes of AD patients compared with healthy controls is associated with reduced keratinocyte proliferation and migration through targeting hyaluronan synthase (HAS3), a damage-associated positive regulator of keratinocyte function, thereby potentially impairing skin barrier in AD [119]. Upregulation of miR-29b in lesional skin and serum samples from AD patients is associated with enhanced keratinocyte apoptosis through inhibiting Bcl-2-like protein 2 (Bcl2L2), contributing to epithelial barrier dysfunction impaired in AD [120].
In contrast, elevated plasma miR-151a levels in AD patients compared with healthy controls appear to be beneficial as they are associated with downregulation of IL-12 receptor β2 (IL12RB2) expression, a subunit of the IL-12 receptor [121]. Therefore, downregulation of IL12RB2 expression by miR-151a may reduce the responsiveness of immune cells to IL-12 signaling. The binding of this receptor activates the JAK/STAT pathway involved in the production of IFNγ and TNFα by T cells and natural killer (NK) cells, thereby enhancing inflammation [122].
Stimulation of human epidermal keratinocytes with IL-13 results in the reduced expression of miR-143, whereas overexpression of miR-143 mimics IL-13-induced downregulation of FLG, loricrin, and involucrin in epidermal keratinocytes via targeting IL-13Rα1 [123]. These data suggest that activation of miR-143 may attenuate AD and that this miRNA may serve as a potential preventive and therapeutic target in AD. In AD lesional skin, miR-335 expression is downregulated, whereas SOX6 is upregulated throughout the epidermis, resulting in reduced keratinocyte differentiation and impaired skin barrier [124].
Bioinformatics analysis of microarray data identified two downregulated differently expressed miRNAs (DEMs), namely, let-7a-5p and miR-26a-5p [125]. let-7a-5p potentially targets the chemokine receptor CCR7, overexpressed in T cells and DCs in AD lesions [126]. miR-26a-5p probably regulates the HAS3 gene, involved in the synthesis of hyaluronic acid, a major component of the extracellular matrix and overexpressed in AD lesions compared with healthy skin and non-lesional AD skin [127]. miR-939 is found to be highly upregulated in S. aureus-stimulated keratinocytes and AD lesions, accompanied by increased expression of several matrix metalloproteinases (MMPs) to promote the colonization of S. aureus and exacerbated S. aureus-induced AD-like skin inflammation [128].
In addition to miRNAs, long non-coding RNAs (lncRNAs) are also likely to be involved in the pathogenesis of AD. In the murine model of AD, the expression of lncRNA MALAT1 is upregulated in the skin, and its knockdown represses NLRP3 inflammasome activation and mitigates Th1/Th2 imbalance, thus potentially ameliorating AD [129]. In another study, downregulation of MALAT1 expression resulted in the attenuation of the expression of the CCR7 gene via a competing endogenous RNA mechanism involving miR-590-5p [130]. This pathway effectively inhibits TNFα/IFNγ-induced keratinocyte proliferation and inflammation. A study of macro-phages revealed that lncRNA, lncFAO, contributes to inflammation resolution and tissue repair in mice by promoting fatty acid oxidation in macrophages [131].
Taken together, these findings indicate the profound involvement of ncRNAs, especially miRNAs, in AD pathogenesis, suggesting their targeting as a promising therapeutic approach for AD. In support, one of the new anticancer drugs, the pan-HDAC inhibitor belinostat, has been shown to promote keratinocyte differentiation and restoration of skin barrier function by upregulating the expression of miR-335, which, in turn, targets the transcription factor SOX6 to promote terminal differentiation of keratinocytes [124].

4. The Role of Skin-Gut Axis in the Pathogenesis of AD

Healthy skin is colonized by a variety of commensal microorganisms while preventing the growth and penetration of pathogenic microorganisms [132,133]. AD is associated with dramatic changes in the skin microbiota, particularly the loss of commensal diversity and dominant colonization by pathogenic Staphylococcus bacteria, mainly S. aureus [46] and also S. capitis, and S. lugdunensis [134]. Not all patients with AD are colonized with S. aureus; however, a high abundance of S. aureus in AD is associated with more severe disease and greater type 2 immune deviation, allergen sensitization, and barrier disruption than in non-infected patients [135,136]. Analysis of S. aureus clinical isolates demonstrates that most pathogenic strains predominate in AD skin in correlation with disease severity [50,137], whereas restoration of commensal assortment has been suggested to precede and predict disease resolution [138,139].
Commensal skin microbiota produces tryptophan metabolites, such as indole-3-aldehyde (IAld), that block Th2 induction through the aryl hydrocarbon receptor (AHR) expressed mainly by keratinocytes [140]. The skin of AD patients displays a lower level of IA1d compared to that of healthy subjects. Collectively, these findings indicate that skin dysbiosis plays a detrimental role in AD pathogenesis, mainly via exacerbating the Th2-associated immune response. Notably, this immune dysregulation can influence the skin microbiota in AD. For example, S. aureus binds more efficiently to IL-4-stimulated skin samples compared to untreated control skin in a murine model of AD, suggesting that a Th2 inflammatory environment promotes skin binding by S. aureus [141]. Additionally, in cultured human keratinocytes, Th2 cytokines inhibit the expression of AMPs, namely human β-defensin 2 (HBD2), HBD3 and cathelicidin (LL-37), which have anti-staphylococcal activity [142,143]. A similar reduction in AMP expression was observed in skin explants from AD patients compared to healthy controls, which is reversed by neutralizing antibodies against IL-4, IL-10, and IL-13 [144,145]. Together, these data suggest the existence of a bidirectional link between skin dysbiosis and chronic inflammation in AD, and the mechanisms involved in the process are schematically summarized in Figure 3.
AD pathogenesis is also characterized by gut dysbiosis in which, similar to skin, the prevalence and diversity of commensals are reduced, whereas the prevalence of pathobionts is increased [52,55,146,147]. AD-associated gut dysbiosis has two major detrimental consequences: compromised gut barrier integrity (leaky gut) and reduced production of short-chain fatty acids (SCFAs), mainly anti-inflammatory butyrate and propionate. A leaky gut is followed by increased blood levels of intestinal endotoxins such as lipopolysaccharide (LPS), which, through binding toll-like receptor 4 (TLR4) on macrophages and T cells, trigger signaling cascades that culminate in the production of pro-inflammatory cytokines (TNFα, IL-1β, IL-6, IL-12) and type I interferons participating in the creation of chronic inflammation [148,149]. The main producers of butyrate and propionate are Faecalibacterium prausnitzii [150,151] and Dialister [152]. In fecal samples from AD patients, their numbers were reduced compared to healthy controls [153,154]. Moreover, a recent Mendelian randomization analysis suggests an inverse correlation between Dialister and AD [155]. Lack of SCFA-producing microbes preceded AD symptoms [46] and is found to be positively correlated with the disease severity in infants [53], suggesting that SCFAs play a role in improving AD manifestations [156]. In support, high fecal levels of propionate and butyrate were found to be associated with decreased atopic sensitization in children [157]. Moreover, butyrate administration attenuated skin bleeding, scarring, dryness, abrasions and erosions in an AD murine model [158]. Notably, butyrate increases the expression of mRNA and protein levels of FLG in normal human epidermal keratinocytes [159] and attenuates S. aureus-aggravated skin inflammation with decreased IL-13, IL-33, and leukocyte infiltration in the skin [54]. Moreover, butyrate derivative BA-NH-NH-BA reduces skin colonization by S. aureus and ameliorates S. aureus-induced production of IL-6 in a murine model of AD [160]. Overall, these results suggest that gut dysbiosis-induced SCFA deficiency is involved in AD-associated S. aureus accumulation in the skin, keratinocyte activation, and skin inflammation, thereby reinforcing the deleterious role of the skin-gut axis in AD pathogenesis (Figure 3) [52,147,161]. Remarkably, there is evidence suggesting a link between epigenetic disturbance and skin dysbiosis in AD pathogenesis. Indeed, low levels of propionate on the skin surface of AD patients are associated with increased expression of HDAC2 and HDAC3 [162]. Reduced concentrations of butyrate are associated with increased growth of S. aureus via inhibition of acetylation of histone H3 lysine 9 (AcH3K9) in human AD keratinocytes [160].

5. Failed Resolution of Chronic Inflammation as a Key Mechanism of AD Pathogenesis

The abundant data unequivocally show that chronic skin inflammation is the main pathogenetic mechanism of AD. Its successful resolution can, therefore, lead to a weakening of disease manifestations. Inflammation resolution is a process primarily driven by dietary polyunsaturated fatty acid (PUFA)-derived specialized pro-resolving mediators (SPMs) through the sequential enzymatic activities of several lipoxygenases and hydrolases. The omega-3 PUFA docosahexaenoic acid (DHA) serves as the substrate for D-series resolvins (RvD1-RvD6), maresins (MaR1, MaR2, and eMaR), and protectins/neuroprotectins (PD1/NPD1), cysteinyl-SPMs (MCTR1-R3, PCTR1-R3, and RCTR1-R3) as well as n-3 docosapentaenoic acid (DPA)-derived SPMs (PD1n-3 DPA), while eicosapentaenoic acid (EPA) serves as a substrate for E-series resolvins (RvE1-RvE4). Lipoxins (LXs), such as LXA4 and LXB4, are derivatives of the omega-6 PUFA arachidonic acid (AA) [163]. SPMs exert their biological actions via cognate G protein-coupled receptors (GPCRs), namely ALX/FPR2, GPR32, ChemR23, BLT1, GPR18, GRP37, and LGR6. The underlying mechanisms of inflammation resolution, which are still under investigation, have been comprehensively reviewed (e.g., [164,165,166,167,168]). In short, this multistep process involves cessation of neutrophil infiltration, counter-regulation of pro-inflammatory cytokines and chemokines, reduction of reactive oxygen species (ROS) and NLRP3 inflammasome generation, induction of neutrophil apoptosis and their efferocytosis by macrophages, accumulation of anti-inflammatory M2 macrophages, and induction of Tregs, all of which initiate the healing processes and culminate in a return to tissue homeostasis. Uncontrolled, excessive inflammation or failure to promptly resolve inflammation results in non-resolving inflammation, which is often chronic or transient and recurrent. Reduced SPM production and functional activity are hypothesized to be the underlying mechanisms of failure to resolve inflammation [169]. Inflammation resolution has emerged as a critical physiological process that protects host tissues from prolonged or excessive inflammation that can become detrimentally chronic [168].
One of the most remarkable features of SPM receptors (in particular, ALX/FPR2) is their unusually high degree of molecular heterogeneity in recognizing a wide range of ligands, indicating that not only each receptor is engaged by different SPMs but also that a single SPM can act through the activation of different receptors [170]. This mutual redundancy is reflected in the relationships between the SPMs, their receptors and immune cells during inflammation resolution. Indeed, it has been repeatedly demonstrated that most SPMs bind various receptors expressed by neutrophils, macrophages, lymphocytes, and other immune cells, thereby ensuring effective inflammation resolution [171,172,173,174]. Accordingly, dysregulation of an SPM/receptor/immune cells cross-talk is suggested as a main cause of inflammation chronicity [31,175], which maintains and exacerbates multiple associated disorders [176,177,178,179]. Hence, enhancing this cross-talk by upregulating SPM functional activity and/or targeting SPM receptors may potentially be an effective approach to attenuate the manifestations of chronic inflammation-associated disorders, including AD.
Importantly, AD patients are characterized by reduced levels of the derivatives of omega-3 PUFAs, suggesting that the natural mechanisms of inflammation resolution in AD are suppressed. In accordance with this idea, transcriptomic and lipidomic pro-filing of patients with AD revealed decreased EPA and DHA levels and omega-3/omega-6 PUFA ratio in the serum and in lesional and even non-lesional skin [180]. Moreover, an increased ratio of pro-inflammatory vs pro-resolving lipid media-tors was found, overall suggesting a strong inflammatory background towards the maintenance of chronic inflammatory status in AD, displaying no tendency of its re-solving. Two recent Mendelian randomization studies suggested a negative correlation between AD risk and omega-3 PUFA [36] and DHA [181] serum levels. In the study by Lin et al. [36], leave-one-out analyses revealed that the protective effect of omega-3 PUFA was mainly mediated by SNP (rs174546) located in the fatty acid desaturase (FADS) gene cluster, highlighting its importance in the fatty acid synthesis pathway in the development of AD. In another recent study [181], DHA levels were negatively associated with the levels of the proinflammatory cytokine TNFSF14. Moreover, a positive correlation was found between TNFSF14 levels and the risk of AD. These results imply that genetic factors have a potential role in the observed PUFA deficiency in AD.
The potential beneficial role of inflammation resolution in patients with AD was first observed in a 1987 study in which taking 10 g of fish oil (containing approximately 1.8 g of EHA) daily for 12 weeks was associated with reduced AD symptomatology [181]. Since then, similar effects of omega-3 PUFAs and their derivatives in AD patients have been repeatedly reported (e.g., [34,182,183,184,185]), although the results of meta-analyses are not consistent [186,187,188,189]. One reason for this discrepancy may be that in cases of oxidative stress, which is a key feature of chronic inflammation in AD [190], omega-3 PUFAs become very oxidized [191,192]. This, in turn, may lead to the formation of highly bioactive lipid peroxidation products such as 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), which are known to induce inflammation and other immune changes, including increased cytokine secretion and activation of inflammatory transcription factors [193,194]. These findings point to the existence of complex relationships between PUFA oxidation and their biological activity [195,196,197], which may influence the beneficial effects of omega-3 PUFA in patients with oxidative stress-related conditions, including AD.
Several preclinical studies have shown clear preventive/therapeutic effects of SPMs in skin inflammation models. For example, in a 2,4-dinitrofluorobenzene (DNFB)—a murine model of AD-like skin lesion, intraperitoneal injection of RvE1 significantly decreased ear swelling and improved skin lesions, accompanied by decreased serum IgE levels and production of IFNγ and IL-4 by activated T cells, and reduced infiltration of eosinophils, mast cells and T cells in the skin lesions [37]. In a psoriatic murine model, RvE1 suppressed the inflammatory cell infiltration, epidermal hyperplasia and mRNA expression of IL-23 in the skin and inhibited migration of cutaneous DCs and γδ T cells, a major IL-17-producing cell population in mice, presumably via binding to the BLT1 receptor [198]. In a murine model of allergic inflammation induced by intraperitoneal injections of ovalbumin (OVA), RvE1 suppressed the production of IL-23 and IL-6 in the lung [199] as well as reduced airway eosinophil and lymphocyte recruitment, IL-13 secretion, and IgE levels [200,201]. In a murine model of imiquimod (IMQ)-induced psoriasiform skin inflammation, RvD1 downregulated skin mRNA expression of IL-17, IL-22, IL-23, and TNFα via inhibition of MAPKs and NF-κB signaling pathways [202]. In a similar model, protectin D1 (PD1) reduced skin thickness, redness, and scaling, decreased IL-1β, IL-6, IL-17, and CXCL1 mRNA expression in the skin and serum via inhibition of STAT1 and NF-κB signaling pathways [203]. Notably, decreased mRNA expression of IL-1β, IL-6, IL-8, and CCL17 was also observed in keratinocytes of PD1-treated mice.
Since SPMs function through triggering their cognate receptors, these data suggest the expression of SPM receptors in keratinocytes. Indeed, human keratinocytes were shown to express ChemR23 [204] and BLT1 [205], which are both receptors for RvE1 and RvE2, as well as ALX/FRP2, the receptor for RvDs and LXA4 [206]. In the latter study, ALX/FRP2-deficient mice showed an endogenous defect in re-epithelialization, and topical application of RvD2 accelerated re-epithelialization during skin injury and enhanced migration of human epidermal keratinocytes in an ALX/FRP2-dependent manner. These data indicate that resolvins play a direct, keratinocyte-mediated role in the tissue repair program. Another study showed that mouse and human keratinocytes use FPR2 to detect S. aureus and trigger antimicrobial defenses in the skin [207]. Moreover, in the murine IMQ-induced psoriasiform inflammation model, topical application and systemic administration of RvD3 results in reduced skin inflammation, acute pain and itch [208].
Overall, these observations suggest that deficiency of SPMs in AD not only reduces the pro-resolving activity of immune cells but also affects the functional activity of SPM receptor-expressing keratinocytes, along with increasing the pain and itch associated with AD. In support, increased levels of omega-6-PUFA-derived major itch mediators, such as leukotriene B4 (LTB4), thromboxane B2 (TXB2), 12-hydroxyeicosatetraenoic acid (12-HETE), prostaglandin E2 (PGE2) and PGF2, as well as BLT1 (receptor for RvE1 and RvE2), have been detected in affected and non-affected skin of AD patients [180]. The authors suggest that in patients with AD, even healthy-looking skin is prone to a pro-inflammatory status and pro-itchy conditions, which are most likely caused by a systemic disorder [209].
Itch triggers scratching responses following activation of pruriceptors in primary sensory neurons, which are a subset of nociceptors and also express transient receptor potential vanilloid 1 and ankyrin 1 (TRPV1 and TRPA1, respectively) [210,211,212]. Both TRPV1 and TRPA1 are co-expressed in a wide range of sensory nerves, where they integrate multiple noxious stimuli, and in non-neuronal cells, such as keratinocytes, mast cells, DCs, and endothelial cells, serving as nociceptive sensors that amplify the inflammatory process [211]. AD is commonly associated with persistent itch [213], and SPMs have been shown to alleviate it by reducing skin inflammation. Indeed, topical application of LXA4 reduces the severity of childhood AD and improves the quality of life through controlling skin inflammation by downregulating TLR4, p-ERK1/2, and NF-κB signaling, and proinflammatory cytokines [214,215]. RvD3 administration prevents the development of psoriasiform itch and skin inflammation via inhibition of TRPV1 signaling in mouse and human dorsal root ganglion (DRG) neurons, which may account for the anti-itch effects of RvD3 [208]. Another study reported an important role of spinal glial cells in driving chronic itch [216]. Intrathecal administration of PD-1, PD1n-3 DPA, and the new analog 3-oxa-PD1n-3 DPA significantly reduced scratching for several hours [217]. These observations suggest the expression of SPM receptors on neuronal cells. In support, receptors for LXA4, RvD1, RvD3, RvD5 and some of their aspirin-triggered analogs were shown to bind to either astrocytic and neuronal ALX/FPR2 in both humans and mice or neuronal and microglial GPR32 receptor in humans. GPR18, the receptor for RvD2, is expressed in all brain cells except oligodendrocytes, and GPR37, the receptor for PD1, is expressed in all brain cells except microglia. RvE1/2 binds the ChemR23 receptor in astrocytes and the BLT1 receptor in astrocytes, microglia and neurons. MaR1 can bind and activate surface LgR6 receptors in astrocytes and neurons and intracellular RORα receptors expressed in all brain cells except oligodendrocytes [218,219]. Activation of these receptors by SPMs may attenuate neuroinflammation, which is known to be involved in pain/itch exacerbation [220,221,222,223]. Figure 4 schematically illustrates the hypothetical mechanisms of SPM action in resolving inflammation in AD. The beneficial effects of SPM in alleviating pain/itch also occur, among others, through the suppression of TRPV1 and TRPA1. For example, intrathecal capsaicin-induced spontaneous pain is blocked by RvE1 [224]. Capsaicin-induced pain is suppressed by MaR1, RvD2, and NPD1, but not RvD1, whereas allyl isothiocyanate-induced pain is inhibited by RvD1 and RvD2, but not RvE1, which suggests different modulation of inflammatory pain by SPM [219]. Moreover, in the IMQ-induced psoriasiform inflammation murine model, topical application and systemic administration of RvD3 resulted in reduced skin inflammation, acute pain and itch [208]. Mechanistically, RvD3 inhibited capsaicin-induced TRPV1 currents in DRG neurons via ALX/FPR2 with a concomitant decrease in calcitonin gene-related peptide (CGRP), which is also expressed by keratinocytes [225] and plays an apparent anti-inflammatory role in AD [226]. Taken together, these findings suggest that SPMs may alleviate AD-associated chronic pain/itch via effective control of skin inflammation by activation of SPM receptors on immune and non-immune cells.

