Next Article in Journal
Opportunities and Challenges of Data-Driven Virus Discovery
Next Article in Special Issue
Transcriptome Meta-Analysis Confirms the Hidradenitis Suppurativa Pathogenic Triad: Upregulated Inflammation, Altered Epithelial Organization, and Dysregulated Metabolic Signaling
Previous Article in Journal
CD, UV, and In Silico Insights on the Effect of 1,3-Bis(1′-uracilyl)-2-propanone on Serum Albumin Structure
Previous Article in Special Issue
Hair Follicle-Related MicroRNA-34a Serum Expression and rs2666433A/G Variant in Patients with Alopecia: A Cross-Sectional Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Inflammatory microRNAs for Treating Inflammatory Skin Diseases

1
Department of Microbiology, Soochow University, Taipei 111, Taiwan
2
Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
3
Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
4
Department of Environmental Engineering and Health, Yuanpei University, Hsinchu 300, Taiwan
5
Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11362, Saudi Arabia
6
Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan 333, Taiwan
7
Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan
*
Author to whom correspondence should be addressed.
Biomolecules 2022, 12(8), 1072; https://doi.org/10.3390/biom12081072
Submission received: 12 July 2022 / Revised: 1 August 2022 / Accepted: 2 August 2022 / Published: 3 August 2022

Abstract

:
Skin inflammation occurs due to immune dysregulation because of internal disorders, infections, and allergic reactions. The inflammation of the skin is a major sign of chronic autoimmune inflammatory diseases, such as psoriasis, atopic dermatitis (AD), and lupus erythematosus. Although there are many therapies for treating these cutaneous inflammation diseases, their recurrence rates are high due to incomplete resolution. MicroRNA (miRNA) plays a critical role in skin inflammation by regulating the expression of protein-coding genes at the posttranscriptional level during pathogenesis and homeostasis maintenance. Some miRNAs possess anti-inflammatory features, which are beneficial for mitigating the inflammatory response. miRNAs that are reduced in inflammatory skin diseases can be supplied transiently using miRNA mimics and agomir. miRNA-based therapies that can target multiple genes in a given pathway are potential candidates for the treatment of skin inflammation. This review article offers an overview of the function of miRNA in skin inflammation regulation, with a focus on psoriasis, AD, and cutaneous wounds. Some bioactive molecules can target and modulate miRNAs to achieve the objective of inflammation suppression. This review also reports the anti-inflammatory efficacy of these molecules through modulating miRNA expression. The main limitations of miRNA-based therapies are rapid biodegradation and poor skin and cell penetration. Consideration was given to improving these drawbacks using the approaches of cell-penetrating peptides (CPPs), nanocarriers, exosomes, and low-frequency ultrasound. A formulation design for successful miRNA delivery into skin and target cells is also described in this review. The possible use of miRNAs as biomarkers and therapeutic modalities could open a novel opportunity for the diagnosis and treatment of inflammation-associated skin diseases.

1. Introduction

Ribonucleic acids (RNAs) are able to regulate gene expression at the transcriptional, posttranscriptional, and epigenetic stages. Noncoding RNAs comprise a major portion of the human transcriptome. These functional RNAs include ribosomal RNA (rRNA), long noncoding RNA (lncRNA), small interfering RNA (siRNA), piwi-interacting RNA (piRNA), and microRNA (miRNA) [1]. Among these, miRNA is a small and highly conserved noncoding RNA sequence containing 19–25 nucleotides. This single-stranded RNA can regulate the expression of protein-coding genes at the posttranscriptional level to join the maintenance of correct cell homeostasis [2]. Since the discovery of miRNA in 1993, 5000–10,000 miRNAs have been found in mammals. miRNAs comprise 1–5% of all genes in the human genome [3]. Approximately 20–60% of protein-coding genes are regulated by miRNAs. miRNAs participate in cell development, morphogenesis, proliferation, apoptosis, differentiation, immune regulation, and wound healing [4]. Under the condition of disease, miRNAs can change to induce altered gene expressions, leading to aberrant phenotypes. On the other hand, they also predominate the protective capacity by reestablishing cell homeostasis [5]. The balance of miRNA plays a key role in the correct functioning of cell physiology. The exploration of miRNA has advanced the development of molecular biology, bioinformatics, and translational investigation.
Most miRNAs are transcribed from deoxyribonucleic acid (DNA) sequences in the nucleus by RNA polymerases [6]. Drosha is a member of the RNase III family that cleaves the primary miRNA (pri-miRNA) to generate a 70-nucleotide precursor miRNA (pre-miRNA). The pre-miRNA is transported to the cytoplasm by exportin-5 and is then processed by the RNase III endonuclease dicer to produce mature miRNA. The mature miRNA is loaded onto the RNA-induced silencing complex (RISC) as guided by the Argonaute (AGO) family of proteins for binding to the 3′-untranslated region (3′UTR) of the target messenger RNA (mRNA) [7]. This can result in the translation-suppression or degradation of the target mRNA. The biogenesis of miRNA and its impact on mRNA are illustrated in Figure 1. miRNA dysregulation is involved in a broad range of diseases, including developmental abnormalities, cancer, metabolic disease, autoimmune disorders, and cardiovascular dysfunction [8]. The modulation of disease-associated miRNAs is beneficial for the targeted therapy of several diseases. Different from the conventional approaches of turning off specific targets, miRNAs exert a biological function by tuning protein-coding genes. miRNA expression modulation has some advantages. Introducing siRNA into cells can reduce the expression of specific genes. However, an unpredicted effect of siRNA on the cells via off-target effects may happen. In the case of miRNA, one exogenous miRNA can modulate several genes that often act in the same biological pathway. In addition, the action of miRNA is designed by nature itself. The intervention of miRNA-based therapies usually causes limited toxicity or adverse impacts [9]. Besides its action in cell interiors, miRNA can be released into plasma, tissue fluid, urine, and milk. miRNA is protected by exosomes or combined with high-density lipoproteins to avoid enzymatic degradation in the plasma [10]. The exosomal miRNA secreted by the cells exerts a vital role in cell-to-cell communication. It is capable of penetrating neighboring cells and can control the expression of genes [11]. In the past few years, several miRNA-based therapeutics have been developed and are currently in different phases of clinical trials [12]. Clinical trials of numerous miRNAs have shown positive results in their initial phases. Some miRNA molecules are in different stages of clinical trials, including the treatment of hepatitis C virus infection (phase II), mycosis fungoides (phase II), polycystic kidney disease (phase I), cutaneous T cell lymphoma (phase I), hepatocellular carcinoma (phase I), malignant pleural mesothelioma (phase I), ischemia (phase I), heart failure (phase I), and idiopathic pulmonary fibrosis (phase I) [13,14]. Although a future therapeutic application of miRNAs is appealing, there are still great practical difficulties to overcome, such as the identification of proper administration routes, the control of in-body stability, the targeting of specific cells, and the attaining of the intended intracellular effects.
Inflammation is a protective strategy of the cells to neutralize the stimuli-including pathogens, toxins, irritants, mechanical stress, and allergens. However, inappropriate inflammation can cause tissue damage. The activation of excessive inflammation is detected by sensors, such as toll-like receptors (TLRs), which are found in macrophages, dendritic cells, and mast cells [15]. This activation induces the production of proinflammatory mediators, including cytokines and chemokines. Apart from the role of regulating cell-normalization processes, miRNA exhibits disturbed expression in inflammatory and autoimmune diseases. Altered miRNA expression is associated with inflammatory signaling, increased cytokine release, and preservation of the vicious cycle in autoimmunity [16]. On the other hand, miRNA can act as part of a negative regulatory loop to keep inflammation in check by elevating anti-inflammatory mediator generation for the return to homeostasis [17]. As the largest organ of the human body, the skin requires a large amount of highly regulated miRNAs for its development and morphogenesis. miRNAs are involved in skin immunity, cell proliferation, aging, pigmentation, wound healing, and cutaneous microbiomes [18,19]. miRNAs also play a role in skin cancers, inflammatory skin diseases, and autoimmune skin disorders. Thus, miRNAs can be biomarkers for skin diseases because of the different expression levels of miRNAs between lesional and healthy skin. miRNAs can also be cell-specific markers for skin disease diagnosis and prognosis. The treatment efficacy and the therapeutic outcome can also be evaluated by changes in the miRNA levels. For example, psoriasis patients show higher levels of miR-125b, miR-146a, miR-203, and miR-223 in serum as compared with healthy subjects [20,21]. It has also been observed that miR-424 is largely detected in the hair shafts of psoriasis patients as compared with normal subjects and those with atopic dermatitis (AD) [22]. The serum of pediatric AD patients shows upregulated miR-203 and miR-483-5p levels compared with healthy groups [23]. miR-194-5p is a useful biomarker for AD diagnosis because of its downregulation in the plasma of AD patients [24]. Another AD biomarker, detected in peripheral CD4+ T cells, is the significant elevation of miR-155 in AD patients [25].
To date, no successful therapy has been found to completely cure autoimmune and inflammatory skin diseases and prevent their recurrence. Current long-term therapy is also difficult because of the inefficiency after prolonged application and the adverse effects of the treatments. There is an emerging need to develop efficient therapeutic strategies to manage chronic inflammatory skin diseases. miRNA-based therapies have become potential candidates for treating inflammatory skin diseases over the last decade. The dermatological or cosmeceutical application of active ingredients to regulate miRNA expression for treating skin diseases has also been largely recognized [26]. There are two approaches for employing miRNA as a gene modulator: miRNA inhibitors/antagomirs, and miRNA mimics/agomirs. The miRNA inhibitor/antagomir approach is utilized for the aberrantly expressed miRNAs that are upregulated in diseases, while anti-miRNA or miRNA inhibitors specifically bind to endogenous mature miRNA, thus preventing targeted miRNA expression [27]. In contrast, miRNAs that are reduced in diseases can be supplied transiently using miRNA mimics and agomirs. Mimics are chemically designed and synthesized to simulate endogenous miRNAs. This review focuses on miRNA mimics that exert anti-inflammatory activity for treating cutaneous inflammation-related diseases, including psoriasis, AD, lupus, skin wounds, and skin aging. Natural or synthetic actives and drugs that can modulate miRNAs for mitigating skin inflammation are also discussed in this study. Most naked miRNAs can usually neither permeate the skin nor facilely cross the cell membrane because of their large size and negative charge [28]. A delivery system is, therefore, required for miRNA administration. This study additionally highlights the emerging approach of formulating designs for miRNAs to achieve successful and efficient delivery into the nidus or target cells.