6. Current AD Treatment

Recent substantial progress in deciphering the mechanisms of AD pathogenesis has brought to the forefront the immune dysregulation that we have discussed above and illustrated in Figure 1. This, in turn, has led to a real “translational revolution” [17], consisting of a significant expansion of the assortment of AD therapeutic agents, mainly including JAKi and monoclonal antibodies against key pathogenetic cytokines and their receptors [227,228,229,230]. The list of the most commonly used JAKi includes baricitinib, upadacitinib, and abrocitinib, which inhibit the signaling pathways of key cytokines involved in inflammatory diseases [231,232,233,234]. Among the monoclonal cytokine inhibitors, the most comprehensive clinical data are available for dupilumab, lebrikizumab and tralokinumab. Dupilumab acts against IL-4Rα, a common receptor to both IL-4 and IL-13. Lebrikizumab binds soluble IL-13 with a high affinity, preventing IL-4Rα/IL-13Rα1 heterodimerization (type 2 receptor). Tralokinumab binds IL-13 [61,62,235]. A comprehensive analysis of the clinical efficacy of current AD treatments has shown that high-dose upadacitinib, abrocitinib and low-dose upadacitinib are the most effective in addressing multiple patient-important outcomes. However, these JAKi are among the most harmful in terms of increasing adverse events. Dupilumab, lebrikizumab, and tralokinumab have intermediate efficacy and are among the safest. Low-dose baricitinib is one of the least effective [29].
Notably, although the therapeutic armamentarium for AD is rapidly expanding, the results of clinical trials have demonstrated that a significant number of patients fail to achieve clear or almost clear status (Investigator Global Assessment (IGA) score 0/1) with currently available treatments. For example, dupilumab achieves this score in <40% of patients when given as monotherapy or in combination with topical corticosteroids (TCSs) [17]. Less than 25% and <40% of tralokinumab-treated patients reach this score when used as monotherapy or in combination with TCSs [236]. JAK inhibitors demonstrate higher rates of achieving this score, with upadacitinib able to clear ~60% of patients and abrocitinib <50%, with slightly higher rates when used in combination with TCSs [17,29,237].
As discussed in the review, potential targets for alleviating the manifestations of AD may include agents that reduce itch. Indeed, preliminary data suggest that topical application of a cream containing N-palmitoylethanolamine (PEA), a TRVP1 antagonist, reduces itch and improves quality of life in patients with mild to moderate AD [238]. Results from the phase III study (NCT02965118) have not yet been published.

7. Application of Epidrugs, Dysbiosis Correctors and Inflammation-Resolving Agents for Alleviation of AD Manifestations

The lack of efficacy of JAKi and anti-cytokine antibodies in a significant proportion of patients with AD, as well as the unclear effect of anti-itch drugs, highlight the need for additional therapeutic approaches that could improve the outcome of AD treatment. Several potential options are schematically presented in Figure 5. The proposal starts with epidrugs because of the significant role of epigenetic disturbances in the pathogenesis of AD. Epidrugs, which are chemical factors that inhibit enzymes with epigenetic activity, are capable of restoring disrupted pathological changes in epigenetic mechanisms [239]. They, therefore, are possible candidates for future AD treatment based on their ability to control gene expression directly at the pre-transcriptional stage, thereby correcting gene dysregulation at its source [240]. Epidrugs target epigenetic marks, which are responsible for epigenetic alterations, such as DNMT and HDAC, or miRNAs. These drugs inhibit or activate disease-associated epigenetic proteins and lead to the improvement, treatment, or prevention of diseases [239,240].
Currently, epidrug research and clinical translation are mostly focused on cancer [241]. However, there is also research on other diseases, such as osteoarthritis [242], selected autoimmune diseases [243], and mental and neurodegenerative disorders [244]. Unfortunately, till the present time, clinical trials of epidrugs have led only to modest success alongside notable side effects, such as high toxicity and drug resistance. This is most probably due to the effects on off-target genes, presumably because many epigenetic regulators are not very specific in gene targeting [245]. Moreover, epidrugs may compromise gene regulation and the genomic stability of normal cells [246], altogether limiting their applicable therapeutic use. Various efforts are currently underway to overcome these obstacles and other issues arising from epigenome editing, including improvements in target specificity, enzymatic activity and drug delivery [245]. Another possibility is to combine genetic and epigenetic approaches, in which synthetic modules consisting of epigenetic regulators are coupled to a custom-designed genome targeting system (e.g., CRISPR-Cas9-based). These synthetic modules can provide precise targeting of the epigenetic regulator gene or locus, especially when used as an inhibitor [246,247,248]. This molecular engineering strategy has been explored to target miRNAs [249]. For example, the knockdown of miRNA-155 (which enhances CTLA-mediated T-cell activation) by CRISPR/Cas9 in macrophage cell lines showed a significant reduction in the development of rheumatoid arthritis-related symptoms [250]. In addition, the pan-HDAC inhibitor, belinostat, resolved skin barrier defects in AD by targeting the dysregulated miR-335:SOX6 axis [124]. In several AD murine models, selective HDAC6 inhibitor tubastatin A and selective SIRT1 inhibitor sirtinol attenuated symptoms associated with AD, accompanied by reduced expression of PGE2 and COX2, and serum levels of TSLP and chemokine CXCL13 [251]. HDAC6 knockout in mice prevented HDAC6-mediated passive cutaneous and systemic anaphylaxis [252]. Taken together, these observations suggest the potential application of epidrugs in AD.
Another potential therapeutic target is the treatment of AD-associated skin and gut dysbiosis. The results of some clinical studies [253,254,255,256] and most meta-analyses report clear beneficial effects of prebiotic/probiotic supplementation in children [257,258,259,260] and adults [260,261,262,263,264], although some meta-analyses found no difference between treatment and placebo groups [265,266]. The mechanisms by which prebiotic/probiotic supplements may benefit AD remain unclear. However, studies in other diseases associated with chronic inflammation suggest that the underlying mechanism is their ability to enhance the production of anti-inflammatory cytokines, such as IL-10 and suppress the production of pro-inflammatory cytokines, such as IL-1, IL-6, and TNFα [267,268,269,270,271,272,273]. This, in turn, suggests that the attenuation of AD resulting from the microbiota restoration occurs through the resolution of chronic inflammation.
PUFA supplementation and numerous preclinical studies of SPMs in AD models have revealed clear beneficial effects, supporting the validity of the idea of using agents that resolve chronic inflammation to treat AD. In this regard, it is noteworthy that SPMs facilitate the termination of the inflammatory response and initiate tissue repair and healing without being immunosuppressive [165,274,275]. This is in stark contrast to JAKi, anti-cytokine antibodies and corticosteroids known to suppress immune responses to pathogens. Furthermore, corticosteroids reduce SPM production and block the inhibitory effects of 17-HDHA and RvD1 on IgE production by B cells from asthma patients [276], thus potentially delaying inflammation resolution.
An additional and intriguing aspect of the potential therapeutic activity of SPMs is their ability to modulate HDACs, mainly SIRTs, confirming a link between epigenetics and chronic inflammation [277,278] and suggesting that combining pro-resolving compounds with epidrugs may enhance their efficacy. Consistent with this idea, RvE1 and lipoxin A4 (LXA4), acting via ChemR23 and ALX/FPR2 receptors, respectively, inhibited NF-κB activation and increased the expression of SIRT1, SIRT6 and SIRT7 in cultured human dental pulp fibroblasts in a synergistic manner [279]. In this study, co-administration of RvE1 and LXA4 markedly promoted the resolution of mouse pulpitis. In septic mice, RvD1 improved animal survival and attenuated lung inflammation by downregulating STAT3, NF-κB, ERK, and p38 expression through a mechanism partly dependent on SIRT1 [280]. In mice with cerebral ischemia/reperfusion, MaR1 attenuated mitochondrial damage and reduced TNFα and IL-1 production via triggering SIRT1 signaling, resulting in a reduction in infarction size and subsequent neurological defects [281]. In human macrophages and PBMCs, MaR1 reversed LPS-induced increased expression and secretion of TNFα, IL-1β and IL-6, concomitantly with increased expression of SIRT1, PGC-1α, and PPARγ [282]. Moreover, SPMs were shown to alleviate inflammation through the regulation of miRNAs. For example, in a murine model of collagen-induced arthritis, RvD1 decreased pannus formation and cartilage damage through the upregulation of miRNA-146a-5p [283]. In a murine model of systemic lupus erythematosus, RvD1 effectively ameliorated disease progression through up-regulating Tregs and down-regulating Th17 cells via miR-30e-5p [284].
However, SPMs are complex molecules that are susceptible to rapid enzymatic degradation, which limits their translational potential [163]. To overcome this problem, stable, small-molecule SPM mimetics and receptor agonists have been developed. They have shown encouraging therapeutic efficacy in various preclinical models, likely by reversing failed chronic inflammation (Reviewed in [176,177,285]). As an example, BML-111, a synthetic analog of LXA4 that is an ALF/FPR2s agonist, showed beneficial effects in animal models of pancreatitis [286], experimental autoimmune myocarditis [287,288], and chronic obstructive pulmonary disease (COPD) [289]. All of the above-mentioned beneficial effects of SPM mimetics and receptor agonists have been obtained in experimental model systems. Yet, the field of “resolution pharmacology” [290] is rapidly evolving, and the first clinical trial results of pro-inflammatory compounds have already been published, providing some insight into their translational potential [173]. For example, a pilot study of inhaled BML-111 and lipoxin A4 mimetic 5(S),6(R) LXA4 methyl ester demonstrated preliminary efficacy in the treatment of childhood asthma with acute moderate episodes [291]. Noteworthy, the treated study participants did not experience any clinical adverse events according to blood, urine, and stool tests, electrocardiogram, and liver and kidney function tests on day 7 of the intervention. In support, mice treated with BML-111 or 5(S),6(R)-LXA4 methyl ester showed no significant differences in pulmonary, renal, or liver function. Another stable LXR4, 15(R/S)-methyl-lipoxin A(4) cream demonstrated therapeutic efficacy in patients with childhood AD [214]. In a double-blind, placebo-controlled, randomized, parallel-group study, the application of benzolipoxin A4 methyl ester (BLXA4), a stable LXA4 mimetic, has effectively reduced gingival inflammation without any adverse safety signals [292]. Notably, this study shows that the topical application of BLXA4, in addition to its local effects, significantly increased circulating SPM levels.
Collectively, these observations suggest that stable SPM mimetics and receptor agonists could be applied as pharmaceuticals in disorders characterized by chronic inflammation, including AD. Given the critical role of failed resolution of chronic inflammation in the development of AD, we propose the combination of inflammation-resolving agents with epidrugs and prebiotics/probiotics as a new therapeutic approach for AD to effectively relieve the disease manifestations (Figure 5).

8. Concluding Remarks

This review provides extensive data supporting the concept that failed resolution of chronic inflammation is a key pathogenic mechanism underlying the development of AD. The establishment of chronic inflammation occurs under the synergistic effects of at least three fundamental inflammation-promoting inducers: (i) the reduced production and functional activity of SPMs, (ii) skin and gut dysbiosis, and (iii) disturbed epigenetic regulation of AD-associated pathogenic mechanisms. As such, the diminution of the activity of these inducers could attenuate AD manifestations. In accordance with this idea, we propose a multilevel treatment strategy: (1) the application of stable, small molecule SPM mimetics and receptor agonists; (2) the recovery of dysbiosis manifestation by supplementation of prebiotics/probiotics; and (3) the implementation of epidrugs that restore impaired epigenetic mechanisms. This treatment strategy can be used in conjunction with current AD therapy, which mainly includes JAKi and anti-cytokine monoclonal antibodies, resulting in the gradual mitigation of AD (Figure 5).
As discussed in the review, stable SPM mimetics and receptor agonists demonstrated beneficial effects in numerous preclinical AD and inflammation models. This may address the shortcomings of current AD immunotherapies. A critical difference between pro-resolving mediators and anti-inflammatory drugs is that although most anti-inflammatory agents can cause immunosuppression, the resolution of inflammation occurs through active endogenous reprogramming of the immune response, allowing inflammation to cease without causing immunosuppressive effects. Recently, it has been found that an agonistic antibody against the ChemR23 receptor is capable of accelerating the resolution of acute inflammation, stimulating macrophage efferocytosis and reducing neutrophil apoptosis at the site of inflammation [293]; these impressive results allow this agonistic antibody to be included in the list of potential pro-resolving agents.
However, the implementation of the proposed combinatory therapeutic strategy requires overcoming critical research challenges. One of the major ones is the search for the optimal animal model of AD. Currently, murine models of AD are classified into three groups: (i) inbred models, (ii) genetically modified mice in which the genes of interest are overexpressed or deleted in a specific cell type, and (iii) models induced by local application of exogenous agents. However, each animal model represents only limited aspects of human AD, and a significant translational gap remains between mouse AD models and human AD [294,295,296]. Therefore, the selection of the most appropriate model or any new model to test our idea of combinatorial treatment of AD is a challenge and requires further research. What are the dominant factors driving epi-genetic dysregulation, dysbiosis and chronic inflammation in AD pathogenesis? Are the relationships between these pathogenic events and AD causal, or do they rather represent independent pathological processes? In this regard, identifying and characterizing the epigenetic, dysbiosis, and chronic inflammation profiles of AD patients could provide a unique opportunity to subtype patients, thereby gaining further insight into the disease. Furthermore, in order to harness the benefits of the proposed multilevel treatment strategy, the crucial next step is to conduct a scientific search for highly specific targets (receptors, cells, signaling pathways, and microbes). However, there is currently a wide variety of possible targets, making the development of such a complex therapeutic strategy a highly challenging task. Once we overcome this challenge, it will open the door to developing personalized treatments for AD patients to significantly reduce manifestations.

Author Contributions

Both authors contributed to the study’s conception and design. Both authors wrote the first draft of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by the Ariel University Research and Development Department (Grant Number: RA2000000457) to G.L.