2. miRNAs and Inflammation

Inflammation is a complicated pathophysiological cascade of the response to infection or injury. The mechanism of inflammation is closely associated with many human diseases. The magnitude and network of pro- and anti-inflammatory factors affect the development and progression of various diseases. Inflammation regulation can be governed by the coordinated control of gene expression in participating immune cells and systems [29]. miRNA is the key gene regulator to achieve inflammation control. Anti-inflammatory miRNAs are fine-tuned signaling regulators that allow the resolution and prevention of inflammatory reactions in immune cells [30]. miRNA has an extensive spectrum of biofunctions for inflammation regulation in immune cells (Figure 2). miRNAs can either enhance or inhibit inflammation, depending on the target mRNAs. The immune system employs multiple miRNAs to manage the functional capacity for constructing a balance between activation and suppression. Innate defense pathway stimulation, such as that found in TLR signaling, contributes to the altered expression of miRNAs that modulate inflammatory genes. Some anti-inflammatory miRNAs modulate the translation of transcripts, leading to a reduction in the immunomodulating factor levels for inhibiting or regulating inflammatory responses [31].
Some miRNAs inhibit multiple target genes involved in inflammation-related signaling. The manipulation of the miRNA expression level offers an applicable therapy against inflammatory diseases. The targeting of the inflammatory response through miRNA mimics could be an effective treatment. Anti-inflammatory miRNA mimics for inflammation mitigation have been previously reported [17]. These include miR-10a, miR-21, miR-24, miR-106b, miR-124, miR-143, miR-145, miR-146, miR-155, and miR-375. These miRNAs can be a negative regulator of inflammation by targeting several inflammation-related pathways, such as TLR, signal transducer and activator of transcription (STAT), nuclear factor-κB (NF-κB), tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6), and Janus kinase (JAK). The overexpression of anti-inflammatory miRNAs in turn abrogates the production of proinflammatory cytokines and chemokines in the immune cells, resulting in the attenuation of the inflammatory response [32]. Numerous miRNAs function in the downregulation of inflammatory pathways. For biological consideration, this is an ideal and precise coordination system to control inflammation. As inflammation is initiated, the fast transcriptional upregulation of the proinflammatory mediators occurs. At the same time, the expression of some miRNAs is initiated by the same transcription. These miRNAs either restrain the expression of the positive signaling proteins or inhibit the same pathway [33]. Altered miRNA expression and supplementary anti-inflammatory miRNA mimics have been successfully used to treat inflammatory and immunological skin disorders. This can open a new field to explore pathogenesis, develop novel biomarkers for diagnosis, and design mechanism-driven therapeutic approaches.

3. miRNAs for Treating Inflammatory Skin Diseases

3.1. Psoriasis

Psoriasis is a chronic autoimmune skin disease delineated by epidermal hyperplasia and inflammatory cell infiltration. The worldwide prevalence of psoriasis is 2–3%, and patients with severe psoriasis have a shortened life expectancy [34]. Both genetic and environmental factors, in association with irregular immune systems, are considered to be involved in psoriatic pathogenesis. Keratinocytes and immune cells are responsible for the production of proinflammatory mediators after activation, leading to keratinocyte proliferation and amplification loops in psoriatic lesions [35]. The suppression of hyperproliferation and inflammation is a target for antipsoriatic therapies [36]. Psoriasis is strongly dependent on genomic variation. A growing number of psoriasis-susceptible genes involved in immunity and keratinocyte function have been discovered [37]. The elucidation of these genes is essential to understand the pathogenetic mechanisms of psoriasis. miRNA dysregulation has been detected in psoriasis patients. Since the first discovery of altered miRNA expression in psoriasis [38], more than 250 miRNAs have been found to be differentially expressed in the skin and blood of psoriasis patients [39]. miRNAs have the potential to predominate the proliferation, apoptosis, differentiation, and proinflammatory mediator production of keratinocytes, as well as the activation of immune cells [40]. Increasing evidence highlights the successful use of miRNAs as psoriasis biomarkers for diagnosis, prognosis, and therapeutic response monitoring (Figure 3).
It has been proven that miR-21, miR-31, miR-146a, miR-155, and miR-203 are greatly upregulated in the lesional skin of psoriasis patients [41], among which miR-21-3p and miR-21-5p play a crucial role in epidermal inflammation. miR-31 functions in the positive vicious loop in psoriatic keratinocytes through proliferative, differentiative, and inflammatory mechanisms [42]. On the other hand, miR-146a and miR-203 are known to hold negative potential in epidermal inflammation, participating in the balance of keratinocyte proliferation and differentiation [43]. The overexpression of miR-155 in psoriasis skin reduces loricrin expression in keratinocytes and disrupts the epidermal barrier’s properties [44]. In addition to lesional skin, some miRNAs in the serum of psoriasis patients are increased as compared with healthy patients. The serum levels of miR-33, miR-126, miR-143, and miR-223 are elevated in psoriasis patients, serving as biomarkers for disease severity and therapeutic outcome [45]. Both inflammatory and anti-inflammatory miRNAs are associated with the initiation, development, and maintenance of psoriasis. When treating psoriasis, some miRNA mimics can be administered to patients to relieve symptoms via genomic regulation.
miR-99a is downregulated in psoriatic lesions by targeting Frizzled (FZD)5 and FZD8. The wingless-related integration site (Wnt)/β-catenin axis plays an important role in cell proliferation. This pathway is activated by binding the Wnt ligand to the FZD receptor protein [46]. Shen et al. [47] delivered miR-99a mimics into keratinocytes (HaCaT) to achieve miR-99a overexpression. The miR-99a mimics suppressed keratinocyte proliferation via the reduction of FZDs by about two-fold. The examination of FZD expression in the lesional skin of psoriasis patients exhibited an inverse correlation of miR99a with FZD5 (p = 0.018) and FZD8 (p = 0.003). miR-125a was found to be intimately related to immunity and inflammation [48]. The quantitative reverse-transcription polymerase chain reaction (RT-qPCR) data from 60 psoriasis patients demonstrated the reduction of miR-125a in lesional skin compared with non-lesional sites [49]. miR-125a was negatively correlated with TNF-α (p = 0.001), IL-1β (p = 0.014), and IL-17 (p = 0.003) in lesional skin. The miR-125a mimic transfection into HaCaT led to the inhibited proliferation and increased apoptosis for abrogating keratinocyte activation.
The miR-146 family consists of miR-146a and miR-146b, which are encoded by genes located on chromosomes 5 and 10, respectively [50]. Srivastava et al. [51] found a protective capability of miR-146a for early psoriasis onset. The genetic deficiencies of miR-146a resulted in exacerbated skin inflammation after imiquimod stimulation in miR-146a−/− mice. Imiquimod is a TLR agonist that induces psoriasiform skin in murine models [52]. The intradermal injection of synthetic miR-146a in wild-type mice bearing psoriasiform dermatitis led to a 14-fold increase in miR-146a expression as compared with the scramble control. This overexpression caused the mitigation of erythema, epidermal thickness, scaling, and neutrophil infiltration. miR-146b can assist miR-146a in the suppression of the inflammatory response in psoriasis [53]. Interferon (IFN)-γ- or TNF-α-stimulated keratinocytes were transfected by miR-146b mimics. The result showed a significant inhibition of IL-1R-associated kinase (IRAK1), fermitin family homolog 1 (FERMT1), IL-8, and chemokine (C-C motif) ligand (CCL)5 after miR-146b treatment. This effect was similar to the result of miR-146a mimic intervention, leading to the hindrance of keratinocyte proliferation. Both miRNAs target similar sets of transcripts. SERPINB2 is a serine protease inhibitor subgroup member of the serpin superfamily. This inhibitor is upregulated under infection and inflammation conditions in macrophages, monocytes, fibroblasts, eosinophils, and keratinocytes [54]. Vaher et al. [55] found that overexpressed SERPINB2 in the psoriatic skin is positively related to psoriasis severity and negatively related to miR-146a/b. Silencing the caspase recruitment domain family member 10 (CARD10) and IRAK, the direct targets of miR-146a/b, reduced SERPINB2 expression in keratinocytes. Thus, miR-146a/b and SERPINB2 coordinately act in the hindrance of psoriasis-associated inflammation.
Tang et al. [56] demonstrated that miR-187 declines in cytokine-activated HaCaT and the lesional skin of psoriasis patients. In their study, the exogenous miR-187 agomir (10 nmol) was intradermally delivered to imiquimod-treated psoriasiform mice to increase the level of miR-187. The overexpression of miR-187 lessened acanthosis and inflammation in the mice, and this effect was due to the hyperproliferation inhibition by targeting CD276. CD276, also known as B7 homolog 3 protein, is an immune checkpoint molecule belonging to the B7-CD28 family [57]. miR-193b-3p is another anti-inflammatory miRNA used to achieve the amelioration of psoriasis. Huang et al. [58] transfected miR-193b-3p mimics in HaCaT and observed suppressed proliferation and NF-κB/STAT3 signaling. The bioinformatic analysis and dual-luciferase reporter assay indicated that miR-193b-3p could diminish keratinocyte activation by directly targeting the Erb-B2 receptor tyrosine kinase 4 (ERBB4). Intradermal injections of miR-193b-3p agomirs into the imiquimod-treated mice dramatically increased miR-193b-3p expression by about six-fold. This overexpression reduced the epidermal thickness from 160 to 50 μm in the psoriasiform skin. miR-203 is largely expressed in keratinocytes to inhibit p63 and suppressors of cytokine signaling (SOCS)3 for regulating cell differentiation [59]. Wang et al. [60] verified the role of miR-203a in inflammation regulation in psoriasis by pcDNA3.1-miR-203 plasmid transfection into HaCaT. The 3′-untranslated region (UTR) of kynureninase was the conserved target area of the miR-203a. The overexpressed miR-203a (by eight-fold) inhibited kynureninase, thereby inhibiting the production of IL-1β in the keratinocytes. The in vivo psoriasiform mouse model also exhibited miR-203a-induced inversed kynureninase expression during the development of psoriatic inflammation.
The transcriptomic profile of clinical psoriasis verified a downregulation of miR-214-3p in psoriatic lesions compared with healthy skin [61]. The TNF inhibitor adalimumab can increase the miR-241-3p levels in the lesional skin of psoriasis patients by 1.7-fold [41]. Zhao et al. [62] demonstrated a negative regulation of forkhead box M1 (FOXM1) by miR-214-3p, inhibiting keratinocyte hyperproliferation. FOXM1 is a proliferation-specific transcription factor belonging to the forkhead family. The intradermal administration of miR-214-3p mimics in imiquimod-induced psoriasis-like mice alleviated the signs of erythema, scales, and epidermal thickness. FOXM1 expression in the lesions was reduced by about two-fold after miR-214-3p application. Liu et al. [63] indicated the downregulation of miR-215-5p in cytokine-stimulated HaCaT and imiquimod-treated skin tissue. The treatment of miR-215-5p agomirs on imiquimod-treated mice decreased the number of Ki67-positive cells in the epidermis. The luciferase assay suggested that miR-215-5p bound to the 3′UTR of dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) as the direct target. Both cell- and animal-based studies showed that miR-215-5p negatively regulated DYRK1A, inhibiting the downstream pathways of protein kinase B (AKT) and extracellular signal-regulated kinase (ERK). Bian et al. [64] demonstrated that miR-340 reduced IL-17A expression in 293T cells through IL-17A 3′UTR. Imiquimod-stimulated skin inflammation in mice treated with intravenous miR-340 agomir resulted in substantially lower scores for cutaneous redness, scaling, and thickening. The cumulative psoriasis severity score could be reduced from nine to six after agomir treatment. miR-383 functions as a suppressor of tumor progression and cell proliferation [65]. It has been reported that miR-383 can target the 3′UTR of lipocalin 2 (LCN2) and block JAK/STAT activation [66]. Skin cells from imiquimod-treated rats were transfected with miR-383 mimics. Overexpressed miR-383 and decreased LCN2 expression were detected by this transfection. Compared with the control, the miR-383 mimic treatment reduced cell proliferation while increasing cell apoptosis. Ye et al. [67] reported downregulated miR-489-3p expression in psoriasis patients. A further bioinformatic assay and luciferase reporter study indicated the direct targeting of miR-489-3p to TLR4 in keratinocytes. HaCaT cells transfected with miR-489-3p mimics inhibited cell proliferation and TLR4/NF-κB signaling. The TNF-α, IL-1β, IL-22, and IFN-γ levels declined by about two-fold after miR-489-3p transfection. The targets and biological mechanisms of the anti-inflammatory miRNAs for treating psoriasis are summarized in Table 1.