Data Availability Statement

No data were used for the research described in the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Faye, O.; Flohr, C.; Kabashima, K.; Ma, L.; Paller, A.S.; Rapelanoro, F.R.; Steinhoff, M.; Su, J.C.; Takaoka, R.; Wollenberg, A.; et al. Atopic Dermatitis: A Global Health Perspective. J. Eur. Acad. Dermatol. Venereol. 2024, 38, 801–811. [Google Scholar] [CrossRef] [PubMed]
  2. Werfel, T.; Heratizadeh, A.; Aberer, W.; Augustin, M.; Biedermann, T.; Bauer, A.; Fölster-Holst, R.; Kahle, J.; Kinberger, M.; Nemat, K.; et al. S3 Guideline Atopic Dermatitis: Part 1-General Aspects, Topical and Non-drug Therapies, Special Patient Groups. JDDG J. Dtsch. Dermatol. Ges. 2024, 22, 137–153. [Google Scholar] [CrossRef] [PubMed]
  3. Lv, J.; Kong, X.; Zhao, Y.; Li, X.; Guo, Z.; Zhang, Y.; Cheng, Z. Global, Regional and National Epidemiology of Allergic Disorders in Children from 1990 to 2019: Findings from the Global Burden of Disease Study 2019. BMJ Open 2024, 14, e080612. [Google Scholar] [CrossRef] [PubMed]
  4. Langan, S.M.; Irvine, A.D.; Weidinger, S. Atopic Dermatitis. Lancet 2020, 396, 345–360. [Google Scholar] [CrossRef] [PubMed]
  5. Patrick, G.J.; Archer, N.K.; Miller, L.S. Which Way Do We Go? Complex Interactions in Atopic Dermatitis Pathogenesis. J. Investig. Dermatol. 2021, 141, 274–284. [Google Scholar] [CrossRef]
  6. Ständer, S. Atopic Dermatitis. N. Engl. J. Med. 2021, 384, 1136–1143. [Google Scholar] [CrossRef]
  7. Traidl-Hoffmann, C.; Afghani, J.; Akdis, C.A.; Akdis, M.; Aydin, H.; Bärenfaller, K.; Behrendt, H.; Bieber, T.; Bigliardi, P.; Bigliardi-Qi, M.; et al. Navigating the Evolving Landscape of Atopic Dermatitis: Challenges and Future Opportunities: The 4th Davos Declaration. Allergy 2024, 79, 2605–2624. [Google Scholar] [CrossRef]
  8. Guttman-Yassky, E.; Waldman, A.; Ahluwalia, J.; Ong, P.Y.; Eichenfield, L. Atopic Dermatitis: Pathogenesis. Semin. Cutan. Med. Surg. 2017, 36, 100–103. [Google Scholar] [CrossRef]
  9. Weidinger, S.; Beck, L.A.; Bieber, T.; Kabashima, K.; Irvine, A.D. Atopic Dermatitis. Nat. Rev. Dis. Primers 2018, 4, 1. [Google Scholar] [CrossRef]
  10. Uchida, T. Preferential Expression of Th2-Type Chemokine and Its Receptor in Atopic Dermatitis. Int. Immunol. 2002, 14, 1431–1438. [Google Scholar] [CrossRef]
  11. Gittler, J.K.; Shemer, A.; Suárez-Fariñas, M.; Fuentes-Duculan, J.; Gulewicz, K.J.; Wang, C.Q.F.; Mitsui, H.; Cardinale, I.; de Guzman Strong, C.; Krueger, J.G.; et al. Progressive Activation of TH2/TH22 Cytokines and Selective Epidermal Proteins Characterizes Acute and Chronic Atopic Dermatitis. J. Allergy Clin. Immunol. 2012, 130, 1344–1354. [Google Scholar] [CrossRef] [PubMed]
  12. Suárez-Fariñas, M.; Dhingra, N.; Gittler, J.; Shemer, A.; Cardinale, I.; de Guzman Strong, C.; Krueger, J.G.; Guttman-Yassky, E. Intrinsic Atopic Dermatitis Shows Similar TH2 and Higher TH17 Immune Activation Compared with Extrinsic Atopic Dermatitis. J. Allergy Clin. Immunol. 2013, 132, 361–370. [Google Scholar] [CrossRef] [PubMed]
  13. Czarnowicki, T.; He, H.; Krueger, J.G.; Guttman-Yassky, E. Atopic Dermatitis Endotypes and Implications for Targeted Therapeutics. J. Allergy Clin. Immunol. 2019, 143, 1–11. [Google Scholar] [CrossRef] [PubMed]
  14. Tokura, Y.; Hayano, S. Subtypes of Atopic Dermatitis: From Phenotype to Endotype. Allergol. Int. 2022, 71, 14–24. [Google Scholar] [CrossRef] [PubMed]
  15. Ratchataswan, T.; Banzon, T.M.; Thyssen, J.P.; Weidinger, S.; Guttman-Yassky, E.; Phipatanakul, W. Biologics for Treatment of Atopic Dermatitis: Current Status and Future Prospect. J. Allergy Clin. Immunol. Pract. 2021, 9, 1053–1065. [Google Scholar] [CrossRef]
  16. Eichenfield, L.F.; Stripling, S.; Fung, S.; Cha, A.; O’Brien, A.; Schachner, L.A. Recent Developments and Advances in Atopic Dermatitis: A Focus on Epidemiology, Pathophysiology, and Treatment in the Pediatric Setting. Pediatr. Drugs 2022, 24, 293–305. [Google Scholar] [CrossRef]
  17. Facheris, P.; Jeffery, J.; Del Duca, E.; Guttman-Yassky, E. The Translational Revolution in Atopic Dermatitis: The Paradigm Shift from Pathogenesis to Treatment. Cell. Mol. Immunol. 2023, 20, 448–474. [Google Scholar] [CrossRef]
  18. Wollenberg, A.; Werfel, T.; Ring, J.; Ott, H.; Gieler, U.; Weidinger, S. Atopic Dermatitis in Children and Adults. Dtsch. Arztebl. Int. 2023, 120, 224–234. [Google Scholar] [CrossRef]
  19. Butala, S.; Castelo-Soccio, L.; Seshadri, R.; Simpson, E.L.; O’Shea, J.J.; Bieber, T.; Paller, A.S. Biologic Versus Small Molecule Therapy for Treating Moderate to Severe Atopic Dermatitis: Clinical Considerations. J. Allergy Clin. Immunol. Pract. 2023, 11, 1361–1373. [Google Scholar] [CrossRef]
  20. Bonelli, M.; Kerschbaumer, A.; Kastrati, K.; Ghoreschi, K.; Gadina, M.; Heinz, L.X.; Smolen, J.S.; Aletaha, D.; O’Shea, J.; Laurence, A. Selectivity, Efficacy and Safety of JAKinibs: New Evidence for a Still Evolving Story. Ann. Rheum. Dis. 2024, 83, 139–160. [Google Scholar] [CrossRef]
  21. Paller, A.S.; Spergel, J.M.; Mina-Osorio, P.; Irvine, A.D. The Atopic March and Atopic Multimorbidity: Many Trajectories, Many Pathways. J. Allergy Clin. Immunol. 2019, 143, 46–55. [Google Scholar] [CrossRef] [PubMed]
  22. Renert-Yuval, Y.; Guttman-Yassky, E. What’s New in Atopic Dermatitis. Dermatol. Clin. 2019, 37, 205–213. [Google Scholar] [CrossRef] [PubMed]
  23. Boguniewicz, M.; Leung, D.Y.M. Recent Insights into Atopic Dermatitis and Implications for Management of Infectious Complications. J. Allergy Clin. Immunol. 2010, 125, 4–13. [Google Scholar] [CrossRef] [PubMed]
  24. Tang, T.S.; Bieber, T.; Williams, H.C. Are the Concepts of Induction of Remission and Treatment of Subclinical Inflammation in Atopic Dermatitis Clinically Useful? J. Allergy Clin. Immunol. 2014, 133, 1615–1625.e1. [Google Scholar] [CrossRef] [PubMed]
  25. Bieber, T. Atopic Dermatitis: An Expanding Therapeutic Pipeline for a Complex Disease. Nat. Rev. Drug Discov. 2022, 21, 21–40. [Google Scholar] [CrossRef]
  26. Werfel, T.; Allam, J.-P.; Biedermann, T.; Eyerich, K.; Gilles, S.; Guttman-Yassky, E.; Hoetzenecker, W.; Knol, E.; Simon, H.-U.; Wollenberg, A.; et al. Cellular and Molecular Immunologic Mechanisms in Patients with Atopic Dermatitis. J. Allergy Clin. Immunol. 2016, 138, 336–349. [Google Scholar] [CrossRef]
  27. Silverberg, J.I.; Thyssen, J.P.; Fahrbach, K.; Mickle, K.; Cappelleri, J.C.; Romero, W.; Cameron, M.C.; Myers, D.E.; Clibborn, C.; DiBonaventura, M. Comparative Efficacy and Safety of Systemic Therapies Used in Moderate-to-severe Atopic Dermatitis: A Systematic Literature Review and Network Meta-analysis. J. Eur. Acad. Dermatol. Venereol. 2021, 35, 1797–1810. [Google Scholar] [CrossRef]
  28. Drucker, A.M.; Morra, D.E.; Prieto-Merino, D.; Ellis, A.G.; Yiu, Z.Z.N.; Rochwerg, B.; Di Giorgio, S.; Arents, B.W.M.; Burton, T.; Spuls, P.I.; et al. Systemic Immunomodulatory Treatments for Atopic Dermatitis. JAMA Dermatol. 2022, 158, 523. [Google Scholar] [CrossRef]
  29. Chu, A.W.L.; Wong, M.M.; Rayner, D.G.; Guyatt, G.H.; Díaz Martinez, J.P.; Ceccacci, R.; Zhao, I.X.; McMullen, E.; Srivastava, A.; Wang, J.; et al. Systemic Treatments for Atopic Dermatitis (Eczema): Systematic Review and Network Meta-Analysis of Randomized Trials. J. Allergy Clin. Immunol. 2023, 152, 1470–1492. [Google Scholar] [CrossRef]
  30. Buckley, C.D.; Gilroy, D.W.; Serhan, C.N. Proresolving Lipid Mediators and Mechanisms in the Resolution of Acute Inflammation. Immunity 2014, 40, 315–327. [Google Scholar] [CrossRef]
  31. Serhan, C.N.; Levy, B.D. Resolvins in Inflammation: Emergence of the pro-Resolving Superfamily of Mediators. J. Clin. Investig. 2018, 128, 2657–2669. [Google Scholar] [CrossRef] [PubMed]
  32. Reese, I.; Werfel, T. Do Long-chain Omega-3 Fatty Acids Protect from Atopic Dermatitis? JDDG J. Dtsch. Dermatol. Ges. 2015, 13, 879–885. [Google Scholar] [CrossRef] [PubMed]
  33. Williams, H.; Chalmers, J. Prevention of Atopic Dermatitis. Acta Derm. Venereol. 2020, 100, adv00166. [Google Scholar] [CrossRef] [PubMed]
  34. Balić, A.; Vlašić, D.; Žužul, K.; Marinović, B.; Bukvić Mokos, Z. Omega-3 Versus Omega-6 Polyunsaturated Fatty Acids in the Prevention and Treatment of Inflammatory Skin Diseases. Int. J. Mol. Sci. 2020, 21, 741. [Google Scholar] [CrossRef] [PubMed]
  35. Olejnik, A.; Gornowicz-Porowska, J.; Jenerowicz, D.; Polańska, A.; Dobrzyńska, M.; Przysławski, J.; Sansone, A.; Ferreri, C. Fatty Acids Profile and the Relevance of Membranes as the Target of Nutrition-Based Strategies in Atopic Dermatitis: A Narrative Review. Nutrients 2023, 15, 3857. [Google Scholar] [CrossRef]
  36. Lin, J.-Y.; Ma, L.-J.; Yuan, J.-P.; Yu, P.; Bai, B.-X. Causal Effects of Fatty Acids on Atopic Dermatitis: A Mendelian Randomization Study. Front. Nutr. 2023, 10, 1083455. [Google Scholar] [CrossRef]
  37. Kim, T.-H.; Kim, G.-D.; Jin, Y.-H.; Park, Y.S.; Park, C.-S. Omega-3 Fatty Acid-Derived Mediator, Resolvin E1, Ameliorates 2,4-Dinitrofluorobenzene-Induced Atopic Dermatitis in NC/Nga Mice. Int. Immunopharmacol. 2012, 14, 384–391. [Google Scholar] [CrossRef]
  38. Hu, F.; Qu, Z.; Chen, K.; Zhang, P.; Wang, B.; Jiang, R.; Zuo, Y.; Xia, P.; Chen, H. Lipoxin A4 Ameliorates Imiquimod-Induced Psoriasis-Like Dermatitis via Promoting the Regression of Inflammation. Clin. Cosmet. Investig. Dermatol. 2023, 16, 2103–2111. [Google Scholar] [CrossRef]
  39. Schmidt, A.D.; de Guzman Strong, C. Current Understanding of Epigenetics in Atopic Dermatitis. Exp. Dermatol. 2021, 30, 1150–1155. [Google Scholar] [CrossRef]
  40. Nedoszytko, B.; Reszka, E.; Gutowska-Owsiak, D.; Trzeciak, M.; Lange, M.; Jarczak, J.; Niedoszytko, M.; Jablonska, E.; Romantowski, J.; Strapagiel, D.; et al. Genetic and Epigenetic Aspects of Atopic Dermatitis. Int. J. Mol. Sci. 2020, 21, 6484. [Google Scholar] [CrossRef]
  41. Andersen, B.; Millar, S. Skin Epigenetics. Exp. Dermatol. 2021, 30, 1004–1008. [Google Scholar] [CrossRef] [PubMed]
  42. Yu, X.; Wang, M.; Li, L.; Zhang, L.; Chan, M.T.V.; Wu, W.K.K. MicroRNAs in Atopic Dermatitis: A Systematic Review. J. Cell. Mol. Med. 2020, 24, 5966–5972. [Google Scholar] [CrossRef] [PubMed]
  43. Weidner, J.; Bartel, S.; Kılıç, A.; Zissler, U.M.; Renz, H.; Schwarze, J.; Schmidt-Weber, C.B.; Maes, T.; Rebane, A.; Krauss-Etschmann, S.; et al. Spotlight on MicroRNAs in Allergy and Asthma. Allergy 2021, 76, 1661–1678. [Google Scholar] [CrossRef]
  44. Yang, S.-C.; Alalaiwe, A.; Lin, Z.-C.; Lin, Y.-C.; Aljuffali, I.A.; Fang, J.-Y. Anti-Inflammatory MicroRNAs for Treating Inflammatory Skin Diseases. Biomolecules 2022, 12, 1072. [Google Scholar] [CrossRef] [PubMed]
  45. Moltrasio, C.; Romagnuolo, M.; Marzano, A.V. Epigenetic Mechanisms of Epidermal Differentiation. Int. J. Mol. Sci. 2022, 23, 4874. [Google Scholar] [CrossRef] [PubMed]
  46. Byrd, A.L.; Deming, C.; Cassidy, S.K.B.; Harrison, O.J.; Ng, W.-I.; Conlan, S.; Belkaid, Y.; Segre, J.A.; Kong, H.H. Staphylococcus aureus and Staphylococcus epidermidis Strain Diversity Underlying Pediatric Atopic Dermatitis. Sci. Transl. Med. 2017, 9, eaal4651. [Google Scholar] [CrossRef]
  47. Simpson, E.L.; Villarreal, M.; Jepson, B.; Rafaels, N.; David, G.; Hanifin, J.; Taylor, P.; Boguniewicz, M.; Yoshida, T.; De Benedetto, A.; et al. Patients with Atopic Dermatitis Colonized with Staphylococcus aureus Have a Distinct Phenotype and Endotype. J. Investig. Dermatol. 2018, 138, 2224–2233. [Google Scholar] [CrossRef]
  48. Edslev, S.; Agner, T.; Andersen, P. Skin Microbiome in Atopic Dermatitis. Acta Derm. Venereol. 2020, 100, adv00164. [Google Scholar] [CrossRef]
  49. Koh, L.F.; Ong, R.Y.; Common, J.E. Skin Microbiome of Atopic Dermatitis. Allergol. Int. 2022, 71, 31–39. [Google Scholar] [CrossRef]
  50. Demessant-Flavigny, A.; Connétable, S.; Kerob, D.; Moreau, M.; Aguilar, L.; Wollenberg, A. Skin Microbiome Dysbiosis and the Role of Staphylococcus aureus in Atopic Dermatitis in Adults and Children: A Narrative Review. J. Eur. Acad. Dermatol. Venereol. 2023, 37, 3–17. [Google Scholar] [CrossRef]
  51. Pothmann, A.; Illing, T.; Wiegand, C.; Hartmann, A.A.; Elsner, P. The Microbiome and Atopic Dermatitis: A Review. Am. J. Clin. Dermatol. 2019, 20, 749–761. [Google Scholar] [CrossRef] [PubMed]
  52. Wrześniewska, M.; Wołoszczak, J.; Świrkosz, G.; Szyller, H.; Gomułka, K. The Role of the Microbiota in the Pathogenesis and Treatment of Atopic Dermatitis—A Literature Review. Int. J. Mol. Sci. 2024, 25, 6539. [Google Scholar] [CrossRef] [PubMed]
  53. Nylund, L.; Nermes, M.; Isolauri, E.; Salminen, S.; de Vos, W.M.; Satokari, R. Severity of Atopic Disease Inversely Correlates with Intestinal Microbiota Diversity and Butyrate-Producing Bacteria. Allergy 2015, 70, 241–244. [Google Scholar] [CrossRef] [PubMed]
  54. Luo, C.-H.; Lai, A.C.-Y.; Chang, Y.-J. Butyrate Inhibits Staphylococcus aureus-Aggravated Dermal IL-33 Expression and Skin Inflammation through Histone Deacetylase Inhibition. Front. Immunol. 2023, 14, 1114699. [Google Scholar] [CrossRef]
  55. De Pessemier, B.; Grine, L.; Debaere, M.; Maes, A.; Paetzold, B.; Callewaert, C. Gut–Skin Axis: Current Knowledge of the Interrelationship between Microbial Dysbiosis and Skin Conditions. Microorganisms 2021, 9, 353. [Google Scholar] [CrossRef]
  56. Moniaga, C.S.; Tominaga, M.; Takamori, K. An Altered Skin and Gut Microbiota Are Involved in the Modulation of Itch in Atopic Dermatitis. Cells 2022, 11, 3930. [Google Scholar] [CrossRef]
  57. Rothenberg-Lausell, C.; Bar, J.; Del Duca, E.; Guttman-Yassky, E. Diversity of Atopic Dermatitis and Selection of Immune Targets. Ann. Allergy Asthma Immunol. 2024, 132, 177–186. [Google Scholar] [CrossRef]
  58. Martin, M.J.; Estravís, M.; García-Sánchez, A.; Dávila, I.; Isidoro-García, M.; Sanz, C. Genetics and Epigenetics of Atopic Dermatitis: An Updated Systematic Review. Genes 2020, 11, 442. [Google Scholar] [CrossRef]
  59. Brys, A.K.; Rodriguez-Homs, L.G.; Suwanpradid, J.; Atwater, A.R.; MacLeod, A.S. Shifting Paradigms in Allergic Contact Dermatitis: The Role of Innate Immunity. J. Investig. Dermatol. 2020, 140, 21–28. [Google Scholar] [CrossRef]
  60. Yamaguchi, H.L.; Yamaguchi, Y.; Peeva, E. Role of Innate Immunity in Allergic Contact Dermatitis: An Update. Int. J. Mol. Sci. 2023, 24, 12975. [Google Scholar] [CrossRef]
  61. Fania, L.; Moretta, G.; Antonelli, F.; Scala, E.; Abeni, D.; Albanesi, C.; Madonna, S. Multiple Roles for Cytokines in Atopic Dermatitis: From Pathogenic Mediators to Endotype-Specific Biomarkers to Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 2684. [Google Scholar] [CrossRef] [PubMed]
  62. Yamamura, Y.; Nakashima, C.; Otsuka, A. Interplay of Cytokines in the Pathophysiology of Atopic Dermatitis: Insights from Murin Models and Human. Front. Med. 2024, 11, 1342176. [Google Scholar] [CrossRef] [PubMed]
  63. Krupka-Olek, M.; Bożek, A.; Aebisher, D.; Bartusik-Aebisher, D.; Cieślar, G.; Kawczyk-Krupka, A. Potential Aspects of the Use of Cytokines in Atopic Dermatitis. Biomedicines 2024, 12, 867. [Google Scholar] [CrossRef] [PubMed]
  64. Çetinarslan, T.; Kümper, L.; Fölster-Holst, R. The Immunological and Structural Epidermal Barrier Dysfunction and Skin Microbiome in Atopic Dermatitis-an Update. Front. Mol. Biosci. 2023, 10, 1159404. [Google Scholar] [CrossRef] [PubMed]
  65. Yoshida, T.; Beck, L.A.; De Benedetto, A. Skin Barrier Defects in Atopic Dermatitis: From Old Idea to New Opportunity. Allergol. Int. 2022, 71, 3–13. [Google Scholar] [CrossRef]
  66. Izquierdo, E.; Rodriguez-Coira, J.; Delgado-Dolset, M.; Gomez-Casado, C.; Barber, D.; Escribese, M. Epithelial Barrier: Protector and Trigger of Allergic Disorders. J. Investig. Allergy Clin. Immunol. 2022, 32, 81–96. [Google Scholar] [CrossRef]
  67. Schuler, C.F.; Billi, A.C.; Maverakis, E.; Tsoi, L.C.; Gudjonsson, J.E. Novel Insights into Atopic Dermatitis. J. Allergy Clin. Immunol. 2023, 151, 1145–1154. [Google Scholar] [CrossRef]
  68. Katsarou, S.; Makris, M.; Vakirlis, E.; Gregoriou, S. The Role of Tight Junctions in Atopic Dermatitis: A Systematic Review. J. Clin. Med. 2023, 12, 1538. [Google Scholar] [CrossRef]
  69. Luger, T.; Amagai, M.; Dreno, B.; Dagnelie, M.-A.; Liao, W.; Kabashima, K.; Schikowski, T.; Proksch, E.; Elias, P.M.; Simon, M.; et al. Atopic Dermatitis: Role of the Skin Barrier, Environment, Microbiome, and Therapeutic Agents. J. Dermatol. Sci. 2021, 102, 142–157. [Google Scholar] [CrossRef]
  70. Makowska, K.; Nowaczyk, J.; Blicharz, L.; Waśkiel-Burnat, A.; Czuwara, J.; Olszewska, M.; Rudnicka, L. Immunopathogenesis of Atopic Dermatitis: Focus on Interleukins as Disease Drivers and Therapeutic Targets for Novel Treatments. Int. J. Mol. Sci. 2023, 24, 781. [Google Scholar] [CrossRef]
  71. Thepen, T.; Langeveldwildschut, E.; Bihari, I.; Wichen, D.; Reijsen, F.; Mudde, G.; Bruijnzeelkoomen, C. Biphasic Response against Aeroallergen in Atopic Dermatitis Showing a Switch from an Initial TH2 Response to a TH1 Response in Situ: An Immunocytochemical Study. J. Allergy Clin. Immunol. 1996, 97, 828–837. [Google Scholar] [CrossRef] [PubMed]
  72. Yamanaka, K.; Mizutani, H. The Role of Cytokines/Chemokines in the Pathogenesis of Atopic Dermatitis. In Pathogenesis and Management of Atopic Dermatitis; S.Karger AG: Basel, Switzerland, 2011; pp. 80–92. [Google Scholar]
  73. Brandt, E.B.; Sivaprasad, U. Th2 Cytokines and Atopic Dermatitis. J. Clin. Cell. Immunol. 2011, 2, 110. [Google Scholar] [CrossRef] [PubMed]
  74. Tsoi, L.C.; Rodriguez, E.; Stölzl, D.; Wehkamp, U.; Sun, J.; Gerdes, S.; Sarkar, M.K.; Hübenthal, M.; Zeng, C.; Uppala, R.; et al. Progression of Acute-to-Chronic Atopic Dermatitis Is Associated with Quantitative Rather than Qualitative Changes in Cytokine Responses. J. Allergy Clin. Immunol. 2020, 145, 1406–1415. [Google Scholar] [CrossRef] [PubMed]
  75. Nemmer, J.M.; Kuchner, M.; Datsi, A.; Oláh, P.; Julia, V.; Raap, U.; Homey, B. Interleukin-31 Signaling Bridges the Gap Between Immune Cells, the Nervous System and Epithelial Tissues. Front. Med. 2021, 8, 639097. [Google Scholar] [CrossRef] [PubMed]
  76. Das, P.; Mounika, P.; Yellurkar, M.L.; Prasanna, V.S.; Sarkar, S.; Velayutham, R.; Arumugam, S. Keratinocytes: An Enigmatic Factor in Atopic Dermatitis. Cells 2022, 11, 1683. [Google Scholar] [CrossRef]
  77. Ong, P.Y. Atopic Dermatitis: Is Innate or Adaptive Immunity in Control? A Clinical Perspective. Front. Immunol. 2022, 13, 943640. [Google Scholar] [CrossRef]
  78. Luo, J.; Zhu, Z.; Zhai, Y.; Zeng, J.; Li, L.; Wang, D.; Deng, F.; Chang, B.; Zhou, J.; Sun, L. The Role of TSLP in Atopic Dermatitis: From Pathogenetic Molecule to Therapeutical Target. Mediat. Inflamm. 2023, 2023, 7697699. [Google Scholar] [CrossRef]
  79. Savva, M.; Papadopoulos, N.G.; Gregoriou, S.; Katsarou, S.; Papapostolou, N.; Makris, M.; Xepapadaki, P. Recent Advancements in the Atopic Dermatitis Mechanism. Front. Biosci. 2024, 29, 84. [Google Scholar] [CrossRef]
  80. Feinberg, A.P. The Key Role of Epigenetics in Human Disease Prevention and Mitigation. N. Engl. J. Med. 2018, 378, 1323–1334. [Google Scholar] [CrossRef]
  81. Cavalli, G.; Heard, E. Advances in Epigenetics Link Genetics to the Environment and Disease. Nature 2019, 571, 489–499. [Google Scholar] [CrossRef]
  82. Möbus, L.; Weidinger, S.; Emmert, H. Epigenetic Factors Involved in the Pathophysiology of Inflammatory Skin Diseases. J. Allergy Clin. Immunol. 2020, 145, 1049–1060. [Google Scholar] [CrossRef] [PubMed]
  83. Sawada, Y.; Gallo, R.L. Role of Epigenetics in the Regulation of Immune Functions of the Skin. J. Investig. Dermatol. 2021, 141, 1157–1166. [Google Scholar] [CrossRef] [PubMed]
  84. Yi, J.Z.; McGee, J.S. Epigenetic-modifying Therapies: An Emerging Avenue for the Treatment of Inflammatory Skin Diseases. Exp. Dermatol. 2021, 30, 1167–1176. [Google Scholar] [CrossRef] [PubMed]
  85. Gibson, F.; Hanly, A.; Grbic, N.; Grunberg, N.; Wu, M.; Collard, M.; Alani, R.M. Epigenetic Dysregulation in Autoimmune and Inflammatory Skin Diseases. Clin. Rev. Allergy Immunol. 2022, 63, 447–471. [Google Scholar] [CrossRef]
  86. Deichmann, U. Epigenetics: The Origins and Evolution of a Fashionable Topic. Dev. Biol. 2016, 416, 249–254. [Google Scholar] [CrossRef]
  87. Deaton, A.M.; Bird, A. CpG Islands and the Regulation of Transcription. Genes Dev. 2011, 25, 1010–1022. [Google Scholar] [CrossRef]
  88. Jones, P.A. Functions of DNA Methylation: Islands, Start Sites, Gene Bodies and Beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef]
  89. Ma, K.; Lu, N.; Zou, F.; Meng, F.-Z. Sirtuins as Novel Targets in the Pathogenesis of Airway Inflammation in Bronchial Asthma. Eur. J. Pharmacol. 2019, 865, 172670. [Google Scholar] [CrossRef]
  90. Ming, M.; Zhao, B.; Shea, C.R.; Shah, P.; Qiang, L.; White, S.R.; Sims, D.M.; He, Y.-Y. Loss of Sirtuin 1 (SIRT1) Disrupts Skin Barrier Integrity and Sensitizes Mice to Epicutaneous Allergen Challenge. J. Allergy Clin. Immunol. 2015, 135, 936–945.e4. [Google Scholar] [CrossRef]
  91. Gillette, T.G.; Hill, J.A. Readers, Writers, and Erasers. Circ. Res. 2015, 116, 1245–1253. [Google Scholar] [CrossRef]
  92. Torres, I.O.; Fujimori, D.G. Functional Coupling between Writers, Erasers and Readers of Histone and DNA Methylation. Curr. Opin. Struct. Biol. 2015, 35, 68–75. [Google Scholar] [CrossRef] [PubMed]
  93. Biswas, S.; Rao, C.M. Epigenetic Tools (The Writers, The Readers and The Erasers) and Their Implications in Cancer Therapy. Eur. J. Pharmacol. 2018, 837, 8–24. [Google Scholar] [CrossRef] [PubMed]
  94. Rodríguez, E.; Baurecht, H.; Wahn, A.F.; Kretschmer, A.; Hotze, M.; Zeilinger, S.; Klopp, N.; Illig, T.; Schramm, K.; Prokisch, H.; et al. An Integrated Epigenetic and Transcriptomic Analysis Reveals Distinct Tissue-Specific Patterns of DNA Methylation Associated with Atopic Dermatitis. J. Investig. Dermatol. 2014, 134, 1873–1883. [Google Scholar] [CrossRef]
  95. Saito-Sasaki, N.; Sawada, Y. S100 Proteins in the Pathogenesis of Psoriasis and Atopic Dermatitis. Diagnostics 2023, 13, 3167. [Google Scholar] [CrossRef]
  96. Liang, Y.; Wang, P.; Zhao, M.; Liang, G.; Yin, H.; Zhang, G.; Wen, H.; Lu, Q. Demethylation of the FCER1G Promoter Leads to FcεRI Overexpression on Monocytes of Patients with Atopic Dermatitis. Allergy 2012, 67, 424–430. [Google Scholar] [CrossRef]
  97. Lee, J.; Jang, A.; Seo, S.J.; Myung, S.C. Epigenetic Regulation of Filaggrin Gene Expression in Human Epidermal Keratinocytes. Ann. Dermatol. 2020, 32, 122. [Google Scholar] [CrossRef]
  98. Ziyab, A.H.; Karmaus, W.; Holloway, J.W.; Zhang, H.; Ewart, S.; Arshad, S.H. DNA Methylation of the Filaggrin Gene Adds to the Risk of Eczema Associated with Loss-of-function Variants. J. Eur. Acad. Dermatol. Venereol. 2013, 27, e420–e423. [Google Scholar] [CrossRef]
  99. Han, J.; Park, S.-G.; Bae, J.-B.; Choi, J.; Lyu, J.-M.; Park, S.H.; Kim, H.S.; Kim, Y.-J.; Kim, S.; Kim, T.-Y. The Characteristics of Genome-Wide DNA Methylation in Naïve CD4+ T Cells of Patients with Psoriasis or Atopic Dermatitis. Biochem. Biophys. Res. Commun. 2012, 422, 157–163. [Google Scholar] [CrossRef]
  100. Acevedo, N.; Benfeitas, R.; Katayama, S.; Bruhn, S.; Andersson, A.; Wikberg, G.; Lundeberg, L.; Lindvall, J.M.; Greco, D.; Kere, J.; et al. Epigenetic Alterations in Skin Homing CD4+CLA+ T Cells of Atopic Dermatitis Patients. Sci. Rep. 2020, 10, 18020. [Google Scholar] [CrossRef]
  101. Nakamura, T.; Sekigawa, I.; Ogasawara, H.; Mitsuishi, K.; Hira, K.; Ikeda, S.; Ogawa, H. Expression of DNMT-1 in Patients with Atopic Dermatitis. Arch. Dermatol. Res. 2006, 298, 253–256. [Google Scholar] [CrossRef]
  102. Luo, Y.; Zhou, B.; Zhao, M.; Tang, J.; Lu, Q. Promoter Demethylation Contributes to TSLP Overexpression in Skin Lesions of Patients with Atopic Dermatitis. Clin. Exp. Dermatol. 2014, 39, 48–53. [Google Scholar] [CrossRef] [PubMed]
  103. Noh, Y.-H.; Lee, J.; Seo, S.J.; Myung, S.C. Promoter DNA Methylation Contributes to Human β -Defensin-1 Deficiency in Atopic Dermatitis. Anim. Cells Syst. 2018, 22, 172–177. [Google Scholar] [CrossRef] [PubMed]
  104. Stevens, M.L.; Zhang, Z.; Johansson, E.; Ray, S.; Jagpal, A.; Ruff, B.P.; Kothari, A.; He, H.; Martin, L.J.; Ji, H.; et al. Disease-Associated KIF3A Variants Alter Gene Methylation and Expression Impacting Skin Barrier and Atopic Dermatitis Risk. Nat. Commun. 2020, 11, 4092. [Google Scholar] [CrossRef] [PubMed]
  105. Bielach-Bazyluk, A.; Zbroch, E.; Mysliwiec, H.; Rydzewska-Rosolowska, A.; Kakareko, K.; Flisiak, I.; Hryszko, T. Sirtuin 1 and Skin: Implications in Intrinsic and Extrinsic Aging—A Systematic Review. Cells 2021, 10, 813. [Google Scholar] [CrossRef] [PubMed]
  106. Ahn, S.S.; Yeo, H.; Jung, E.; Lim, Y.; Lee, Y.H.; Shin, S.Y. FRA1:C-JUN:HDAC1 Complex down-Regulates Filaggrin Expression upon TNFα and IFNγ Stimulation in Keratinocytes. Proc. Natl. Acad. Sci. USA 2022, 119, e2123451119. [Google Scholar] [CrossRef]
  107. Ma, X.; Ru, Y.; Luo, Y.; Kuai, L.; Chen, Q.-L.; Bai, Y.; Liu, Y.-Q.; Chen, J.; Luo, Y.; Song, J.-K.; et al. Post-Translational Modifications in Atopic Dermatitis: Current Research and Clinical Relevance. Front. Cell Dev. Biol. 2022, 10, 942838. [Google Scholar] [CrossRef]
  108. Dopytalska, K.; Czaplicka, A.; Szymańska, E.; Walecka, I. The Essential Role of MicroRNAs in Inflammatory and Autoimmune Skin Diseases—A Review. Int. J. Mol. Sci. 2023, 24, 9130. [Google Scholar] [CrossRef]
  109. Brancaccio, R.; Murdaca, G.; Casella, R.; Loverre, T.; Bonzano, L.; Nettis, E.; Gangemi, S. MiRNAs’ Cross-Involvement in Skin Allergies: A New Horizon for the Pathogenesis, Diagnosis and Therapy of Atopic Dermatitis, Allergic Contact Dermatitis and Chronic Spontaneous Urticaria. Biomedicines 2023, 11, 1266. [Google Scholar] [CrossRef]
  110. Sonkoly, E.; Janson, P.; Majuri, M.-L.; Savinko, T.; Fyhrquist, N.; Eidsmo, L.; Xu, N.; Meisgen, F.; Wei, T.; Bradley, M.; et al. MiR-155 Is Overexpressed in Patients with Atopic Dermatitis and Modulates T-Cell Proliferative Responses by Targeting Cytotoxic T Lymphocyte–Associated Antigen 4. J. Allergy Clin. Immunol. 2010, 126, 581–589.e20. [Google Scholar] [CrossRef]
  111. Ma, L.; Xue, H.-B.; Wang, F.; Shu, C.-M.; Zhang, J.-H. MicroRNA-155 May Be Involved in the Pathogenesis of Atopic Dermatitis by Modulating the Differentiation and Function of T Helper Type 17 (Th17) Cells. Clin. Exp. Immunol. 2015, 181, 142–149. [Google Scholar] [CrossRef]
  112. Yao, R.; Ma, Y.-L.; Liang, W.; Li, H.-H.; Ma, Z.-J.; Yu, X.; Liao, Y.-H. MicroRNA-155 Modulates Treg and Th17 Cells Differentiation and Th17 Cell Function by Targeting SOCS1. PLoS ONE 2012, 7, e46082. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, X.; Chen, Y.; Yuan, W.; Yao, L.; Wang, S.; Jia, Z.; Wu, P.; Li, L.; Wei, P.; Wang, X.; et al. MicroRNA-155-5p Is a Key Regulator of Allergic Inflammation, Modulating the Epithelial Barrier by Targeting PKIα. Cell Death Dis. 2019, 10, 884. [Google Scholar] [CrossRef] [PubMed]
  114. Taganov, K.D.; Boldin, M.P.; Chang, K.-J.; Baltimore, D. NF-ΚB-Dependent Induction of MicroRNA MiR-146, an Inhibitor Targeted to Signaling Proteins of Innate Immune Responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef] [PubMed]
  115. Rebane, A.; Runnel, T.; Aab, A.; Maslovskaja, J.; Rückert, B.; Zimmermann, M.; Plaas, M.; Kärner, J.; Treis, A.; Pihlap, M.; et al. MicroRNA-146a Alleviates Chronic Skin Inflammation in Atopic Dermatitis Through Suppression of Innate Immune Responses in Keratinocytes. J. Allergy Clin. Immunol. 2014, 4, 836–847.e11. [Google Scholar] [CrossRef] [PubMed]
  116. Yang, Z.; Zeng, B.; Wang, C.; Wang, H.; Huang, P.; Pan, Y. MicroRNA-124 Alleviates Chronic Skin Inflammation in Atopic Eczema via Suppressing Innate Immune Responses in Keratinocytes. Cell. Immunol. 2017, 319, 53–60. [Google Scholar] [CrossRef]
  117. Cheng, S.; Di, Z.; Hirman, A.R.; Zheng, H.; Duo, L.; Zhai, Q.; Xu, J. MiR-375-3p Alleviates the Severity of Inflammation through Targeting YAP1/LEKTI Pathway in HaCaT Cells. Biosci. Biotechnol. Biochem. 2020, 84, 2005–2013. [Google Scholar] [CrossRef]
  118. Beheshti, R.; Halstead, S.; McKeone, D.; Hicks, S.D. Understanding Immunological Origins of Atopic Dermatitis through Multi-omic Analysis. Pediatr. Allergy Immunol. 2022, 33, e13817. [Google Scholar] [CrossRef]
  119. Vaher, H.; Runnel, T.; Urgard, E.; Aab, A.; Carreras Badosa, G.; Maslovskaja, J.; Abram, K.; Raam, L.; Kaldvee, B.; Annilo, T.; et al. MiR-10a-5p Is Increased in Atopic Dermatitis and Has Capacity to Inhibit Keratinocyte Proliferation. Allergy 2019, 74, 2146–2156. [Google Scholar] [CrossRef]
  120. Gu, C.; Li, Y.; Wu, J.; Xu, J. IFN-γ-Induced MicroRNA-29b up-Regulation Contributes Tokeratinocyte Apoptosis in Atopic Dermatitis through Inhibiting Bcl2L2. Int. J. Clin. Exp. Pathol. 2017, 10, 10117–10126. [Google Scholar]
  121. Chen, X.; Zhang, L.; Zhang, J.; Dou, X.; Shao, Y.; Jia, X.; Zhang, W.; Yu, B. MiR-151a Is Involved in the Pathogenesis of Atopic Dermatitis by Regulating Interleukin-12 Receptor Β2. Exp. Dermatol. 2018, 27, 427–432. [Google Scholar] [CrossRef]
  122. Ullrich, K.A.M.; Schulze, L.L.; Paap, E.M.; Müller, T.M.; Neurath, M.F.; Zundler, S. Immunology of IL-12: An Update on Functional Activities and Implications for Disease. EXCLI J. 2020, 11, 1563–1589. [Google Scholar]
  123. Zeng, Y.-P.; Nguyen, G.H.; Jin, H.-Z. MicroRNA-143 Inhibits IL-13-Induced Dysregulation of the Epidermal Barrier-Related Proteins in Skin Keratinocytes via Targeting to IL-13Rα1. Mol. Cell. Biochem. 2016, 416, 63–70. [Google Scholar] [CrossRef] [PubMed]
  124. Liew, W.C.; Sundaram, G.M.; Quah, S.; Lum, G.G.; Tan, J.S.L.; Ramalingam, R.; Common, J.E.A.; Tang, M.B.Y.; Lane, E.B.; Thng, S.T.G.; et al. Belinostat Resolves Skin Barrier Defects in Atopic Dermatitis by Targeting the Dysregulated MiR-335:SOX6 Axis. J. Allergy Clin. Immunol. 2020, 146, 606–620.e12. [Google Scholar] [CrossRef] [PubMed]
  125. Li, H.M.; Xiao, Y.J.; Min, Z.S.; Tan, C. Identification and Interaction Analysis of Key Genes and MicroRNAs in Atopic Dermatitis by Bioinformatics Analysis. Clin. Exp. Dermatol. 2019, 44, 257–264. [Google Scholar] [CrossRef] [PubMed]
  126. Esaki, H.; Ewald, D.A.; Ungar, B.; Rozenblit, M.; Zheng, X.; Xu, H.; Estrada, Y.D.; Peng, X.; Mitsui, H.; Litman, T.; et al. Identification of Novel Immune and Barrier Genes in Atopic Dermatitis by Means of Laser Capture Microdissection. J. Allergy Clin. Immunol. 2015, 135, 153–163. [Google Scholar] [CrossRef] [PubMed]
  127. Malaisse, J.; Bourguignon, V.; De Vuyst, E.; Lambert de Rouvroit, C.; Nikkels, A.F.; Flamion, B.; Poumay, Y. Hyaluronan Metabolism in Human Keratinocytes and Atopic Dermatitis Skin Is Driven by a Balance of Hyaluronan Synthases 1 and 3. J. Investig. Dermatol. 2014, 134, 2174–2182. [Google Scholar] [CrossRef]
  128. Wang, J.; Huang, Y.; Wu, X.; Li, D. MicroRNA-939 Amplifies Staphylococcus aureus-Induced Matrix Metalloproteinase Expression in Atopic Dermatitis. Front. Immunol. 2024, 15, 1354154. [Google Scholar] [CrossRef]
  129. Zhao, W.; Yu, H.-H.; Meng, W.-W.; Liu, A.-M.; Zhang, B.-X.; Wang, Y.; Li, J.; Wang, L.; Fang, Y.-F. Icariin Restrains NLRP3 Inflammasome-Mediated Th2 Immune Responses and Ameliorates Atopic Dermatitis Through Modulating a Novel LncRNA MALAT1/MiR-124-3p Axis. Pharm. Biol. 2023, 61, 1249–1259. [Google Scholar] [CrossRef]
  130. Yu, H.; Zhao, W.; Zhang, B.; Wang, Y.; Li, J.; Fang, Y. Morinda officinalis Extract Exhibits Protective Effects against Atopic Dermatitis by Regulating the MALAT1/miR-590-5p/CCR7 Axis. J. Cosmet. Dermatol. 2023, 22, 1602–1612. [Google Scholar] [CrossRef]
  131. Nakayama, Y.; Fujiu, K.; Yuki, R.; Oishi, Y.; Morioka, M.S.; Isagawa, T.; Matsuda, J.; Oshima, T.; Matsubara, T.; Sugita, J.; et al. A Long Noncoding RNA Regulates Inflammation Resolution by Mouse Macrophages Through Fatty Acid Oxidation Activation. Proc. Natl. Acad. Sci. USA 2020, 117, 14365–14375. [Google Scholar] [CrossRef]
  132. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The Human Skin Microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef] [PubMed]
  133. Paller, A.S.; Kong, H.H.; Seed, P.; Naik, S.; Scharschmidt, T.C.; Gallo, R.L.; Luger, T.; Irvine, A.D. The Microbiome in Patients with Atopic Dermatitis. J. Allergy Clin. Immunol. 2019, 143, 26–35. [Google Scholar] [CrossRef] [PubMed]
  134. Edslev, S.M.; Olesen, C.M.; Nørreslet, L.B.; Ingham, A.C.; Iversen, S.; Lilje, B.; Clausen, M.-L.; Jensen, J.S.; Stegger, M.; Agner, T.; et al. Staphylococcal Communities on Skin Are Associated with Atopic Dermatitis and Disease Severity. Microorganisms 2021, 9, 432. [Google Scholar] [CrossRef]
  135. Iwamoto, K.; Moriwaki, M.; Miyake, R.; Hide, M. Staphylococcus aureus in Atopic Dermatitis: Strain-specific Cell Wall Proteins and Skin Immunity. Allergol. Int. 2019, 68, 309–315. [Google Scholar] [CrossRef]
  136. Hülpüsch, C.; Rohayem, R.; Reiger, M.; Traidl-Hoffmann, C. Exploring the Skin Microbiome in Atopic Dermatitis Pathogenesis and Disease Modification. J. Allergy Clin. Immunol. 2024, 154, 31–41. [Google Scholar] [CrossRef]
  137. Geoghegan, J.A.; Irvine, A.D.; Foster, T.J. Staphylococcus aureus and Atopic Dermatitis: A Complex and Evolving Relationship. Trends Microbiol. 2018, 26, 484–497. [Google Scholar] [CrossRef]
  138. Kong, H.H.; Oh, J.; Deming, C.; Conlan, S.; Grice, E.A.; Beatson, M.A.; Nomicos, E.; Polley, E.C.; Komarow, H.D.; Murray, P.R.; et al. Temporal Shifts in the Skin Microbiome Associated with Disease Flares and Treatment in Children with Atopic Dermatitis. Genome Res. 2012, 22, 850–859. [Google Scholar] [CrossRef]
  139. Shi, B.; Leung, D.Y.M.; Taylor, P.A.; Li, H. Methicillin-Resistant Staphylococcus aureus Colonization Is Associated with Decreased Skin Commensal Bacteria in Atopic Dermatitis. J. Investig. Dermatol. 2018, 138, 1668–1671. [Google Scholar] [CrossRef]
  140. Yu, J.; Luo, Y.; Zhu, Z.; Zhou, Y.; Sun, L.; Gao, J.; Sun, J.; Wang, G.; Yao, X.; Li, W. A Tryptophan Metabolite of the Skin Microbiota Attenuates Inflammation in Patients with Atopic Dermatitis through the Aryl Hydrocarbon Receptor. J. Allergy Clin. Immunol. 2019, 143, 2108–2119.e12. [Google Scholar] [CrossRef]
  141. Cho, S.-H.; Strickland, I.; Tomkinson, A.; Fehringer, A.P.; Gelfand, E.W.; Leung, D.Y.M. Preferential Binding of Staphylococcus aureus to Skin Sites of Th2-Mediated Inflammation in a Murine Model. J. Investig. Dermatol. 2001, 116, 658–663. [Google Scholar] [CrossRef]