3.2. Atopic Dermatitis (AD)

AD is an inflammatory skin disease characterized by erythema, edema, vesicles, and lichenification. The pathogenesis of AD is involved in inflammation dysregulation and response to antigens. AD can be featured by skin barrier dysfunction, skin microbiome alteration, and type 2 immune responses [68]. The increases in immunoglobulin (Ig)E and eosinophils in the development of AD boost inflammation and skin disruption through the production of oxidative stress, toxic granule proteins, cytokines, and chemokines [69]. Th2-related cytokines, such as IL-4, IL-5, IL-13, IL-22, and IL-31, are largely expressed in AD skin [70]. There are increasing reports of AD comorbidities, including neuropsychiatric, cardiovascular, and malignant disorders [71]. AD is the most common inflammatory skin disorder, affecting 10–25% of children and 2–10% of adults [72]. Recent investigations have illustrated the fundamental role of miRNA in AD pathogenesis (Figure 4) [73]. Elevated expressions of miR-10a, miR-24, miR-27a, miR-29b, miR-146a, miR-151a, miR-193a, miR-199, miR-211, miR-222, miR-4207, and miR-4529-3p were observed in the lesional skin of AD patients [25,74]. On the other hand, miR-135a, miR-143, miR-184, miR-194-5p, and miR-4454 were downregulated in clinical AD. miR-155-5p is also highly expressed in AD lesions, which can activate T cells, increase cutaneous inflammation, and disintegrate tight junctions [75]. miR-720 is upregulated in AD, possibly because of its role in keratinocyte cell cycle regulation [76]. The dysregulation of miR-143, miR-146a, miR-155, and miR-451a in AD can be used as a biomarker to diagnose this inflammatory disorder [77]. These miRNAs function in keratinocyte proliferation regulation, cytokine signaling, the NF-κB-dependent inflammation response, and T cell activation. Since plasma platelets are also involved in the pathogenesis of AD, recent studies [78] have indicated that platelet-associated miRNAs, such as miR-24 and miR-191, are responsible for the worsening of AD symptoms due to platelet activation.
Some miRNAs alleviate inflammation caused by AD through the suppression of the immune response in keratinocytes or immune cells. The mimics or agomirs of these miRNAs are potential candidates for anti-AD therapy. CCL22 is a macrophage-derived chemokine correlated with the severity of AD. Yoon et al. [79] assessed the suppression of the CCL22 gene by miRNA for treating AD in mice. A recombinant strain of Salmonella typhimurium expressing CCL22 miRNA (ST-miRCCL22) was prepared for CCL22 knockdown, and the successful transport of ST-miRCCL22 into the RAW264.7 macrophages was observed. The expression of CCL22 in the mouse splenocytes was reduced by about 10-fold after the ST-miRCCL22 treatment. In the in vivo study of atopic mice, the oral inoculation of ST-miRCCL22 lowered the total scratching counts for seven days. The numbers of IgE, IL-4, and Th17 cells were reduced after this treatment due to the CCL22 downregulation in the activated lymphocytes. miR-10a-5p has been acknowledged as a regulator of cell proliferation and inflammatory responses, and has been found to be upregulated in AD patients and in the proliferation of keratinocytes [80]. After the transfection of miR-10a-5p mimics into IL-1β-stimulated keratinocytes, IL-8 and CCL5 expression was significantly reduced. In the transfected cells, 48% were in the G1/G0 phase, compared with 38% for the untreated control, suggesting that proliferation was inhibited by the mimics. The luciferase assay verified that hyaluronan synthase 3, a positive regulator of keratinocyte proliferation and migration, is the direct target of miR-10a-5p.
Yang et al. [81] found a decreased expression of miR-124 in the lesional skin of AD patients compared with the non-lesional sites. The transfection of miR-124 mimics into keratinocytes elicited a 130-fold increase in miR-124 expression. This increase led to the downregulation of IL-8, CCL5, and CCL8 in the IFN-γ- or TNF-α-activated cells. RELA (the gene name of p65) is the direct target of miR-124 to control the NF-κB-associated inflammatory pathways in activated keratinocytes. IL-13 is a Th2-derived cytokine that can impair the epidermal barrier. IL-13Rα1 is a direct target of miR-143 [82]. IL-13 stimulation on keratinocytes resulted in a decrease in the miR-143 level [83]. The amount of IL-13Rα1 in the IL-13-stimulated keratinocytes was diminished by about 10-fold after transfection with the miR-143 mimics. The forced miR-143 expression prevented the IL-13-induced downregulation of filaggrin, loricrin, and involucrin. The skin barrier function was expected to be restored by this effect. In addition to psoriasis, miR-146a is applicable for AD treatment because of its involvement in immune regulation [84]. Meisgen et al. [85] transfected keratinocytes with synthetic miR-146a and found a remarkable suppression of the TLR2-induced production of TNF-α, IL-8, and CCL20. This downregulation was mediated by the direct targeting of miR-146a to TRAF6 and IRAK1. The transcriptomic analysis revealed that the miR-146a mimics regulated the genes involved in cell–cell communication, keratinocyte immunity, cytokines, chemokines, and antimicrobial peptides. miR-146a overexpression in the keratinocytes also lessened the chemotactic migration of neutrophils (0.54-fold as compared with the control). The evidence of miR-146a’s ability to alleviate skin inflammation in AD was further evaluated in vivo [86]. The AD-like model was established by the topical treatment of MC903, a vitamin D3 analog, on the ears of wild-type and miR-146a−/− mice. The miR-146a-deficient mice developed a stronger inflammation response characterized by increased immune cell infiltration, as well as IFN-γ, CCL5, and CCL8 expression, in the skin. The keratinocyte-based study testified the direct targets of CARD10 and IRAK1 for miR-146a.
The lympho–epithelial kazal-type inhibitor (LEKTI) has relatively low expression in the keratinocytes of AD patients [87]. This decrease might have resulted from the overexpression of Yes-associated protein 1 (YAP1), a regulator of the proliferation of epidermal stem cells [88]. Cheng et al. [89] employed a luciferase reporter assay to approve the target binding of miR-375-3p with the 3′UTR of YAP1. The miR-375-3p expression was upregulated six-fold after the transfection of mimics in HaCaT cells. This upregulation contributed to the inhibition of IL-1β and IL-6, accompanied by a reduction of NF-κ nuclear translocation. The cell proliferation was also restrained by miR-375-3p transfection. miR-1294 has been recognized as a tumor suppressor. The role and regulatory mechanism of miR-1294 in AD were explored by Yan et al. [90]. In an in vitro 3D skin-equivalent model, the miR-1294 mimic treatment reduced the thickening of the lamellar bilayer structure stimulated with IFN-γ and TNF-α. The filaggrin level was reduced by IFN-γ and TNF-α was also recovered by the mimic. The in vivo dinitrochlorobenzene-induced AD-like mouse model exhibited a reduction of the injury score from three to one after the miR-1294 mimic treatment. miR-1294 upregulation decreased inflammation and skin barrier destruction by targeting STAT3 to inhibit NF-κB signaling. The targets and biological mechanisms of the anti-inflammatory miRNAs used to treat AD are summarized in Table 2.

3.3. Skin Wounds

Commonly observed cutaneous wounds include open wounds, infected wounds, diabetic wounds, burn wounds, and acne wounds. Wound healing is a complicated process consisting of four overlapping stages: hemostasis, inflammation, proliferation, and tissue remodeling. After the occurrence of a skin wound, numerous inflammatory cells migrate into the wound area to protect against microbial invasion and repair the damage. The dysregulation of inflammation generates unsuccessful healing, hypertrophic scarring, and keloids [91]. Appropriate inflammation is important for promoting skin wound healing. Nevertheless, redundant inflammation responses prompt pathological damage to wound tissue and delay repair [92]. miRNAs possess a strong potential to regulate both the induction and resolution of inflammation in skin wound healing [93]. The overexpression of miR-21, miR-29b, miR-106b, and miR-146a has been reported to accelerate re-epithelialization and reduce excessive scar generation in wound healing [94]. Contrary to this effect, the downregulation of miR-200c, miR-210, and miR155 is effective in improving wound healing. Because of the role of regulating inflammation and immunity, epidermal keratinocytes are the major cells participating in skin wound healing [95]. The re-epithelialization of the wound area by keratinocyte migration is an essential step of wound closure. Some miRNAs involved in psoriasis, such as miR-21, miR-31, and miR-203, are also implicated in keratinocyte migration [96]. Neutrophils are the primary immune cells in the early inflammatory response during wound repair [97]. The neutrophil-derived miR-142 is required to promote neutrophil migration and increase the ability of the wound site to resist microbial infection [98]. Macrophages are another type of immune cell governing the inflammatory phase during wound repair. miR-21 and miR-223 participate in the regulation of macrophage polarization in cutaneous wounds [99,100]. A clinical trial has investigated the effect of miR-29 mimics (Remlarsen) on skin wounds [101]. Intradermal miR-29 mimic injection into the incisional wound site reduced collagen expression and the development of fibroplasia accompanied by the downregulation of the miR-29 target genes COL1A1, COL1A2, and COL3A1.
The wound-healing process can be accelerated by treatment with anti-inflammatory miRNAs. Li et al. [102] signified the potential of miR-23b to inhibit inflammatory reactions in wound repair. miR-23b agomir transfection into HaCaT showed a wound closure of more than 90% ten days post-wounding in a scratch wound healing assay. Subcutaneous miR-23b injections into excisional wounds in mice decreased the immune cell accumulation and cytokine expression for accelerating healing. α-Smooth-muscle actin (α-SMA) in fibroblasts can secrete collagen for strengthening the wound [103]. miR-23b can promote the release of α-SMA in the fiber pattern. miR-23b inhibits inflammation by targeting apoptotic signal-regulating kinase 1 (ASK1). The miR-31 mimics were effective at enhancing wound healing via increased keratinocyte migration [104]. A surgical wound was created in healthy subjects. The basal level of miR-31 in the skin was low, but quickly increased by 1.9-fold one day after the injury. The miR-31 expression was continuously upregulated to 7.7-fold after seven days. miR-31 overexpression was induced using transforming growth factor (TGF)-β2. Epithelial membrane protein 1 (EMP-1) was the direct target of miR-31 in the keratinocytes. The in vitro scratch assay indicated that the miR-31 mimic increased the migration capability of the keratinocytes by 3.6-fold.
miR-34a is a tumor suppressor with the ability to regulate the immune response [105]. An excisional wound was made in mice to check the effect of miR-34a on wound closure [106]. miR-34a was downregulated in the inflammatory stage and returned to the baseline in the proliferative phase. The miR-34a−/− mice showed impaired healing as compared with the wild-type animals. The re-epithelialization was faster in the group of wild-type mice than in the miR-34a-knockout mice (re-epithelialization percentage of 100% versus 10% after seven days). IL-6/STAT3 signaling was essential in the wound healing of the miR-34a−/− mice. This result indicated the importance of miR-34a in inhibiting the inflammation of excisional wounds. Diabetic ulcers in the foot are the main comorbidity in diabetic patients. The anti-inflammatory property of miRNA could be applicable to accelerate the healing of diabetic wounds. Ban et al. [107] stated that miR-497 mimics could reduce the overexpression of TNF-α, IL-1β, and IL-6 in human dermal fibroblasts under hyperglycemic situations. The therapeutic efficacy of the mimics was investigated by intradermal injections into wounds in diabetic mice. A faster reduction of the wound area was observed in the group receiving miR-497 treatment, with healing of 66% as compared with the negative control group (23%) on day four. The levels of TNF-α, IL-1β, and IL-6 in the injury site decreased by more than 20% compared with the negative control. The targets and biological mechanisms of the anti-inflammatory miRNAs for cutaneous wound healing are summarized in Table 3.

3.4. Other Uses

Hosts with pathogenic infections usually undergo inflammation due to proinflammatory cytokine/chemokine bursts by immune cells [108]. Some anti-inflammatory miRNAs have the potential to treat microbe-stimulated inflammation in the skin [109]. C. acnes is reported to represent more than 30% of the facial microbes in acne patients [110]. miR-146a has been successfully used to repress biofilm-derived C. acnes-triggered inflammation [111]. The overexpression of miR-146a by mimic transfection to keratinocytes showed markedly reduced TLR2-induced TNF-α, IL-6, and IL-8 expression. The data of the luciferase reporter assay suggested that miR-146a bound to the 3′UTR of IRAK1 and TRAF6, resulting in the inhibition of the ERK1/2, NF-κB, and mitogen-activated protein kinase (MAPK) pathways. Candida species are the most common fungal pathogens evoking skin and system infection. Dectin-1 is a significant sensor for β-glucan from Candida [112]. Dectin-1 and β-glucan can trigger the intracellular transduction pathways of CARD. CARD10 is the direct target of miR-146a. Du et al. [113] appraised the effect of miR-146a on the inflammation induced by Candida albicans. The transfection of miR-146a into C. albicans-stimulated THP-1 cells significantly inhibited Dectin-1-elicited TNF-α and IL-6 production by about two-fold. The miR-146a mimic inhibited the C. albicans-induced translocation of NF-κB.
Lupus erythematosus is an autoimmune disorder with a wide range of dermatological manifestations. Lupus erythematosus lesions in the skin share extensive lymphocyte infiltration with a high predominance of CD4 T cells and cytokines, including TNF-α, IL-1α, IL-1β, IL-6, and IL-8 [114]. Huang et al. [115] found a correlation between the increase in Th17 cells and the decrease in miR-590-3p in systemic lupus erythematosus patients and MRL/lpr mice. miR-590-3p agomir transfection promoted the apoptosis of Th17 cells by autophagy suppression via direct targeting of autophagy-related 7 (Atg7). The in vivo treatment of lupus mice using agomir lessened lupus nephritis and the size of skin lesions. Chronic idiopathic urticaria (CIU) is a polyetiological dermatological inflammation disorder. A total of 16 miRNAs were found to be differentially expressed in patients with CIU [116]. Among them, five miRNAs (29c-5p, 361-3p, 2355-3p, 2355-5p, and 4264) were largely increased in CIU, making them potential biomarkers for diagnosing autoimmune urticaria. miRNAs are active in the cell regulation of CIU. The CIU patients showed lower expression of miR-194 and higher thrombospondin 1 (THBS1) as compared with the healthy control [117]. THBS1 was proven to be the target of miR-194 in the luciferase activity assay in 293T cells. miR-194 mimics decreased the amount of TNF-α, IL-1β, IL-6, and IL-8 in mast cells. The mast cell degranulation and histamine release were also lowered by transfection with the mimic.