  142. Albanesi, C.; Fairchild, H.R.; Madonna, S.; Scarponi, C.; De Pità, O.; Leung, D.Y.M.; Howell, M.D. IL-4 and IL-13 Negatively Regulate TNF-α- and IFN-γ-Induced β-Defensin Expression through STAT-6, Suppressor of Cytokine Signaling (SOCS)-1, and SOCS-3. J. Immunol. 2007, 179, 984–992. [Google Scholar] [CrossRef] [PubMed]
  143. Howell, M.D.; Fairchild, H.R.; Kim, B.E.; Bin, L.; Boguniewicz, M.; Redzic, J.S.; Hansen, K.C.; Leung, D.Y.M. Th2 Cytokines Act on S100/A11 to Downregulate Keratinocyte Differentiation. J. Investig. Dermatol. 2008, 128, 2248–2258. [Google Scholar] [CrossRef] [PubMed]
  144. Howell, M.D.; Boguniewicz, M.; Pastore, S.; Novak, N.; Bieber, T.; Girolomoni, G.; Leung, D.Y.M. Mechanism of HBD-3 Deficiency in Atopic Dermatitis. Clin. Immunol. 2006, 121, 332–338. [Google Scholar] [CrossRef] [PubMed]
  145. Howell, M.D.; Kim, B.E.; Gao, P.; Grant, A.V.; Boguniewicz, M.; DeBenedetto, A.; Schneider, L.; Beck, L.A.; Barnes, K.C.; Leung, D.Y.M. Cytokine Modulation of Atopic Dermatitis Filaggrin Skin Expression. J. Allergy Clin. Immunol. 2009, 124, R7–R12. [Google Scholar] [CrossRef]
  146. Fang, Z.; Li, L.; Zhang, H.; Zhao, J.; Lu, W.; Chen, W. Gut Microbiota, Probiotics, and Their Interactions in Prevention and Treatment of Atopic Dermatitis: A Review. Front. Immunol. 2021, 12, 720393. [Google Scholar] [CrossRef]
  147. Sadowsky, R.L.; Sulejmani, P.; Lio, P.A. Atopic Dermatitis: Beyond the Skin and Into the Gut. J. Clin. Med. 2023, 12, 5534. [Google Scholar] [CrossRef]
  148. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef]
  149. Kawai, T.; Ikegawa, M.; Ori, D.; Akira, S. Decoding Toll-like Receptors: Recent Insights and Perspectives in Innate Immunity. Immunity 2024, 57, 649–673. [Google Scholar] [CrossRef]
  150. Barcenilla, A.; Pryde, S.E.; Martin, J.C.; Duncan, S.H.; Stewart, C.S.; Henderson, C.; Flint, H.J. Phylogenetic Relationships of Butyrate-Producing Bacteria from the Human Gut. Appl. Environ. Microbiol. 2000, 66, 1654–1661. [Google Scholar] [CrossRef]
  151. Duncan, S.H.; Hold, G.L.; Harmsen, H.J.M.; Stewart, C.S.; Flint, H.J. Growth Requirements and Fermentation Products of Fusobacterium prausnitzii, and a Proposal to Reclassify It as Faecalibacterium prausnitzii Gen. Nov., Comb. Nov. Int. J. Syst. Evol. Microbiol. 2002, 52, 2141–2146. [Google Scholar] [CrossRef]
  152. Louis, P.; Hold, G.L.; Flint, H.J. The Gut Microbiota, Bacterial Metabolites and Colorectal Cancer. Nat. Rev. Microbiol. 2014, 12, 661–672. [Google Scholar] [CrossRef] [PubMed]
  153. Song, H.; Yoo, Y.; Hwang, J.; Na, Y.-C.; Kim, H.S. Faecalibacterium prausnitzii Subspecies–Level Dysbiosis in the Human Gut Microbiome Underlying Atopic Dermatitis. J. Allergy Clin. Immunol. 2016, 137, 852–860. [Google Scholar] [CrossRef] [PubMed]
  154. Mao, R.; Yu, Q.; Li, J. The Causal Relationship between Gut Microbiota and Inflammatory Dermatoses: A Mendelian Randomization Study. Front. Immunol. 2023, 14, 1231848. [Google Scholar] [CrossRef]
  155. Zhong, Y.; Wang, F.; Meng, X.; Zhou, L. The Associations between Gut Microbiota and Inflammatory Skin Diseases: A Bi-Directional Two-Sample Mendelian Randomization Study. Front. Immunol. 2024, 15, 1297240. [Google Scholar] [CrossRef]
  156. Stec, A.; Sikora, M.; Maciejewska, M.; Paralusz-Stec, K.; Michalska, M.; Sikorska, E.; Rudnicka, L. Bacterial Metabolites: A Link between Gut Microbiota and Dermatological Diseases. Int. J. Mol. Sci. 2023, 24, 3494. [Google Scholar] [CrossRef]
  157. Roduit, C.; Frei, R.; Ferstl, R.; Loeliger, S.; Westermann, P.; Rhyner, C.; Schiavi, E.; Barcik, W.; Rodriguez-Perez, N.; Wawrzyniak, M.; et al. High Levels of Butyrate and Propionate in Early Life Are Associated with Protection against Atopy. Allergy 2019, 74, 799–809. [Google Scholar] [CrossRef]
  158. Hu, C.; Zeng, D.; Huang, Y.; Deng, Q.; Liu, S.; Zhou, W.; Zhou, W. Sodium Butyrate Ameliorates Atopic Dermatitis-Induced Inflammation by Inhibiting HDAC3-Mediated STAT1 and NF-ΚB Pathway. Inflammation 2024, 47, 989–1001. [Google Scholar] [CrossRef]
  159. Leon Carrion, S.; Sutter, C.H.; Sutter, T.R. Combined Treatment with Sodium Butyrate and PD 153035 Enhances Keratinocyte Differentiation. Exp. Dermatol. 2014, 23, 211–214. [Google Scholar] [CrossRef]
  160. Traisaeng, S.; Herr, D.R.; Kao, H.-J.; Chuang, T.-H.; Huang, C.-M. A Derivative of Butyric Acid, the Fermentation Metabolite of Staphylococcus epidermidis, Inhibits the Growth of a Staphylococcus aureus Strain Isolated from Atopic Dermatitis Patients. Toxins 2019, 11, 311. [Google Scholar] [CrossRef]
  161. Park, D.H.; Kim, J.W.; Park, H.-J.; Hahm, D.-H. Comparative Analysis of the Microbiome across the Gut–Skin Axis in Atopic Dermatitis. Int. J. Mol. Sci. 2021, 22, 4228. [Google Scholar] [CrossRef]
  162. Qiu, Z.; Zhu, Z.; Liu, X.; Chen, B.; Yin, H.; Gu, C.; Fang, X.; Zhu, R.; Yu, T.; Mi, W.; et al. A Dysregulated Sebum–Microbial Metabolite–IL-33 Axis Initiates Skin Inflammation in Atopic Dermatitis. J. Exp. Med. 2022, 219, e20212397. [Google Scholar] [CrossRef] [PubMed]
  163. Chiang, N.; Serhan, C.N. Specialized Pro-Resolving Mediator Network: An Update on Production and Actions. Essays Biochem. 2020, 64, 443–462. [Google Scholar] [CrossRef] [PubMed]
  164. Headland, S.E.; Norling, L.V. The Resolution of Inflammation: Principles and Challenges. Semin. Immunol. 2015, 27, 149–160. [Google Scholar] [CrossRef]
  165. Basil, M.C.; Levy, B.D. Specialized Pro-Resolving Mediators: Endogenous Regulators of Infection and Inflammation. Nat. Rev. Immunol. 2016, 16, 51–67. [Google Scholar] [CrossRef]
  166. Fullerton, J.N.; Gilroy, D.W. Resolution of Inflammation: A New Therapeutic Frontier. Nat. Rev. Drug Discov. 2016, 15, 551–567. [Google Scholar] [CrossRef]
  167. Schett, G.; Neurath, M.F. Resolution of Chronic Inflammatory Disease: Universal and Tissue-Specific Concepts. Nat. Commun. 2018, 9, 3261. [Google Scholar] [CrossRef]
  168. Panigrahy, D.; Gilligan, M.M.; Serhan, C.N.; Kashfi, K. Resolution of Inflammation: An Organizing Principle in Biology and Medicine. Pharmacol. Ther. 2021, 227, 107879. [Google Scholar] [CrossRef]
  169. Serhan, C.N.; Gupta, S.K.; Perretti, M.; Godson, C.; Brennan, E.; Li, Y.; Soehnlein, O.; Shimizu, T.; Werz, O.; Chiurchiù, V.; et al. The Atlas of Inflammation Resolution (AIR). Mol. Asp. Med. 2020, 74, 100894. [Google Scholar] [CrossRef]
  170. Chiang, N.; Serhan, C.N. Structural Elucidation and Physiologic Functions of Specialized Pro-Resolving Mediators and Their Receptors. Mol. Asp. Med. 2017, 58, 114–129. [Google Scholar] [CrossRef]
  171. Filep, J.G.; Sekheri, M.; El Kebir, D. Targeting Formyl Peptide Receptors to Facilitate the Resolution of Inflammation. Eur. J. Pharmacol. 2018, 833, 339–348. [Google Scholar] [CrossRef]
  172. Krepel, S.A.; Wang, J.M. Chemotactic Ligands That Activate G-Protein-Coupled Formylpeptide Receptors. Int. J. Mol. Sci. 2019, 20, 3426. [Google Scholar] [CrossRef] [PubMed]
  173. Park, J.; Langmead, C.J.; Riddy, D.M. New Advances in Targeting the Resolution of Inflammation: Implications for Specialized Pro-Resolving Mediator GPCR Drug Discovery. ACS Pharmacol. Transl. Sci. 2020, 3, 88–106. [Google Scholar] [CrossRef] [PubMed]
  174. Mastromarino, M.; Lacivita, E.; Colabufo, N.A.; Leopoldo, M. G-Protein Coupled Receptors Involved in the Resolution of Inflammation: Ligands and Therapeutic Perspectives. Mini-Rev. Med. Chem. 2021, 20, 2090–2103. [Google Scholar] [CrossRef]
  175. Tylek, K.; Trojan, E.; Regulska, M.; Lacivita, E.; Leopoldo, M.; Basta-Kaim, A. Formyl Peptide Receptor 2, as an Important Target for Ligands Triggering the Inflammatory Response Regulation: A Link to Brain Pathology. Pharmacol. Rep. 2021, 73, 1004–1019. [Google Scholar] [CrossRef]
  176. Livshits, G.; Kalinkovich, A. Receptors for Pro-Resolving Mediators as a Therapeutic Tool for Smooth Muscle Remodeling-Associated Disorders. Pharmacol. Res. 2021, 164, 105340. [Google Scholar] [CrossRef]
  177. Livshits, G.; Kalinkovich, A. Specialized, Pro-Resolving Mediators as Potential Therapeutic Agents for Alleviating Fibromyalgia Symptomatology. Pain Med. 2022, 23, 977–990. [Google Scholar] [CrossRef]
  178. Kalinkovich, A.; Becker, M.; Livshits, G. New Horizons in the Treatment of Age-Associated Obesity, Sarcopenia and Osteoporosis. Drugs Aging 2022, 39, 673–683. [Google Scholar] [CrossRef]
  179. Gusakov, K.; Kalinkovich, A.; Ashkenazi, S.; Livshits, G. Nature of the Association between Rheumatoid Arthritis and Cervical Cancer and Its Potential Therapeutic Implications. Nutrients 2024, 16, 2569. [Google Scholar] [CrossRef]
  180. Töröcsik, D.; Weise, C.; Gericke, J.; Szegedi, A.; Lucas, R.; Mihaly, J.; Worm, M.; Rühl, R. Transcriptomic and Lipidomic Profiling of Eicosanoid/Docosanoid Signalling in Affected and Non-affected Skin of Human Atopic Dermatitis Patients. Exp. Dermatol. 2019, 28, 177–189. [Google Scholar] [CrossRef]
  181. Huang, X.W.; Pang, S.W.; Yang, L.Z.; Han, T.; Chen, J.; Huang, C.W.; Liao, L.; Xie, P.J. TNFSF14 Mediates the Impact of Docosahexaenoic Acid on Atopic Dermatitis: A Mendelian Randomization Study. Eur. Rev. Med. Pharmacol. Sci. 2024, 28, 107–117. [Google Scholar]
  182. Bjorneboe, A.; Soyland, E.; Bjorneboe, G.-E.A.; Rajka, G.; Drevon, C.A. Effect of Dietary Supplementation with Eicosapentaenoic Acid in the Treatment of Atopic Dermatitis. Br. J. Dermatol. 1987, 117, 463–469. [Google Scholar] [CrossRef] [PubMed]
  183. Oien, T.; Storro, O.; Johnsen, R. Do Early Intake of Fish and Fish Oil Protect against Eczema and Doctor-Diagnosed Asthma at 2 Years of Age? A Cohort Study. J. Epidemiol. Community Health 2010, 64, 124–129. [Google Scholar] [CrossRef] [PubMed]
  184. Øien, T.; Schjelvaag, A.; Storrø, O.; Johnsen, R.; Simpson, M. Fish Consumption at One Year of Age Reduces the Risk of Eczema, Asthma and Wheeze at Six Years of Age. Nutrients 2019, 11, 1969. [Google Scholar] [CrossRef] [PubMed]
  185. Zhang, B.; Li, P.; Fu, P. Association between Polyunsaturated Fatty Acid Intake and Eczema in Children and Adolescents. Int. Arch. Allergy Immunol. 2023, 184, 681–691. [Google Scholar] [CrossRef] [PubMed]
  186. Best, K.P.; Gold, M.; Kennedy, D.; Martin, J.; Makrides, M. Omega-3 Long-Chain PUFA Intake during Pregnancy and Allergic Disease Outcomes in the Offspring: A Systematic Review and Meta-Analysis of Observational Studies and Randomized Controlled Trials. Am. J. Clin. Nutr. 2016, 103, 128–143. [Google Scholar] [CrossRef]
  187. Miles, E.; Calder, P. Can Early Omega-3 Fatty Acid Exposure Reduce Risk of Childhood Allergic Disease? Nutrients 2017, 9, 784. [Google Scholar] [CrossRef]
  188. Zhang, Y.; Lin, J.; Zhou, R.; Zheng, X.; Dai, J. Effect of Omega-3 Fatty Acids Supplementation during Childhood in Preventing Allergic Disease: A Systematic Review and Meta-Analysis. J. Asthma 2021, 58, 523–536. [Google Scholar] [CrossRef]
  189. Jia, Y.; Huang, Y.; Wang, H.; Jiang, H. Effect of Prenatal Omega-3 Polyunsaturated Fatty Acid Supplementation on Childhood Eczema: A Systematic Review and Meta-Analysis. Int. Arch. Allergy Immunol. 2023, 184, 21–32. [Google Scholar] [CrossRef]
  190. Bertino, L.; Guarneri, F.; Cannavò, S.P.; Casciaro, M.; Pioggia, G.; Gangemi, S. Oxidative Stress and Atopic Dermatitis. Antioxidants 2020, 9, 196. [Google Scholar] [CrossRef]
  191. Crupi, R.; Marino, A.; Cuzzocrea, S. n-3 Fatty Acids: Role in Neurogenesis and Neuroplasticity. Curr. Med. Chem. 2013, 24, 2953–2963. [Google Scholar] [CrossRef]
  192. Panda, C.; Varadharaj, S.; Voruganti, V.S. PUFA, Genotypes and Risk for Cardiovascular Disease. Prostaglandins Leukot. Essent. Fat. Acids 2022, 176, 102377. [Google Scholar] [CrossRef] [PubMed]
  193. Li, Y.; Zhao, T.; Li, J.; Xia, M.; Li, Y.; Wang, X.; Liu, C.; Zheng, T.; Chen, R.; Kan, D.; et al. Oxidative Stress and 4-hydroxy-2-nonenal (4-HNE): Implications in the Pathogenesis and Treatment of Aging-related Diseases. J. Immunol. Res. 2022, 2022, 2233906. [Google Scholar] [CrossRef] [PubMed]
  194. Cordiano, R.; Di Gioacchino, M.; Mangifesta, R.; Panzera, C.; Gangemi, S.; Minciullo, P.L. Malondialdehyde as a Potential Oxidative Stress Marker for Allergy-Oriented Diseases: An Update. Molecules 2023, 28, 5979. [Google Scholar] [CrossRef]
  195. Zaloga, G.P. Narrative Review of n-3 Polyunsaturated Fatty Acid Supplementation upon Immune Functions, Resolution Molecules and Lipid Peroxidation. Nutrients 2021, 13, 662. [Google Scholar] [CrossRef] [PubMed]
  196. Leong, X.F. Lipid Oxidation Products on Inflammation-Mediated Hypertension and Atherosclerosis: A Mini Review. Front. Nutr. 2021, 8, 717740. [Google Scholar] [CrossRef]
  197. Atalay Ekiner, S.; Gęgotek, A.; Skrzydlewska, E. Inflammasome Activity Regulation by PUFA Metabolites. Front. Immunol. 2024, 15, 1452749. [Google Scholar] [CrossRef]
  198. Sawada, Y.; Honda, T.; Nakamizo, S.; Otsuka, A.; Ogawa, N.; Kobayashi, Y.; Nakamura, M.; Kabashima, K. Resolvin E1 Attenuates Murine Psoriatic Dermatitis. Sci. Rep. 2018, 8, 11873. [Google Scholar] [CrossRef]
  199. Haworth, O.; Cernadas, M.; Yang, R.; Serhan, C.N.; Levy, B.D. Resolvin E1 Regulates Interleukin 23, Interferon-γ and Lipoxin A4 to Promote the Resolution of Allergic Airway Inflammation. Nat. Immunol. 2008, 9, 873–879. [Google Scholar] [CrossRef]
  200. Aoki, H.; Hisada, T.; Ishizuka, T.; Utsugi, M.; Kawata, T.; Shimizu, Y.; Okajima, F.; Dobashi, K.; Mori, M. Resolvin E1 Dampens Airway Inflammation and Hyperresponsiveness in a Murine Model of Asthma. Biochem. Biophys. Res. Commun. 2008, 367, 509–515. [Google Scholar] [CrossRef]
  201. Aoki, H.; Hisada, T.; Ishizuka, T.; Utsugi, M.; Ono, A.; Koga, Y.; Sunaga, N.; Nakakura, T.; Okajima, F.; Dobashi, K.; et al. Protective Effect of Resolvin E1 on the Development of Asthmatic Airway Inflammation. Biochem. Biophys. Res. Commun. 2010, 400, 128–133. [Google Scholar] [CrossRef]
  202. Xu, J.; Duan, X.; Hu, F.; Poorun, D.; Liu, X.; Wang, X.; Zhang, S.; Gan, L.; He, M.; Zhu, K.; et al. Resolvin D1 Attenuates Imiquimod-Induced Mice Psoriasiform Dermatitis through MAPKs and NF-ΚB Pathways. J. Dermatol. Sci. 2018, 89, 127–135. [Google Scholar] [CrossRef] [PubMed]
  203. Park, K.-D.; Kim, N.; Kang, J.; Dhakal, H.; Kim, J.Y.; Jang, Y.H.; Lee, W.J.; Lee, S.-J.; Kim, S.-H. Protectin D1 Reduces Imiquimod-Induced Psoriasiform Skin Inflammation. Int. Immunopharmacol. 2021, 98, 107883. [Google Scholar] [CrossRef] [PubMed]
  204. Kanda, N.; Watanabe, S. Leukotriene B 4 Enhances Tumour Necrosis Factor-α-induced CCL27 Production in Human Keratinocytes. Clin. Exp. Allergy 2007, 37, 1074–1082. [Google Scholar] [CrossRef] [PubMed]
  205. Wang, Y.; Huo, J.; Zhang, D.; Hu, G.; Zhang, Y. Chemerin/ChemR23 Axis Triggers an Inflammatory Response in Keratinocytes through ROS-sirt1-NF-κB Signaling. J. Cell. Biochem. 2019, 120, 6459–6470. [Google Scholar] [CrossRef]
  206. Hellmann, J.; Sansbury, B.E.; Wong, B.; Li, X.; Singh, M.; Nuutila, K.; Chiang, N.; Eriksson, E.; Serhan, C.N.; Spite, M. Biosynthesis of D-Series Resolvins in Skin Provides Insights into Their Role in Tissue Repair. J. Investig. Dermatol. 2018, 138, 2051–2060. [Google Scholar] [CrossRef]
  207. Lebtig, M.; Scheurer, J.; Muenkel, M.; Becker, J.; Bastounis, E.; Peschel, A.; Kretschmer, D. Keratinocytes Use FPR2 to Detect Staphylococcus aureus and Initiate Antimicrobial Skin Defense. Front. Immunol. 2023, 14, 1188555. [Google Scholar] [CrossRef]
  208. Lee, S.H.; Tonello, R.; Im, S.-T.; Jeon, H.; Park, J.; Ford, Z.; Davidson, S.; Kim, Y.H.; Park, C.-K.; Berta, T. Resolvin D3 Controls Mouse and Human TRPV1-Positive Neurons and Preclinical Progression of Psoriasis. Theranostics 2020, 10, 12111–12126. [Google Scholar] [CrossRef]
  209. Mihály, J.; Gericke, J.; Aydemir, G.; Weiss, K.; Carlsen, H.; Blomhoff, R.; Garcia, J.; Rühl, R. Reduced Retinoid Signaling in the Skin after Systemic Retinoid-X Receptor Ligand Treatment in Mice with Potential Relevance for Skin Disorders. Dermatology 2012, 225, 304–311. [Google Scholar] [CrossRef]
  210. LaMotte, R.H.; Dong, X.; Ringkamp, M. Sensory Neurons and Circuits Mediating Itch. Nat. Rev. Neurosci. 2014, 15, 19–31. [Google Scholar] [CrossRef]
  211. Gouin, O.; L’Herondelle, K.; Lebonvallet, N.; Le Gall-Ianotto, C.; Sakka, M.; Buhé, V.; Plée-Gautier, E.; Carré, J.-L.; Lefeuvre, L.; Misery, L.; et al. TRPV1 and TRPA1 in Cutaneous Neurogenic and Chronic Inflammation: Pro-Inflammatory Response Induced by Their Activation and Their Sensitization. Protein Cell 2017, 8, 644–661. [Google Scholar] [CrossRef]
  212. Moore, C.; Gupta, R.; Jordt, S.-E.; Chen, Y.; Liedtke, W.B. Regulation of Pain and Itch by TRP Channels. Neurosci. Bull. 2018, 34, 120–142. [Google Scholar] [CrossRef] [PubMed]
  213. Butler, D.C.; Berger, T.; Elmariah, S.; Kim, B.; Chisolm, S.; Kwatra, S.G.; Mollanazar, N.; Yosipovitch, G. Chronic Pruritus. JAMA 2024, 331, 2114. [Google Scholar] [CrossRef] [PubMed]
  214. Wu, S.-H.; Chen, X.-Q.; Liu, B.; Wu, H.-J.; Dong, L. Efficacy and Safety of 15(R/S)-Methyl-Lipoxin A 4 in Topical Treatment of Infantile Eczema. Br. J. Dermatol. 2013, 168, 172–178. [Google Scholar] [CrossRef] [PubMed]
  215. Liu, X.; Wang, X.; Duan, X.; Poorun, D.; Xu, J.; Zhang, S.; Gan, L.; He, M.; Zhu, K.; Ming, Z.; et al. Lipoxin A4 and Its Analog Suppress Inflammation by Modulating HMGB1 Translocation and Expression in Psoriasis. Sci. Rep. 2017, 7, 7100. [Google Scholar] [CrossRef]
  216. Ji, R.-R.; Donnelly, C.R.; Nedergaard, M. Astrocytes in Chronic Pain and Itch. Nat. Rev. Neurosci. 2019, 20, 667–685. [Google Scholar] [CrossRef]
  217. Nesman, J.I.; Chen, O.; Luo, X.; Ji, R.-R.; Serhan, C.N.; Hansen, T.V. A New Synthetic Protectin D1 Analog 3-Oxa-PD1 n-3 DPA Reduces Neuropathic Pain and Chronic Itch in Mice. Org. Biomol. Chem. 2021, 19, 2744–2752. [Google Scholar] [CrossRef]
  218. Tiberi, M.; Chiurchiù, V. Specialized Pro-Resolving Lipid Mediators and Glial Cells: Emerging Candidates for Brain Homeostasis and Repair. Front. Cell. Neurosci. 2021, 15, 673549. [Google Scholar] [CrossRef]
  219. Ji, R.-R. Specialized Pro-Resolving Mediators as Resolution Pharmacology for the Control of Pain and Itch. Annu. Rev. Pharmacol. Toxicol. 2023, 63, 273–293. [Google Scholar] [CrossRef]
  220. Ji, R.-R. Neuroimmune Interactions in Itch: Do Chronic Itch, Chronic Pain, and Chronic Cough Share Similar Mechanisms? Pulm. Pharmacol. Ther. 2015, 35, 81–86. [Google Scholar] [CrossRef]
  221. Yosipovitch, G.; Berger, T.; Fassett, M.S. Neuroimmune Interactions in Chronic Itch of Atopic Dermatitis. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 239–250. [Google Scholar] [CrossRef]
  222. Steinhoff, M.; Ahmad, F.; Pandey, A.; Datsi, A.; AlHammadi, A.; Al-Khawaga, S.; Al-Malki, A.; Meng, J.; Alam, M.; Buddenkotte, J. Neuroimmune Communication Regulating Pruritus in Atopic Dermatitis. J. Allergy Clin. Immunol. 2022, 149, 1875–1898. [Google Scholar] [CrossRef]
  223. Escalante, A.; Serra-Baldrich, E. Pathogenic Mechanisms Underlying Itch in Atopic Dermatitis: The Emerging Role of Neuroimmune Interactions. Eur. J. Dermatol. 2023, 33, 343–349. [Google Scholar] [CrossRef] [PubMed]
  224. Xu, Z.-Z.; Zhang, L.; Liu, T.; Park, J.Y.; Berta, T.; Yang, R.; Serhan, C.N.; Ji, R.-R. Resolvins RvE1 and RvD1 Attenuate Inflammatory Pain via Central and Peripheral Actions. Nat. Med. 2010, 16, 592–597. [Google Scholar] [CrossRef] [PubMed]
  225. Kim, Y.J.; Granstein, R.D. Roles of Calcitonin Gene-Related Peptide in the Skin, and Other Physiological and Pathophysiological Functions. Brain Behav. Immun. Health 2021, 18, 100361. [Google Scholar] [CrossRef] [PubMed]
  226. Abdelhadi, S.; Nordlind, K.; Johansson, B.; Theodorsson, E.; Holst, M.; Lönndahl, L. Expression of Calcitonin Gene-Related Peptide in Atopic Dermatitis and Correlation with Distress. Immunopharmacol. Immunotoxicol. 2024, 46, 67–72. [Google Scholar] [CrossRef]
  227. Na, C.; Baghoomian, W.; Simpson, E. A Therapeutic Renaissance-Emerging Treatments for Atopic Dermatitis. Acta Derm. Venereol. 2020, 100, adv00165. [Google Scholar] [CrossRef] [PubMed]
  228. Lovell, K.; Patel, N.; Rao, S.; Strowd, L.C. The Future of Atopic Dermatitis Treatment. Adv. Exp. Med. Biol. 2024, 1447, 227–244. [Google Scholar] [CrossRef] [PubMed]
  229. Radi, G.; Campanti, A.; Diotallevi, F.; Martina, E.; Marani, A.; Offidani, A. A Systematic Review of Atopic Dermatitis: The Intriguing Journey Starting from Physiopathology to Treatment, from Laboratory Bench to Bedside. Biomedicines 2022, 10, 2700. [Google Scholar] [CrossRef]
  230. Ferrara, F.; Zovi, A.; Capuozzo, M.; Langella, R. Atopic Dermatitis: Treatment and Innovations in Immunotherapy. Inflammopharmacology 2024, 32, 1777–1789. [Google Scholar] [CrossRef]
  231. Traidl, S.; Freimooser, S.; Werfel, T. Janus Kinase Inhibitors for the Therapy of Atopic Dermatitis. Allergol. Sel. 2021, 5, 293–304. [Google Scholar] [CrossRef]
  232. Nakashima, C.; Yanagihara, S.; Otsuka, A. Innovation in the Treatment of Atopic Dermatitis: Emerging Topical and Oral Janus Kinase Inhibitors. Allergol. Int. 2022, 71, 40–46. [Google Scholar] [CrossRef] [PubMed]
  233. Chovatiya, R.; Paller, A.S. JAK Inhibitors in the Treatment of Atopic Dermatitis. J. Allergy Clin. Immunol. 2021, 148, 927–940. [Google Scholar] [CrossRef] [PubMed]
  234. Müller, S.; Maintz, L.; Bieber, T. Treatment of Atopic Dermatitis: Recently Approved Drugs and Advanced Clinical Development Programs. Allergy 2024, 79, 1501–1515. [Google Scholar] [CrossRef] [PubMed]
  235. Dubin, C.; Del Duca, E.; Guttman-Yassky, E. The IL-4, IL-13 and IL-31 Pathways in Atopic Dermatitis. Expert Rev. Clin. Immunol. 2021, 17, 835–852. [Google Scholar] [CrossRef] [PubMed]
  236. Silverberg, J.I.; Toth, D.; Bieber, T.; Alexis, A.F.; Elewski, B.E.; Pink, A.E.; Hijnen, D.; Jensen, T.N.; Bang, B.; Olsen, C.K.; et al. Tralokinumab plus Topical Corticosteroids for the Treatment of Moderate-to-severe Atopic Dermatitis: Results from the Double-blind, Randomized, Multicentre, Placebo-controlled Phase III ECZTRA 3 Trial*. Br. J. Dermatol. 2021, 184, 450–463. [Google Scholar] [CrossRef]
  237. Wan, H.; Jia, H.; Xia, T.; Zhang, D. Comparative Efficacy and Safety of Abrocitinib, Baricitinib, and Upadacitinib for Moderate-to-Severe Atopic Dermatitis: A Network Meta-analysis. Dermatol. Ther. 2022, 35, e15636. [Google Scholar] [CrossRef]
  238. Eberlein, B.; Eicke, C.; Reinhardt, H.; Ring, J. Adjuvant Treatment of Atopic Eczema: Assessment of an Emollient Containing N-palmitoylethanolamine (ATOPA Study). J. Eur. Acad. Dermatol. Venereol. 2008, 22, 73–82. [Google Scholar] [CrossRef]
  239. Umehara, T. Epidrugs: Toward Understanding and Treating Diverse Diseases. Epigenomes 2022, 6, 18. [Google Scholar] [CrossRef]
  240. Feehley, T.; O’Donnell, C.W.; Mendlein, J.; Karande, M.; McCauley, T. Drugging the Epigenome in the Age of Precision Medicine. Clin. Epigenet. 2023, 15, 6. [Google Scholar] [CrossRef]
  241. Miranda Furtado, C.L.; Dos Santos Luciano, M.C.; Silva Santos, R.D.; Furtado, G.P.; Moraes, M.O.; Pessoa, C. Epidrugs: Targeting Epigenetic Marks in Cancer Treatment. Epigenetics 2019, 14, 1164–1176. [Google Scholar] [CrossRef]
  242. Grandi, F.C.; Bhutani, N. Epigenetic Therapies for Osteoarthritis. Trends Pharmacol. Sci. 2020, 41, 557–569. [Google Scholar] [CrossRef] [PubMed]
  243. Liotti, A.; Ferrara, A.L.; Loffredo, S.; Galdiero, M.R.; Varricchi, G.; Di Rella, F.; Maniscalco, G.T.; Belardo, M.; Vastano, R.; Prencipe, R.; et al. Epigenetics: An Opportunity to Shape Innate and Adaptive Immune Responses. Immunology 2022, 167, 451–470. [Google Scholar] [CrossRef] [PubMed]
  244. Gladkova, M.G.; Leidmaa, E.; Anderzhanova, E.A. Epidrugs in the Therapy of Central Nervous System Disorders: A Way to Drive on? Cells 2023, 12, 1464. [Google Scholar] [CrossRef] [PubMed]
  245. Ueda, J.; Yamazaki, T.; Funakoshi, H. Toward the Development of Epigenome Editing-Based Therapeutics: Potentials and Challenges. Int. J. Mol. Sci. 2023, 24, 4778. [Google Scholar] [CrossRef] [PubMed]
  246. Rittiner, J.; Cumaran, M.; Malhotra, S.; Kantor, B. Therapeutic Modulation of Gene Expression in the Disease State: Treatment Strategies and Approaches for the Development of Next-generation of the Epigenetic Frugs. Front. Bioeng. Biotechnol. 2022, 10, 1035543. [Google Scholar] [CrossRef]
  247. Lataniotis, L.; Albrecht, A.; Kok, F.O.; Monfries, C.A.L.; Benedetti, L.; Zampetaki, A. CRISPR/Cas9 Editing Reveals Novel Mechanisms of Clustered microRNA Regulation and Function. Sci. Rep. 2017, 7, 8585. [Google Scholar] [CrossRef] [PubMed]
  248. Kurata, J.S.; Lin, R.J. MicroRNA-focused CRISPR-Cas9 Library Screen Reveals Fitness-associated miRNAs. RNA 2018, 24, 966–981. [Google Scholar] [CrossRef]
  249. Alinejad, T.; Modarressi, S.; Sadri, Z.; Hao, Z.; Chen, C.S. Diagnostic Applications and Therapeutic Option of Cascade CRISPR/Cas in the Modulation of MiRNA in Diverse Cancers: Promises and Obstacles. J. Cancer Res. Clin. Oncol. 2023, 149, 9557–9575. [Google Scholar] [CrossRef]
  250. Jing, W.; Zhang, X.; Sun, W.; Hou, X.; Yao, Z.; Zhu, Y. CRISPR/CAS9-Mediated Genome Editing of MiRNA-155 Inhibits Proinflammatory Cytokine Production by RAW264.7 Cells. Biomed Res. Int. 2015, 2015, 326042. [Google Scholar] [CrossRef]
  251. Kwon, Y.; Choi, Y.; Kim, M.; Jeong, M.S.; Jung, H.S.; Jeoung, D. HDAC6 and CXCL13 Mediate Atopic Dermatitis by Regulating Cellular Interactions and Expression Levels of MiR-9 and SIRT1. Front. Pharmacol. 2021, 12, 691279. [Google Scholar] [CrossRef]
  252. Kwon, Y.; Choi, Y.; Kim, M.; Jo, H.; Jeong, M.S.; Jung, H.S.; Jeoung, D. HDAC6-MYCN-CXCL3 Axis Mediates Allergic Inflammation and Is Necessary for Allergic Inflammation-Promoted Cellular Interactions. Mol. Immunol. 2024, 166, 1–15. [Google Scholar] [CrossRef] [PubMed]
  253. Wu, Y.-J.; Wu, W.-F.; Hung, C.-W.; Ku, M.-S.; Liao, P.-F.; Sun, H.-L.; Lu, K.-H.; Sheu, J.-N.; Lue, K.-H. Evaluation of Efficacy and Safety of Lactobacillus Rhamnosus in Children Aged 4–48 Months with Atopic Dermatitis: An 8-Week, Double-Blind, Randomized, Placebo-Controlled Study. J. Microbiol. Immunol. Infect. 2017, 50, 684–692. [Google Scholar] [CrossRef] [PubMed]
  254. Carucci, L.; Nocerino, R.; Paparo, L.; De Filippis, F.; Coppola, S.; Giglio, V.; Cozzolino, T.; Valentino, V.; Sequino, G.; Bedogni, G.; et al. Therapeutic Effects Elicited by the Probiotic Lacticaseibacillus rhamnosus GG in Children with Atopic Dermatitis. The Results of the ProPAD Trial. Pediatr. Allergy Immunol. 2022, 33, e13836. [Google Scholar] [CrossRef] [PubMed]
  255. Colombo, D.; Rigoni, C.; Cantù, A.; Carnevali, A.; Filippetti, R.; Franco, T.; Grassi, A.; Loi, C.; Mazzotta, A.; Patroi, I.; et al. Probiotics and Prebiotics Orally Assumed as Disease Modifiers for Stable Mild Atopic Dermatitis: An Italian Real-Life, Multicenter, Retrospective, Observational Study. Medicina 2023, 59, 2080. [Google Scholar] [CrossRef] [PubMed]
  256. Greenzaid, J.D.; Chan, L.J.; Chandani, B.M.; Kiritsis, N.R.; Feldman, S.R. Microbiome Modulators for Atopic Eczema: A Systematic Review of Experimental and Investigational Therapeutics. Expert Opin. Investig. Drugs 2024, 33, 415–430. [Google Scholar] [CrossRef] [PubMed]
  257. Chang, Y.-S.; Trivedi, M.K.; Jha, A.; Lin, Y.-F.; Dimaano, L.; García-Romero, M.T. Synbiotics for Prevention and Treatment of Atopic Dermatitis. JAMA Pediatr. 2016, 170, 236. [Google Scholar] [CrossRef]
  258. Fijan, S.; Kolč, N.; Hrašovec, M.; Jamtvedt, G.; Pogačar, M.Š.; Mičetić Turk, D.; Maver, U. Single-Strain Probiotic Lactobacilli for the Treatment of Atopic Dermatitis in Children: A Systematic Review and Meta-Analysis. Pharmaceutics 2023, 15, 1256. [Google Scholar] [CrossRef]
  259. Wang, F.; Wu, F.; Chen, H.; Tang, B. The Effect of Probiotics in the Prevention of Atopic Dermatitis in Children: A Systematic Review and Meta-Analysis. Transl. Pediatr. 2023, 12, 731–748. [Google Scholar] [CrossRef]
  260. Xue, X.; Yang, X.; Shi, X.; Deng, Z. Efficacy of Probiotics in Pediatric Atopic Dermatitis: A Systematic Review and Meta-analysis. Clin. Transl. Allergy 2023, 13, e12283. [Google Scholar] [CrossRef]
  261. Li, Y.; Zhang, B.; Guo, J.; Cao, Z.; Shen, M. The Efficacy of Probiotics Supplementation for the Treatment of Atopic Dermatitis in Adults: A Systematic Review and Meta-Analysis. J. Dermatol. Treat. 2022, 33, 2800–2809. [Google Scholar] [CrossRef]
  262. Umborowati, M.A.; Damayanti, D.; Anggraeni, S.; Endaryanto, A.; Surono, I.S.; Effendy, I.; Prakoeswa, C.R.S. The Role of Probiotics in the Treatment of Adult Atopic Dermatitis: A Meta-Analysis of Randomized Controlled Trials. J. Health Popul. Nutr. 2022, 41, 37. [Google Scholar] [CrossRef] [PubMed]
  263. Kim, K.; Lee, E.; Kim, M.; Hong, S.J. Therapeutic Effectiveness of Probiotics for Atopic Dermatitis: A Systematic Review and Meta-analysis of Randomized Controlled Trials with Subgroup Analysis. Asian Pac. J. Allergy Immunol. 2023. [Google Scholar] [CrossRef]
  264. Husein-ElAhmed, H.; Steinhoff, M. Effects of Probiotic Supplementation in Adult with Atopic Dermatitis: A Systematic Review with Meta-Analysis. Clin. Exp. Dermatol. 2023, 49, 46–52. [Google Scholar] [CrossRef] [PubMed]
  265. Huang, R.; Ning, H.; Shen, M.; Li, J.; Zhang, J.; Chen, X. Probiotics for the Treatment of Atopic Dermatitis in Children: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Cell. Infect. Microbiol. 2017, 7, 392. [Google Scholar] [CrossRef] [PubMed]
  266. Makrgeorgou, A.; Leonardi-Bee, J.; Bath-Hextall, F.J.; Murrell, D.F.; Tang, M.L.; Roberts, A.; Boyle, R.J. Probiotics for Treating Eczema. Cochrane Database Syst. Rev. 2018, 2018, CD006135. [Google Scholar] [CrossRef]
  267. McFarlane, C.; Ramos, C.I.; Johnson, D.W.; Campbell, K.L. Prebiotic, Probiotic, and Synbiotic Supplementation in Chronic Kidney Disease: A Systematic Review and Meta-Analysis. J. Ren. Nutr. 2019, 29, 209–220. [Google Scholar] [CrossRef]
  268. Bock, P.M.; Telo, G.H.; Ramalho, R.; Sbaraini, M.; Leivas, G.; Martins, A.F.; Schaan, B.D. The Effect of Probiotics, Prebiotics or Synbiotics on Metabolic Outcomes in Individuals with Diabetes: A Systematic Review and Meta-Analysis. Diabetologia 2021, 64, 26–41. [Google Scholar] [CrossRef]
  269. Jukic Peladic, N.; Dell’Aquila, G.; Carrieri, B.; Maggio, M.; Cherubini, A.; Orlandoni, P. Potential Role of Probiotics for Inflammaging: A Narrative Review. Nutrients 2021, 13, 2919. [Google Scholar] [CrossRef]
  270. Warman, D.J.; Jia, H.; Kato, H. The Potential Roles of Probiotics, Resistant Starch, and Resistant Proteins in Ameliorating Inflammation during Aging (Inflammaging). Nutrients 2022, 14, 747. [Google Scholar] [CrossRef]
  271. Padhi, P.; Worth, C.; Zenitsky, G.; Jin, H.; Sambamurti, K.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Mechanistic Insights Into Gut Microbiome Dysbiosis-Mediated Neuroimmune Dysregulation and Protein Misfolding and Clearance in the Pathogenesis of Chronic Neurodegenerative Disorders. Front. Neurosci. 2022, 16, 836605. [Google Scholar] [CrossRef]
  272. Ren, J.; Li, H.; Zeng, G.; Pang, B.; Wang, Q.; Wei, J. Gut Microbiome-Mediated Mechanisms in Aging-Related Diseases: Are Probiotics Ready for Prime Time? Front. Pharmacol. 2023, 14, 1178596. [Google Scholar] [CrossRef] [PubMed]
  273. Colletti, A.; Pellizzato, M.; Cicero, A.F. The Possible Role of Probiotic Supplementation in Inflammation: A Narrative Review. Microorganisms 2023, 11, 2160. [Google Scholar] [CrossRef] [PubMed]
  274. Serhan, C.N.; Chiang, N.; Dalli, J. The Resolution Code of Acute Inflammation: Novel pro-Resolving Lipid Mediators in Resolution. Semin. Immunol. 2015, 27, 200–215. [Google Scholar] [CrossRef]
  275. Serhan, C.N.; Sulciner, M.L. Resolution Medicine in Cancer, Infection, Pain and Inflammation: Are We on Track to Address the next Pandemic? Cancer Metastasis Rev. 2023, 42, 13–17. [Google Scholar] [CrossRef] [PubMed]
  276. Kim, N.; Thatcher, T.H.; Sime, P.J.; Phipps, R.P. Corticosteroids Inhibit Anti-IgE Activities of Specialized Proresolving Mediators on B Cells from Asthma Patients. JCI Insight 2017, 2, e88588. [Google Scholar] [CrossRef] [PubMed]
  277. Raghuraman, S.; Donkin, I.; Versteyhe, S.; Barrès, R.; Simar, D. The Emerging Role of Epigenetics in Inflammation and Immunometabolism. Trends Endocrinol. Metab. 2016, 27, 782–795. [Google Scholar] [CrossRef]
  278. Stylianou, E. Epigenetics of Chronic Inflammatory Diseases. J. Inflamm. Res. 2018, 12, 1–14. [Google Scholar] [CrossRef]
  279. Liu, X.; Wang, C.; Pang, L.; Pan, L.; Zhang, Q. Combination of Resolvin E1 and Lipoxin A4 Promotes the Resolution of Pulpitis by Inhibiting NF-κB Activation through Upregulating Sirtuin 7 in Dental Pulp Fibroblasts. Cell Prolif. 2022, 55, e13227. [Google Scholar] [CrossRef]
  280. Zhuo, Y.; Zhang, S.; Li, C.; Yang, L.; Gao, H.; Wang, X. Resolvin D1 Promotes SIRT1 Expression to Counteract the Activation of STAT3 and NF-ΚB in Mice with Septic-Associated Lung Injury. Inflammation 2018, 41, 1762–1771. [Google Scholar] [CrossRef]
  281. Xian, W.; Li, T.; Li, L.; Hu, L.; Cao, J. Maresin 1 Attenuates the Inflammatory Response and Mitochondrial Damage in Mice with Cerebral Ischemia/Reperfusion in a SIRT1-Dependent Manner. Brain Res. 2019, 1711, 83–90. [Google Scholar] [CrossRef]
  282. Wang, W.; Xu, R.-L.; He, P.; Chen, R. MAR1 Suppresses Inflammatory Response in LPS-Induced RAW 264.7 Macrophages and Human Primary Peripheral Blood Mononuclear Cells via the SIRT1/PGC-1α/PPAR-γ Pathway. J. Inflamm. 2021, 18, 8. [Google Scholar] [CrossRef] [PubMed]
  283. Sun, W.; Ma, J.; Zhao, H.; Xiao, C.; Zhong, H.; Ling, H.; Xie, Z.; Tian, Q.; Chen, H.; Zhang, T.; et al. Resolvin D1 Suppresses Pannus Formation via Decreasing Connective Tissue Growth Factor Caused by Upregulation of MiRNA-146a-5p in Rheumatoid Arthritis. Arthritis Res. Ther. 2020, 22, 61. [Google Scholar] [CrossRef] [PubMed]
  284. Cheng, T.; Ding, S.; Liu, S.; Li, X.; Tang, X.; Sun, L. Resolvin D1 Improves the Treg/Th17 Imbalance in Systemic Lupus Erythematosus Through MiR-30e-5p. Front. Immunol. 2021, 12, 668760. [Google Scholar] [CrossRef]
  285. Livshits, G.; Kalinkovich, A. Targeting Chronic Inflammation as a Potential Adjuvant Therapy for Osteoporosis. Life Sci. 2022, 306, 120847. [Google Scholar] [CrossRef] [PubMed]