4. miRNAs as the Targets to Inhibit Skin Inflammation

Besides the capability of miRNAs to directly block mRNA activity and inhibit inflammation, miRNAs can be a target to mediate the anti-inflammatory response [118]. Some chemicals are able to target and modulate miRNAs to attain the aim of inflammation suppression. It has been proposed that miRNAs could be a group of biological target molecules for therapeutic intention. The application of compounds that target pri-miRNAs, pre-miRNAs, miRNA processing, and loading into the RISC structure has potential for drug design and development. Some bioactive molecules may impact endogenous miRNA synthesis through downregulation or upregulation, thereby contributing to inflammation suppression [119]. For instance, resveratrol, from the stilbene group, is considered to be beneficial for skin health. This polyphenol has been broadly reported as a potential molecule to treat various skin disorders, including skin cancer, photoaging, allergy, dermatitis, melanogenesis, and microbial infections [120]. Wang and Zhang [121] demonstrated the upregulation of miR-17 by resveratrol for reducing lipopolysaccharide-induced skin inflammation. The resveratrol intervention inhibited the production of TNF-α, IL-6, and IL-8 in the lipopolysaccharide-activated HaCaT. miR-17 was upregulated three-fold after resveratrol treatment. miR-17 silencing enhanced the expression of cytokines in the resveratrol-treated lipopolysaccharide-activated cells. The resveratrol–miR-17 axis was found to stimulate the phosphatase and tensin homolog (PTEN)/phosphoinositide 3-kinases (PI3Ks)/AKT and mammalian target of rapamycin (mTOR) pathways.
Adalimumab (Humira) is a biological drug used to ameliorate psoriatic inflammation via TNF inhibition [122]. In a clinical trial of psoriasis patients [123], adalimumab treatment was found to significantly decrease the psoriasis area and severity score (PASI) and miR-146a-5p in peripheral blood mononuclear cells (PBMCs). The reduction of the miR-146a-5p levels was correlated with the improvement of the PASI. miR-146a-5p could be a dynamic biomarker to predict the therapeutic effectiveness of adalimumab. Ebosin is an exopolysaccharide isolated from fermentation cultures of Streptomyces sp. 139. This compound could mitigate lipopolysaccharide-activated inflammation in HaCaT via IκB kinase (IKK)/NF-κB signaling [124]. Moreover, the PASI score of the imiquimod-treated psoriasiform mice was decreased from 3 to 1.5 with the application of ebosin. Ebosin reduced inflammation by lessening miR-155-3p expression, both in vitro and in vivo. The luciferase activity assay indicated the direct targeting of TNF alpha-induced protein 3 (TNFAIP3) by miR-155-3p. Circular RNA (circRNA) is a type of lncRNA. CircRNA has no 5′ or 3′ end, which endows it with resistance to exonuclease [125]. CircRNA RAB3B, a member of the RAS oncogene family, has been found to be downregulated in psoriasis [126]. CircRAB3B overexpression delayed the proliferation and elevated the apoptotic rate of IL-22-stimulated HaCaT [127]. miR-1228-3p was the target of circRAB3B, and this circRNA negatively regulated the expression of miR-1228-3p in keratinocytes. The luciferase reporter and bioinformatic analyses showed the direct binding of miR-1228-3p to the 3′UTR of PTEN. The combination of 8-methoxypsoralen and UVA (PUVA) is an effective photochemotherapy used to treat psoriasis. Chowdhari and Saini [128] detected significant upregulation of has-miR-4516 in the HaCaT after PUVA application. The transfection of has-miR-4516 mimics decreased STAT3 and pSTAT3 by 1.5-fold in HaCaT cells. The overexpression of has-miR-4516 raised the content of apoptotic keratinocytes from 4% to 24%.
Some bioactives are capable of directly modulating the expression of miRNAs for alleviating inflammatory dermatitis. Berberine is a natural alkaloid derived from Coptis chinensis. This bioactive compound has been proven to show anti-inflammatory, antioxidant, anticancer, and hypolipidemic effects [129]. Berberine attenuated ear swelling from 0.48 to 0.33 mm in a mouse model of allergic dermatitis [130]. This treatment also inhibited miR-21 expression, histamine release, and p38 phosphorylation. The result of the miR-21 mimic transfection in the mast cells indicated that miR-21-mediated suppression in mast cell degranulation was involved in the anti-inflammatory activity of berberine in dermatitis.
Acupuncture, originating from ancient China, involves inserting needles into the body to stimulate sensory nerves in the skin and muscles. Electro-acupuncture is an improvement of traditional acupuncture by the addition of an electrical charge, promoting needle stimulation through electrical impulses [131]. Wang et al. [132] used electro-acupuncture to reduce the inflammation caused by allergic dermatitis. Treatment with electro-acupuncture at the ST36 acupoint resulted in the reduction of the ear thickness from 0.3 to 0.2 mm in allergic dermatitis-like rats. The acupuncture treatment lowered the expression of miR-155 through the signaling of IL-33 for inhibiting p38 phosphorylation. The rat peritoneal mast cells transfected with miR-155 mimics abrogated the inhibitory effect of electro-acupuncture on NF-κB-regulated transcription in response to IL-33. IL-32γ is an anti-inflammatory cytokine that inhibits skin inflammation [133]. Lee et al. [134] proved that the AD severity and epidermal thickness of MC903-induced IL-32γ transgenic mice were lower than those of wild-type animals. The expression of miR-205 was impeded by IL-32γ in the mouse skin and HaCaT cells. The expression of TNF-α, IL-1β, IL-6, and thymic stromal lymphopoietin (TSLP) in IFN-γ/TNF-α-activated keratinocytes. Belinostat is a histone deacetylase inhibitor used for the suppression of hematological and solid malignancies. It could potentially target miR-335 to restore barrier defects in AD [135]. The luciferase reporter analysis confirmed the direct binding of miR-335 to SOX6 3′UTR. The miR-335 level was aberrantly lost in the lesional skin of AD patients. In an ex vivo human organ culture model mimicking the AD phenotype, topical applications of belinostat upregulated filaggrin and involucrin, the downstream of miR-335. Thus, the barrier function of AD-like skin could be recovered by this effect.
Cutaneous wound healing can be accelerated by chemicals such as vitamin D and resveratrol. In addition, some natural extracts are beneficial for treating skin wounds. Ginger has been proven to resolve the problem of poor wound healing [136]. Al-Rawaf et al. [137] combined vitamin D and ginger supplements to treat diabetic wounds in rats. The combined treatment in the diabetic wounds accelerated the epithelialization period from 18.8 to 11.3 days. Compared with healthy rats, diabetic rats exhibited greater levels of miR-155 and lower levels of miR-15a and miR-146a. The combined vitamin D and ginger treatment significantly reversed this tendency. Resveratrol has been shown to be favorable for promoting skin wound repair [138]. Hu et al. [139] discovered the beneficial effect of resveratrol on diabetic wounds by raising the expression of extracellular vesicle (EV)-carried miRNA-129 derived from mesenchymal stem cells (MSCs). Rat MSCs were isolated and treated with resveratrol, and the corresponding EVs were isolated to promote skin wound healing. The size distribution of the EVs ranged between 40 and 150 nm, indicating a nano size. More than 80% of human umbilical-vein endothelial cells (HUVEC) showed internal uptake of EVs based on fluorescence microscopy. The use of resveratrol-treated EVs in diabetic wounds improved the proliferative and migratory potential of the cells. Resveratrol promoted wound healing through TRAF downregulation via MSC-EV-carried miR-219. Curcumin is known to improve diabetic wound repair [140]; however, its low bioavailability and poor aqueous solubility have prevented the clinical application of curcumin. Huang et al. [141] employed (2E,6E)-2,6-bis(2-(trifluoromethyl)benzylidene)cyclohexanone (C66), a synthetic analog of curcumin, to resolve these problems. The C66 treatments in diabetic wounds showed complete closure within 14 days, whereas the non-treatment control had a low closure rate of 64%. The decreased miR-146a level in the diabetic wound was upregulated after C66 treatment. The C66 administration also showed pronounced inhibition of the expression of TNF-α, IL-6, and IL-8. The cell-based study indicated that C66 reversed NF-κB activation due to the overexpressed miR146a in HUVECs.
Jiang Tang Xiao Ke (JTXK) is a traditional Chinese formula containing extracts of pueraria, rehmannia, ginseng, and radix salvia miltiorrhizae. This medicine has been reported to decrease miR-139-5p expression in the pancreatic tissue of diabetic mice [142]. To evaluate the potential of JTXK on S. aureus-infected wound healing, a topical noisome hydrogel was utilized to load JTXK [143]. The in vivo data supported the decreased miR-139-5p expression in the infected wound after topical JTXK administration and the accelerated wound healing rate. Eif4g2, the key downstream mediator of miR-139-5p, was significantly increased by about two-fold by JTXK treatment. Staphylococcus epidermidis plays a vital role in controlling the skin inflammation response. Lipoteichoic acid released from S. epidermidis inhibits C. acnes-mediated inflammation in the skin [144]. Lipoteichoic acid activated TLR2 to upregulate miR-143. This miRNA, in turn, targeted TLR2 to decrease the stability of the TLR2 mRNA and then lessen the TLR2 proteins, thus suppressing the proinflammatory cytokines induced by C. acnes. C. acnes-bearing mice treated with lipoteichoic acid exhibited decreased erythema and ear swelling as compared with the control group. UVB irradiation generates skin photoaging by the induction of cell death and DNA damage. Lee et al. [145] showed the protective activity of troxerutin on UVB-elicited photoaging in keratinocytes. Troxerutin is a natural flavonoid with anti-inflammatory and antioxidant characteristics [146]. An eight-hour pretreatment with 5 μM of troxerutin increased the UVB-irradiated keratinocyte viability by 20%. miRNA gene microarray analysis showed that 68 miRNAs were modulated after troxerutin treatment of UVB-exposed keratinocytes. Among them, the miR-205-3p expression was elevated by 4.3-fold, while miR-483-5p, miR-513b, and miR-3648 were decreased by 16.6-, 23.1-, and 11.6-fold, respectively. Based on these data, the protective effect of troxerutin could be grouped into four functions: apoptosis, proliferation, migration, and DNA repair. The miRNA targets and biological mechanisms of the bioactive molecules for inhibiting skin inflammation in different skin disease models are summarized in Table 4.