  286. Shi, Z.; Wang, Y.; Ye, W.; Lin, Z.; Deng, T.; Zhang, T.; Zhao, J.; Tong, Y.; Shan, Y.; Chen, G. The LipoxinA4 Receptor Agonist BML-111 Ameliorates Intestinal Disruption Following Acute Pancreatitis through the Nrf2-Regulated Antioxidant Pathway. Free Radic. Biol. Med. 2021, 163, 379–391. [Google Scholar] [CrossRef] [PubMed]
  287. Jaén, R.I.; Fernández-Velasco, M.; Terrón, V.; Sánchez-García, S.; Zaragoza, C.; Canales-Bueno, N.; Val-Blasco, A.; Vallejo-Cremades, M.T.; Boscá, L.; Prieto, P. BML-111 Treatment Prevents Cardiac Apoptosis and Oxidative Stress in a Mouse Model of Autoimmune Myocarditis. FASEB J. 2020, 34, 10531–10546. [Google Scholar] [CrossRef]
  288. Val-Blasco, A.; Prieto, P.; Jaén, R.I.; Gil-Fernández, M.; Pajares, M.; Domenech, N.; Terrón, V.; Tamayo, M.; Jorge, I.; Vázquez, J.; et al. Specialized Proresolving Mediators Protect Against Experimental Autoimmune Myocarditis by Modulating Ca2+ Handling and NRF2 Activation. JACC Basic Transl. Sci. 2022, 7, 544–560. [Google Scholar] [CrossRef]
  289. Cao, Y.; Zhou, X.; Yin, Z.; Yu, X.; Yang, Q.; Guo, Q.; Tian, D.; Xiong, X.; Xu, G.; Kuang, X. The Anti-Inflammatory Effect of BML-111 on COPD May Be Mediated by Regulating NLRP3 Inflammasome Activation and ROS Production. Prostaglandins Other Lipid Mediat. 2018, 138, 23–30. [Google Scholar] [CrossRef]
  290. Perretti, M.; Leroy, X.; Bland, E.J.; Montero-Melendez, T. Resolution Pharmacology: Opportunities for Therapeutic Innovation in Inflammation. Trends Pharmacol. Sci. 2015, 36, 737–755. [Google Scholar] [CrossRef]
  291. Kong, X.; Wu, S.-H.; Zhang, L.; Chen, X.-Q. Pilot Application of Lipoxin A4 Analog and Lipoxin A4 Receptor Agonist in Asthmatic Children with Acute Episodes. Exp. Ther. Med. 2017, 14, 2284–2290. [Google Scholar] [CrossRef]
  292. Hasturk, H.; Schulte, F.; Martins, M.; Sherzai, H.; Floros, C.; Cugini, M.; Chiu, C.-J.; Hardt, M.; Van Dyke, T. Safety and Preliminary Efficacy of a Novel Host-Modulatory Therapy for Reducing Gingival Inflammation. Front. Immunol. 2021, 12, 704163. [Google Scholar] [CrossRef] [PubMed]
  293. Lavy, M.; Gauttier, V.; Dumont, A.; Chocteau, F.; Deshayes, S.; Fresquet, J.; Dehame, V.; Girault, I.; Trilleaud, C.; Neyton, S.; et al. ChemR23 Activation Reprograms Macrophages toward a Less Inflammatory Phenotype and Dampens Carcinoma Progression. Front. Immunol. 2023, 14, 1196731. [Google Scholar] [CrossRef] [PubMed]
  294. Jin, H.; He, R.; Oyoshi, M.; Geha, R.S. Animal Models of Atopic Dermatitis. J. Investig. Dermatol. 2009, 129, 31–40. [Google Scholar] [CrossRef] [PubMed]
  295. Gilhar, A.; Reich, K.; Keren, A.; Kabashima, K.; Steinhoff, M.; Paus, R. Mouse Models of Atopic Dermatitis: A Critical Reappraisal. Exp. Dermatol. 2021, 30, 319–336. [Google Scholar] [CrossRef]
  296. Sakamoto, K.; Nagao, K. Mouse Models for Atopic Dermatitis. Curr. Protoc. 2023, 3, e709. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram illustrating the development of skin chronic inflammation in AD. Disruption of the epidermal barrier (mainly caused by mechanical scratch) activates and damages keratinocytes, which is accompanied by secretion of a wide range of pro-inflammatory cytokines and chemokines. Cytokines TSLP, IL-25, and IL-33, classified as alarmins, activate ILC2 cells. In response, ILC2 cells produce IL-5 and IL-13, which activate Th2 cells, leading to increased production of IL-4 and IL-13 that activate B cells to produce IgE. IL-5, produced by Th2 cells, chemoattracts eosinophils (not depicted in the diagram), presumably contributing to AD pathogenesis. Secreted by keratinocytes, TSLP activates dendritic cells (DCs), resulting in enhanced production of TNFα and IL-6, which, in turn, activate Th-22 cells. DCs secrete IL-25, which activates keratinocytes, whereas secreted by DCs IL-12, IL-21, and TGFβ activate Th-17 cells. In turn, IL-22 from Th22 cells and IL-17 from Th17 cells cause thickening of epidermis. Activation of inflammatory dendritic epidermal cells (IDEC) by chemokines TARC, CCL17, CTACK, and MDS leads to enhanced secretion of IL-12 and IL-18, which activate Th1 cells to produce IL-1α, IL-12, IFNγ, and TGFβ capable of keratinocyte stimulation, thereby worsening skin inflammation. IL-31 stimulates the differentiation of keratinocytes, whereas IL-25 and IL-33 promote itch (pruritus) and skin barrier disruption. Adipokines secrete pro-inflammatory molecules, such as free fatty acids (FFAs), leptin, TNFα, IL-6 and some others capable of activating DCs and Th2 cells. All these events create a vicious cycle that governs and maintains chronic skin inflammation in AD. Further explanations are given in the text. Abbreviations: CCL17, CC motif chemokine ligand 17; CTACK, cutaneous T-cell attracting chemokine; DC, dendritic cell; FFAs, free fatty acids; IDEC, inflammatory dendritic epidermal cell; ILC2, group 2 innate lymphoid cell, MDC, macrophage-derived chemokine; TGF, transforming growth factor; TARC, thymus- and activation-regulated chemokine; Th, T helper cell; TNF, tumor necrosis factor; TSLP, thymic stromal lymphopoietin.
Figure 1. Schematic diagram illustrating the development of skin chronic inflammation in AD. Disruption of the epidermal barrier (mainly caused by mechanical scratch) activates and damages keratinocytes, which is accompanied by secretion of a wide range of pro-inflammatory cytokines and chemokines. Cytokines TSLP, IL-25, and IL-33, classified as alarmins, activate ILC2 cells. In response, ILC2 cells produce IL-5 and IL-13, which activate Th2 cells, leading to increased production of IL-4 and IL-13 that activate B cells to produce IgE. IL-5, produced by Th2 cells, chemoattracts eosinophils (not depicted in the diagram), presumably contributing to AD pathogenesis. Secreted by keratinocytes, TSLP activates dendritic cells (DCs), resulting in enhanced production of TNFα and IL-6, which, in turn, activate Th-22 cells. DCs secrete IL-25, which activates keratinocytes, whereas secreted by DCs IL-12, IL-21, and TGFβ activate Th-17 cells. In turn, IL-22 from Th22 cells and IL-17 from Th17 cells cause thickening of epidermis. Activation of inflammatory dendritic epidermal cells (IDEC) by chemokines TARC, CCL17, CTACK, and MDS leads to enhanced secretion of IL-12 and IL-18, which activate Th1 cells to produce IL-1α, IL-12, IFNγ, and TGFβ capable of keratinocyte stimulation, thereby worsening skin inflammation. IL-31 stimulates the differentiation of keratinocytes, whereas IL-25 and IL-33 promote itch (pruritus) and skin barrier disruption. Adipokines secrete pro-inflammatory molecules, such as free fatty acids (FFAs), leptin, TNFα, IL-6 and some others capable of activating DCs and Th2 cells. All these events create a vicious cycle that governs and maintains chronic skin inflammation in AD. Further explanations are given in the text. Abbreviations: CCL17, CC motif chemokine ligand 17; CTACK, cutaneous T-cell attracting chemokine; DC, dendritic cell; FFAs, free fatty acids; IDEC, inflammatory dendritic epidermal cell; ILC2, group 2 innate lymphoid cell, MDC, macrophage-derived chemokine; TGF, transforming growth factor; TARC, thymus- and activation-regulated chemokine; Th, T helper cell; TNF, tumor necrosis factor; TSLP, thymic stromal lymphopoietin.
Cells 13 01899 g001
Figure 2. Schematic presentation of the involvement of miRNAs in the pathogenesis of AD. The upper panel of the figure indicates that down- or up-regulation of the expression of miRNAs leads to T-cell imbalance as a prerequisite for the development and maintenance of chronic inflammation. The lower panel shows skin barrier disruption that exacerbates AD manifestations. Down-arrows and up-arrows show decreased and increased expression of miRNA, respectively. Further explanations are given in the text. Abbreviations: Bcl2L2, Bcl-2-like protein 2; CTLA4, cytotoxic T-lymphocyte associated protein 4; HAS3, hyaluronan synthase 3; HMGB1, high mobility group box 1 protein; IL12RB2, IL-12 receptor subunit beta 2; IL-13Rα1, IL-13 receptor subunit alpha 1; LEKTI, lympho-epithelial Kazal type inhibitor; MMPs, matrix metalloproteinases; SOX6, transcription factor SOX-6; YAP1, Yes-associated protein.
Figure 2. Schematic presentation of the involvement of miRNAs in the pathogenesis of AD. The upper panel of the figure indicates that down- or up-regulation of the expression of miRNAs leads to T-cell imbalance as a prerequisite for the development and maintenance of chronic inflammation. The lower panel shows skin barrier disruption that exacerbates AD manifestations. Down-arrows and up-arrows show decreased and increased expression of miRNA, respectively. Further explanations are given in the text. Abbreviations: Bcl2L2, Bcl-2-like protein 2; CTLA4, cytotoxic T-lymphocyte associated protein 4; HAS3, hyaluronan synthase 3; HMGB1, high mobility group box 1 protein; IL12RB2, IL-12 receptor subunit beta 2; IL-13Rα1, IL-13 receptor subunit alpha 1; LEKTI, lympho-epithelial Kazal type inhibitor; MMPs, matrix metalloproteinases; SOX6, transcription factor SOX-6; YAP1, Yes-associated protein.
Cells 13 01899 g002
Figure 3. Schematic presentation of the skin-gut axis involvement in chronic inflammation development associated with AD pathogenesis. Skin dysbiosis (left-hand side of the diagram) in AD is characterized by a clear predominance of Staphylococcus aureus (S. aureus) along with an increase in the prevalence of some other pathogenic microorganisms (pathobionts) and a decrease in the prevalence of commensals. This leads to the formation of biofilm that weakens the host’s immune response as well as the production of several virulence factors, such as toxins and proteases that damage the epithelial barrier and increase histamine release and IgE levels. Reduced production of antimicrobial peptides (AMPs) (i.e., defensins and Cathelicidin LL-37) and tryptophan metabolites (i.e., indole-3-aldehyde) leads to decreased skin protective activity and increased secretion of TSLP by keratinocytes, thereby deteriorating the epidermal barrier of the skin. Gut dysbiosis (right-hand side of the diagram) is characterized by a decrease in the prevalence of microorganisms that produce short-chain fatty acids (SCFAs) (i.e., butyrate and propionate), which leads to disturbed barrier integrity, lipopolysaccharide (LPS) leakage, increased and reduced production of pro- and anti-inflammatory cytokines, respectively. Similar to skin dysbiosis, gut dysbiosis is also characterized by reduced production of AMPs and tryptophan metabolites. Overall, these events create a skin-gut axis that contributes to AD-associated chronic inflammation and worsening of AD manifestations. Further explanations are given in the text. Abbreviations: AMP, antimicrobial peptides; LPS, lipopolysaccharide; SCFAs, short-chain fatty acids; S. aureus, Staphylococcus aureus; TSLP, thymic stromal lymphopoietin.
Figure 3. Schematic presentation of the skin-gut axis involvement in chronic inflammation development associated with AD pathogenesis. Skin dysbiosis (left-hand side of the diagram) in AD is characterized by a clear predominance of Staphylococcus aureus (S. aureus) along with an increase in the prevalence of some other pathogenic microorganisms (pathobionts) and a decrease in the prevalence of commensals. This leads to the formation of biofilm that weakens the host’s immune response as well as the production of several virulence factors, such as toxins and proteases that damage the epithelial barrier and increase histamine release and IgE levels. Reduced production of antimicrobial peptides (AMPs) (i.e., defensins and Cathelicidin LL-37) and tryptophan metabolites (i.e., indole-3-aldehyde) leads to decreased skin protective activity and increased secretion of TSLP by keratinocytes, thereby deteriorating the epidermal barrier of the skin. Gut dysbiosis (right-hand side of the diagram) is characterized by a decrease in the prevalence of microorganisms that produce short-chain fatty acids (SCFAs) (i.e., butyrate and propionate), which leads to disturbed barrier integrity, lipopolysaccharide (LPS) leakage, increased and reduced production of pro- and anti-inflammatory cytokines, respectively. Similar to skin dysbiosis, gut dysbiosis is also characterized by reduced production of AMPs and tryptophan metabolites. Overall, these events create a skin-gut axis that contributes to AD-associated chronic inflammation and worsening of AD manifestations. Further explanations are given in the text. Abbreviations: AMP, antimicrobial peptides; LPS, lipopolysaccharide; SCFAs, short-chain fatty acids; S. aureus, Staphylococcus aureus; TSLP, thymic stromal lymphopoietin.
Cells 13 01899 g003
Figure 4. Hypothetical mechanisms underlying the role of SPMs in inflammation resolution and attenuation of pain and itch in AD. SPMs (upper line of the diagram) transmit signals through an array of receptors expressed in various immune cells, keratinocytes and skin nociceptors, which also express transient receptor potential (TRP) ion channels, mainly the vanilloid (TRPV) and ankyrin 1 (TRPA1) subtypes, and voltage-gated sodium ion channels (Nav 1.4, Nav1.7-9) (left-hand side of the diagram). These pain/itch-signaling receptors are also expressed in spinal nociceptors, which also express SPM receptors on indicated immune and neuronal cells (right-hand side of the diagram). The inflammation-resolving effects provided by SPMs through binding to their receptors on immune (A) and neuronal (B) cells ultimately result in suppression of pain/itch signaling, thereby attenuating AD manifestations. Further explanations are given in the text. Abbreviations: M2, anti-inflammatory subtype of macrophages; NF-κB, nuclear factor kappa B.
Figure 4. Hypothetical mechanisms underlying the role of SPMs in inflammation resolution and attenuation of pain and itch in AD. SPMs (upper line of the diagram) transmit signals through an array of receptors expressed in various immune cells, keratinocytes and skin nociceptors, which also express transient receptor potential (TRP) ion channels, mainly the vanilloid (TRPV) and ankyrin 1 (TRPA1) subtypes, and voltage-gated sodium ion channels (Nav 1.4, Nav1.7-9) (left-hand side of the diagram). These pain/itch-signaling receptors are also expressed in spinal nociceptors, which also express SPM receptors on indicated immune and neuronal cells (right-hand side of the diagram). The inflammation-resolving effects provided by SPMs through binding to their receptors on immune (A) and neuronal (B) cells ultimately result in suppression of pain/itch signaling, thereby attenuating AD manifestations. Further explanations are given in the text. Abbreviations: M2, anti-inflammatory subtype of macrophages; NF-κB, nuclear factor kappa B.
Cells 13 01899 g004
Figure 5. A proposed therapeutic strategy to resolve failed chronic inflammation in AD. We suggest that failed chronic inflammation is a key element in AD pathogenesis. This occurs under the synergistic actions of at least three processes: (1) the reduced production and functional activity of SPMs, (2) skin and gut dysbiosis; and (3) epigenetic disturbances. In accordance, diminution of the activities of these chronic inflammation inducers could reverse AD manifestations. To achieve this overall aim, we propose a multilevel approach: (1) the application of stable, small-molecule SPM mimetics and receptor agonists; (2) the convalescence of dysbiosis manifestation by supplementation of probiotics/prebiotics; and (3) the correction of epigenetic disturbances by epidrugs. This treatment strategy can be used in conjunction with current AD therapy, which mainly includes JAKi and anti-cytokine monoclonal antibodies. This combined approach, in our view, would lead to the resolution of chronic inflammation, ultimately leading to the gradual amelioration of AD manifestations. Further explanations are provided in the text.
Figure 5. A proposed therapeutic strategy to resolve failed chronic inflammation in AD. We suggest that failed chronic inflammation is a key element in AD pathogenesis. This occurs under the synergistic actions of at least three processes: (1) the reduced production and functional activity of SPMs, (2) skin and gut dysbiosis; and (3) epigenetic disturbances. In accordance, diminution of the activities of these chronic inflammation inducers could reverse AD manifestations. To achieve this overall aim, we propose a multilevel approach: (1) the application of stable, small-molecule SPM mimetics and receptor agonists; (2) the convalescence of dysbiosis manifestation by supplementation of probiotics/prebiotics; and (3) the correction of epigenetic disturbances by epidrugs. This treatment strategy can be used in conjunction with current AD therapy, which mainly includes JAKi and anti-cytokine monoclonal antibodies. This combined approach, in our view, would lead to the resolution of chronic inflammation, ultimately leading to the gradual amelioration of AD manifestations. Further explanations are provided in the text.
Cells 13 01899 g005
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