5. Approaches for Improving miRNA Delivery

Topical administration routes provide a direct way to treat skin inflammation. Topical drug delivery is a noninvasive and convenient strategy for treating cutaneous disorders. It has the advantages of direct access to the nidus, minimal off-target effects, and the avoidance of systemic responses [147]. miRNA administration via topical absorption can be an ideal approach for applying therapies to the skin [96]. However, the intrinsic barrier function of the stratum corneum, combined with the hydrophilic features of miRNA, has precluded the successful permeation of miRNA into the skin. Even if miRNA can penetrate the inflamed skin, miRNA-mediated gene regulation still requires an intracellular entrance into the target cells. Unfortunately, it is difficult for naked miRNA to permeate the skin and the cell membrane. A delivery system is, therefore, required for facile miRNA administration [148]. Effective topical therapies using miRNA require bypassing the skin barrier and the subsequent miRNA transfection into the target cells. The necessity of using carriers to enhance miRNA delivery is urgent to achieve extensive application in skin inflammation treatments.
Cell-penetrating peptides (CPPs) are one of the strategies for enhancing miRNA penetration into cells and the skin [149]. CPPs are short peptides (<30 amino acids) capable of translocating themselves into cells and facilitating cargo or CPP/cargo complexes to translocate across the plasma membrane [150]. The skin-permeation of bioactives can be improved by using CPPs as penetration enhancers [151]. Urgard et al. [152] used the CPP PepFect6 to form a nanocomplex with miR-146a for treating irritant dermatitis. The nanocomplex exhibited a spherical and homogeneous particle distribution with an average diameter of 30–50 nm. The facile internalization of the miR-146a mimic/PepFect6 nanocomplex into the keratinocytes led to the suppression of the direct targets CARD10 (1.8-fold) and IRAK1 (2.2-fold). In a mouse model of irritant contact dermatitis, the administration of the nanocomplex increased the miR-146a expression by 1380-fold after 30 h. The ear swelling was attenuated 2.4-fold after nanocomplex application. Mulholland et al. [153] developed miRNA-31/CPP nanocarriers within an electrospun nanofiber, with the aim of regenerating skin wounds. The CPP used in this nanocomplex was CHAT, which is a 15-amino-acid linear peptide considered useful for enhancing plasmid DNA delivery [154]. The prepared nanocomplex had a mean size and zeta potential of 74 nm and 9.7 mV, respectively. The transfection percentage of the nanocomplex to the HaCaT cells was greater than 40%. The electrospun nanofiber was advantageous for wound healing due to its biocompatibility and close skin coverage [155]. In vivo, topical treatments of the nanocomplex-loaded electrospun nanofiber on the mouse wound increased the epidermal thickness and angiogenesis as compared with the commercial dressing control.
Nanoparticles are promising delivery systems that could ameliorate the cellular uptake of miRNA. The use of nanoparticles protects miRNA from degradation and improves the efficiency of delivery. Zgheib et al. [156] conjugated miR-146a with cerium oxide nanoparticles for accelerating diabetic wound repair. By scavenging reactive oxygen species (ROS), this type of nanoparticle could eliminate oxidative stress and regulate the imbalance between oxidant and antioxidant enzymes in diabetic wounds [157]. The hydrodynamic diameter of the miR-146a-conjugated nanoparticles was approximately 190 nm. Diabetic wounds were induced by injecting streptozocin into a pig. After a 10-day application, the wound surface area of the nanoparticle group (4.8 cm2) was significantly smaller than that of the control (6.8 cm2). The wound was completely closed on days 14 and 18 after the nanoparticle and saline treatments, respectively. Niemiec et al. [158] further incorporated miR-146a-conjugated nanoparticles into silk fibroin to improve diabetic wound repair. Silk fibroin, composed of biocompatible polymers, is characterized by a strong mechanical structure and the ability to exhibit strain hardening [159]. In the murine model of diabetic wounds, the wounds treated with nanoparticle-incorporated nanosilk and saline were reduced to 31% and 8% of the original size after 13 days, respectively. The human skin samples treated with nanosilk had increased biomechanical strength (51 N) compared with the saline control (42 N). The proinflammatories IL-6 and IL-8 in the wound site were also reduced by the nanosilk application.
Amphipathic bile acid-attached polyethyleneimine (BA-PEI) imparts facile cell membrane permeability by membrane fusion and pore creation [160]. Wang et al. [161] fabricated BA-PEI nanocarriers to load synthetic miR-21 and accelerate excisional wound healing. The nanosystem displayed a size of 173 nm with a zeta potential of 27 mV. An 83-fold increase in miR-21 expression was observed after the treatment with the nanocarriers in HaCaT compared with the saline control. Subcutaneous injections of the nanocarriers in the wound sites of the wild-type mice showed a 57% open wound area after eight days, whereas a 100% open wound area was detected for the group receiving the saline treatment. The nanocarrier-treated wound was fully closed on day 16. There was an open wound and scar formation in the saline group on day 16. Saleh et al. [162] developed bioadhesive hydrogels incorporated with miR-223-5p-loaded hyaluronic acid nanoparticles to control macrophage polarization during wound healing. The hydrogels were composed of gelatin methacryloyl because of its robust attachment to the wound [163]. The miR-223-5p-loaded nanoparticles exhibited a mean diameter of 160 nm and a surface charge of −13 mV. The amount of miR-223-5p in the M1 macrophages was increased by 1541-fold after 24 h of nanoparticle incubation. A murine excisional wound model demonstrated a greater wound closure percentage due to the nanoparticle-laden hydrogels (96%) compared with the naked miRNA (67%), hydrogels (61%), and non-treatment control (45%). The collagen level and epidermal thickness in the wound site were also significantly increased by the nanoparticle-laden hydrogels. Feng et al. [164] prepared biomimetic reconstituted high-density lipoprotein nanogels loaded with miR-210 antisense to explore the anti-inflammatory effect on imiquimod-induced psoriasiform lesions in mice. The average size of the nanocarriers was about 30 nm. Topical application of the nanogels significantly reduced the erythema, scales, and immune cell accumulation in the lesions. The proportion of Th1 and Th17 cells in the lesional skin was decreased by this treatment and was accompanied by decreased IFN-γ and IL-17A.
Exosomes are membrane-enclosed nanovesicles released by cells into extracellular spaces or culture mediums for managing cell–cell communication [165]. Genetic materials, lipids, and proteins are contained inside the exosomes. Because of their biomimetic features and targeting capabilities, exosomes can be used as nanocarriers for drug delivery [166]. Xia et al. [167] designed an exosome-guided cell technique with miRNA-125b transfection to elicit cutaneous wound healing. Fibroblast activation to myofibroblasts can alleviate age-related defects in wound repair. Supplementing wounds with exosomes isolated from young mouse wound-edge fibroblasts largely improved the myofibroblast abundance in the aged mice and promoted fibroblast transition to the myofibroblasts, thus accelerating wound closure. The exosomal transfer of miR-125b to the fibroblasts suppressed sirtuin 7, the direct target of muR-125b, to accelerate myofibroblast differentiation. The replenishment of miR-125b could be a therapeutic strategy to enhance wound repair. To prepare the nanocarriers as gene-delivery systems, soluble potato starch was reacted with a quaternization reagent to produce quaternized starch (Q-starch) [168]. This nanosystem, based on natural polysaccharides, was considered useful as a drug delivery carrier due to its biodegradability, minimal immunogenicity, and possible receptor-mediated endocytosis [169]. Lifshiz Zimon et al. [170] assessed the benefits of ultrasound-assisted miR-197/Q-starch nanocomplexes for improving skin absorption as well as its anti-psoriatic activity. The ability of low-frequency ultrasound to enhance cell membrane permeability and skin delivery was elucidated previously [171]. The mean diameter of the nanocomplex was estimated to be 132 nm, with a zeta potential of 32 mV. The ultrasound-mediated delivery contributed to the entrance of the nanocomplex to the epidermis, including the basal cells. The in vivo efficacy of the ultrasound-mediated nanocomplex absorption was evaluated by the xenograft transplantation of human psoriasis skin to the mice. The pathological score data showed a reduction after the topical application of the nanocomplex in the presence of ultrasound. The epidermal hyperplasia was also restrained by the combined ultrasound and nanocarriers. The approaches for improving miRNA delivery into cells and skin are depicted in Table 5.

6. Conclusions

The regulation of miRNA expression is a promising and novel therapy for targeting skin inflammation diseases, such as psoriasis, AD, and cutaneous wounds. The exogenous administration of the anti-inflammatory miRNA mimic is beneficial for inhibiting proinflammatory mediators, leading to the alleviation of skin inflammation. miRNA-based anti-inflammatory therapy is also achieved by treatment with bioactive agents that can modulate the expression of miRNA. In terms of using miRNA treatment for skin diseases, local administration via the skin could be an efficient approach to achieve satisfactory availability. Topical delivery of miRNA usually has an incomplete response. This phenomenon is mainly caused by the barrier features of the skin and the target cells. Hence, the elaboration of delivery carriers that improve skin delivery and cell internalization is important. Considering the efficiency of skin penetration and controlled release, the introduction of nanocarriers could be a potential solution for topical application. Regarding future applications, effort should be paid to connecting the gap between laboratory investigations and clinical trials. Most studies on the anti-inflammatory activity of miRNA have been conducted using cell- and animal-based models, and there have been few clinical studies until now. The high cost of miRNA synthesis and its questionable stability may hinder the progress of its application. Although miRNA-based therapies have some limitations, future approaches aimed at treating cutaneous inflammation in a variety of skin diseases should be considered.