Livshits, G.; Kalinkovich, A. Resolution of Chronic Inflammation, Restoration of Epigenetic Disturbances and Correction of Dysbiosis as an Adjunctive Approach to the Treatment of Atopic Dermatitis. Cells 2024, 13, 1899. https://doi.org/10.3390/cells13221899

AMA Style

Livshits G, Kalinkovich A. Resolution of Chronic Inflammation, Restoration of Epigenetic Disturbances and Correction of Dysbiosis as an Adjunctive Approach to the Treatment of Atopic Dermatitis. Cells. 2024; 13(22):1899. https://doi.org/10.3390/cells13221899

Chicago/Turabian Style

Livshits, Gregory, and Alexander Kalinkovich. 2024. "Resolution of Chronic Inflammation, Restoration of Epigenetic Disturbances and Correction of Dysbiosis as an Adjunctive Approach to the Treatment of Atopic Dermatitis" Cells 13, no. 22: 1899. https://doi.org/10.3390/cells13221899

APA Style

Livshits, G., & Kalinkovich, A. (2024). Resolution of Chronic Inflammation, Restoration of Epigenetic Disturbances and Correction of Dysbiosis as an Adjunctive Approach to the Treatment of Atopic Dermatitis. Cells, 13(22), 1899. https://doi.org/10.3390/cells13221899

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