Author Contributions

Writing—original draft preparation, S.-C.Y., A.A. and Z.-C.L.; writing—review and editing, Y.-C.L., I.A.A. and J.-Y.F.; supervision, Y.-C.L. and J.-Y.F.; funding acquisition, J.-Y.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Ministry of Science and Technology of Taiwan (MOST-110-2330-B-182-011-MY3) and Chang Gung Memorial Hospital (CMRPD1M0101-3).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Slack, F.J.; Chinnaiyan, A.M. The role of non-coding RNAs in oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  2. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [Green Version]
  3. Friedman, R.C.; Farh, K.K.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ivey, K.N.; Srivastava, D. microRNAs as developmental regulators. Cold Spring Harb. Perspect. Biol. 2015, 7, a008144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Condrat, C.E.; Thompson, D.C.; Barbu, M.G.; Bugnar, O.L.; Boboc, A.; Cretoiu, D.; Suciu, N.; Cretoiu, S.M.; Voinea, S.C. miRNAs as biomarkers in disease: Latest findings regarding their role in diagnosis and prognosis. Cells 2020, 9, 276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Herrera, J.A.; Schwartz, M.A. microRNAs in mechanical homeostasis. Cold Spring Harb. Perspect. Med. 2022, 12, a041220. [Google Scholar] [CrossRef] [PubMed]
  7. Siddika, T.; Heinemann, I.U. Bringing microRNAs to light: Methods for microRNA quatification and visualization in live cells. Front. Bioeng. Biotechnol. 2021, 8, 619583. [Google Scholar] [CrossRef]
  8. Rupaimoole, R.; Slack, F.J. microRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017, 16, 203–222. [Google Scholar] [CrossRef] [PubMed]
  9. Rebane, A.; Akdis, C.A. MicroRNAs: Essential players in the regulation of inflammation. J. Allergy Clin. Immunol. 2013, 132, 15–26. [Google Scholar] [CrossRef]
  10. Duan, W.; Zhang, W.; Jia, J.; Lu, Q.; Eric Gershwin, M. Exosomal microRNA in autoimmunity. Cell. Mol. Immunol. 2019, 16, 932–934. [Google Scholar] [CrossRef] [PubMed]
  11. Wang, J.; Yue, B.L.; Huang, Y.Z.; Lan, X.Y.; Liu, W.J.; Chen, H. Exosomal RNAs: Novel potential biomarkers for diseases—A review. Int. J. Mol. Sci. 2022, 23, 2461. [Google Scholar] [CrossRef] [PubMed]
  12. Liang, L.; He, X. A narrative review of microRNA therapeutics: Understanding the future of microRNA research. Precis. Cancer Med. 2021, 4, 33. [Google Scholar] [CrossRef]
  13. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.S. Therapeutic advances of miRNAs: A preclinical and clinical update. J. Adv. Res. 2020, 28, 127–138. [Google Scholar] [CrossRef] [PubMed]
  14. Diener, C.; Keller, A.; Meese, E. Emerging concepts of miRNA therapeutics: From cells to clinic. Trends Genet. 2022, 38, 613–626. [Google Scholar] [CrossRef]
  15. Vijay, K. Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int. Immunopharmacol. 2018, 59, 391–412. [Google Scholar] [CrossRef] [PubMed]
  16. Nejad, C.; Stunden, H.J.; Gantier, M.P. A guide to miRNAs in inflammation and innate immune responses. FEBS J. 2018, 285, 3695–3716. [Google Scholar] [CrossRef] [PubMed]
  17. Tahamtan, A.; Teymoori-Rad, M.; Nakstad, B.; Salimi, V. Anti-inflammatory microRNAs and their potential for inflammatory diseases treatment. Front. Immunol. 2018, 9, 1377. [Google Scholar] [CrossRef] [Green Version]
  18. Lee, H.; Hong, Y.; Kim, M. Structural and functional changes and possible molecular mechanisms in aged skin. Int. J. Mol. Sci. 2021, 22, 12489. [Google Scholar] [CrossRef] [PubMed]
  19. Jiang, Y.; Xu, X.; Xiao, L.; Wang, L.; Qiang, S. The role of microRNA in the inflammatory response of wound healing. Front. Immunol. 2022, 13, 852419. [Google Scholar] [CrossRef] [PubMed]
  20. Chatzikyriakidou, A.; Voulgari, P.V.; Georgiou, I.; Drosos, A.A. The role of microRNA-146a (miR-146a) and its target IL-1R-associated kinase (IRAK1) in psoriatic arthritis susceptibility. Scand. J. Immunol. 2010, 71, 382–385. [Google Scholar] [CrossRef]
  21. Løvendorf, M.B.; Zibert, J.R.; Gyldenløve, M.; Røpke, M.A.; Skov, L. MicroRNA-223 and miR-143 are important systemic biomarkers for disease activity in psoriasis. J. Dermatol. Sci. 2014, 75, 133–139. [Google Scholar] [CrossRef] [PubMed]
  22. Tsuru, Y.; Jinnin, M.; Ichihara, A.; Fujisawa, A.; Moriya, C.; Sakai, K.; Fukushima, S.; Ihn, H. miR-424 levels in hair shaft are increased in psoriatic patients. J. Dermatol. 2014, 41, 382–385. [Google Scholar] [CrossRef] [PubMed]
  23. Lv, Y.; Qi, R.; Xu, J.; Di, Z.; Zheng, H.; Huo, W.; Zhang, L.; Chen, H.; Gao, X. Profiling of serum and urinary microRNAs in children with atopic dermatitis. PLoS ONE 2014, 9, e115448. [Google Scholar] [CrossRef]
  24. Meng, L.; Li, M.; Gao, Z.; Ren, H.; Chen, J.; Liu, X.; Cai, Q.; Jiang, L.; Ren, X.; Yu, Q.; et al. Possible role of hsa-miR-194-5p, via regulation of HS3ST2, in the pathogenesis of atopic dermatitis in children. Eur. J. Dermatol. 2019, 29, 603–613. [Google Scholar] [CrossRef] [PubMed]
  25. 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. [Google Scholar] [CrossRef] [PubMed]
  26. Li, X.; Ponandai-Srinivasan, S.; Nandakumar, K.S.; Fabre, S.; Xu Landén, N.; Mavon, A.; Khmaladze, I. Targeting microRNA for improved skin health. Health Sci. Rep. 2021, 4, e374. [Google Scholar] [CrossRef] [PubMed]
  27. Domingo, S.; Solé, C.; Moliné, T.; Ferrer, B.; Cortés-Hernández, J. MicroRNAs in several cutaneous autoimmune diseases: Psoriasis, cutaneous lupus erythematosus and atopic dermatitis. Cells 2020, 9, 2656. [Google Scholar] [CrossRef]
  28. Lee, S.W.L.; Paoletti, C.; Campisi, M.; Osaki, T.; Adriani, G.; Kamm, R.D.; Mattu, C.; Chiono, V. MicroRNA delivery through nanoparticles. J. Control. Release 2019, 313, 80–95. [Google Scholar] [CrossRef]
  29. Lu, Q.; Wu, R.; Zhao, M.; Garcia-Gomez, A.; Ballestar, E. miRNAs as therapeutic targets in inflammatory disease. Trends Pharmacol. Sci. 2019, 40, 853–865. [Google Scholar] [CrossRef] [PubMed]
  30. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.S. The interplay among miRNAs, major cytokines, and cancer-related inflammation. Mol. Ther. Nucleic Acids 2020, 20, 606–620. [Google Scholar] [CrossRef] [PubMed]
  31. Bayraktar, R.; Bertilaccio, M.T.S.; Calin, G.A. The interaction between two worlds: MicroRNAs and Toll-like receptors. Front. Immunol. 2019, 10, 1053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Specjalski, K.; Jassem, E. MicroRNAs: Potential biomarkers and targets of therapy in allergic diseases? Arch. Immunol. Ther. Exp. 2019, 67, 213–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Curtale, G.; Rubino, M.; Locati, M. MicroRNAs as molecular switches in macrophage activation. Front. Immunol. 2019, 10, 799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Rapalli, V.K.; Waghule, T.; Gorantla, S.; Dubey, S.K.; Saha, R.N.; Singhvi, G. Psoriasis: Pathological mechanisms, current pharmacological therapies, and emerging drug delivery systems. Drug Discov. Today 2020, 25, 2212–2226. [Google Scholar] [CrossRef]
  35. Afonina, I.S.; Van Nuffel, E.; Beyaert, R. Immune responses and therapeutic options in psoriasis. Cell Mol. Life Sci. 2021, 78, 2709–2727. [Google Scholar] [CrossRef] [PubMed]
  36. Huang, T.H.; Lin, C.F.; Alalaiwe, A.; Yang, S.C.; Fang, J.Y. Apoptotic or antiproliferative activity of natural products against keratinocytes for the treatment of psoriasis. Int. J. Mol. Sci. 2019, 20, 2558. [Google Scholar] [CrossRef] [Green Version]
  37. Shao, S.; Gudjonsson, J.E. Epigenetics of psoriasis. Adv. Exp. Med. Biol. 2020, 1253, 209–221. [Google Scholar]
  38. Sonkoly, E.; Wei, T.; Janson, P.C.; Sääf, A.; Lundeberg, L.; Tengvall-Linder, M.; Norstedt, G.; Alenius, H.; Homey, B.; Scheynius, A.; et al. MicroRNAs: Novel regulators involved in the pathogenesis of psoriasis? PLoS ONE 2007, 2, e610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Yang, X.; Wang, H. miRNAs flowing up and down: The concerto of psoriasis. Front. Med. 2021, 8, 646796. [Google Scholar]
  40. Timis, T.L.; Orasan, R.I. Understanding psoriasis: Role of miRNAs. Biomed. Rep. 2018, 9, 367–374. [Google Scholar] [PubMed] [Green Version]
  41. Raaby, L.; Langkilde, A.; Kjellerup, R.B.; Vinter, H.; Khatib, S.H.; Hjuler, K.F.; Johansen, C.; Iversen, L. Changes in mRNA expression precede changes in microRNA expression in lesional psoriatic skin during treatment with adalimumab. Br. J. Dermatol. 2015, 173, 436–447. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, M.J.; Xu, Y.Y.; Huang, R.Y.; Chen, X.M.; Chen, H.M.; Han, L.; Yan, Y.H.; Lu, C.J. Role of an imbalanced miRNAs axis in pathogenesis of psoriasis: Novel perspectives based on review of the literature. Oncotarget 2017, 8, 5498–5507. [Google Scholar] [CrossRef] [Green Version]
  43. Huang, R.Y.; Li, L.; Wang, M.J.; Chen, X.M.; Huang, Q.C.; Lu, C.J. An exploration of the role of microRNAs in psoriasis: A systematic review of the literature. Medicine 2015, 94, e2030. [Google Scholar] [CrossRef] [PubMed]
  44. Beer, L.; Kalinina, P.; Köcher, M.; Laggner, M.; Jeitler, M.; Abbas Zadeh, S.; Copic, D.; Tschachler, E.; Mildner, M. miR-155 contributes to normal keratinocyte differetiation and is upregulated in the epidermis of psoriatic skin lesions. Int. J. Mol. Sci. 2020, 21, 9288. [Google Scholar] [CrossRef] [PubMed]
  45. Nedoszytko, B.; Szczerkowska-Dobosz, A.; Stawczyk-Macieja, M.; Owczarczyk-Saczonek, A.; Reich, A.; Bartosiñska, J.; Batycka-Baran, A.; Czajkowski, R.; Dobrucki, I.T.; Dobrucki, L.W.; et al. Pathogenesis of psoriasis in the “omic” era. Part II. Genetic, genomic and epigenetic changes in psoriasis. Postepy Dermatol. Alergol. 2020, 37, 283–298. [Google Scholar] [CrossRef] [PubMed]
  46. Kasprzak, A. Angiogenesis-related functions of Wnt signaling in colorectal carcimogenesis. Cancers 2020, 12, 3601. [Google Scholar] [CrossRef] [PubMed]
  47. Shen, H.; Tian, Y.; Yao, X.; Liu, W.; Zhang, Y.; Yang, Z. MiR-99a inhibits keratinocyte proliferation by targeting Frizzled-5 (FZD5)/FZD8 through β-catenin signaling in psoriasis. Pharmazie 2017, 72, 461–467. [Google Scholar]
  48. Müller, M.B.; Hübner, M.; Li, L.; Tomasi, S.; Ließke, V.; Effinger, D.; Hirschberger, S.; Pogoda, K.; Sperandio, M.; Kreth, S. Cell-crossing functional network driven by microRNA-125a regulates endothelial permeability and monocyte trafficking in acute inflammation. Front. Immunol. 2022, 13, 826047. [Google Scholar] [CrossRef] [PubMed]
  49. Su, F.; Jin, L.; Liu, W. MicroRNA-125a correlates with decreased psoriasis severity and inflammation and represses keratinocyte proliferation. Dermatology 2021, 237, 568–578. [Google Scholar] [CrossRef] [PubMed]
  50. Paterson, M.R.; Kriegel, A.J. MiR-146a/b: A family with shared seeds and different roots. Physiol. Genom. 2017, 49, 243–252. [Google Scholar] [CrossRef] [PubMed]
  51. Srivastava, A.; Nikamo, P.; Lohcharoenkal, W.; Li, D.; Meisgen, F.; Xu Landén, N.; Ståhle, M.; Pivarcsi, A.; Sonkoly, E. MicroRNA-146a suppresses IL-17-mediated skin inflammation and is genetically associated with psoriasis. J. Allergy Clin. Immunol. 2017, 139, 550–561. [Google Scholar] [CrossRef] [Green Version]
  52. Gangwar, R.S.; Gudjonsson, J.E.; Ward, N.L. Mouse models of psoriasis: A comprehensive review. J. Investig. Dermatol. 2022, 142, 884–897. [Google Scholar] [CrossRef]
  53. Hermann, H.; Runnel, T.; Aab, A.; Baurecht, H.; Rodriguez, E.; Magilnick, N.; Urgard, E.; Šahmatova, L.; Prans, E.; Maslovskaja, J.; et al. miR-146b probably assists miRNA-146a in the suppression of keratinocyte proliferation and inflammatory responses in psoriasis. J. Investig. Dermatol. 2017, 137, 1945–1954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Brauze, D. SERPINB2–Its regulation and interplay with aryl hydrocarbon receptor. J. Appl. Genet. 2021, 62, 99–105. [Google Scholar] [CrossRef] [PubMed]
  55. Vaher, H.; Kivihall, A.; Runnel, T.; Raam, L.; Prans, E.; Maslovskaja, J.; Abram, K.; Kaldvee, B.; Mrowietz, U.; Weidinger, S.; et al. SERPINB2 and miR-146a/b are coordinately regulated and act in the suppression of psoriasis-associated inflammatory responses in keratinocytes. Exp. Dermatol. 2020, 29, 51–60. [Google Scholar] [CrossRef] [PubMed]
  56. Tang, L.; He, S.; Zhu, Y.; Feng, B.; Su, Z.; Liu, B.; Xu, F.; Wang, X.; Liu, H.; Li, C.; et al. Downregulated miR-187 contributes to the keratinocytes hyperproliferation in psoriasis. J. Cell. Physiol. 2019, 234, 3661–3674. [Google Scholar] [CrossRef]
  57. Flem-Karlsen, K.; Fodstad, Ø.; Tan, M.; Nunes-Xavier, C.E. B7-H3 in cancer–Beyond immune regulation. Trends Cancer 2018, 4, 401–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Huang, C.; Zhong, W.; Ren, X.; Huang, X.; Li, Z.; Chen, C.; Jiang, B.; Chen, Z.; Jian, X.; Yang, L.; et al. MiR-193b-3p-ERBB4 axis regulates psoriasis pathogenesis via modulating cellular proliferation and inflammatory-mediator production of keratinocytes. Cell Death Dis. 2021, 12, 963. [Google Scholar] [CrossRef] [PubMed]
  59. Wei, T.; Orfanidis, K.; Xu, N.; Janson, P.; Ståhle, M.; Pivarcsi, A.; Sonkoly, E. The expression of microRNA-203 during human skin morphogenesis. Exp. Dermatol. 2010, 19, 854–856. [Google Scholar] [CrossRef] [Green Version]
  60. Wang, M.; Wang, Y.; Zhang, M.; Duan, Q.; Chen, C.; Sun, Q.; Liu, M.; Zheng, Y.; Shao, Y. Kynureninase contributes to the pathogenesis of psoriasis through pro-inflammatory effect. J. Cell. Physiol. 2022, 237, 1044–1056. [Google Scholar] [CrossRef]
  61. Yu, Z.; Gong, Y.; Cui, L.; Hu, Y.; Zhou, Q.; Chen, Z.; Yu, Y.; Chen, Y.; Xu, P.; Zhang, X.; et al. High-throughput transcriptome and pathogenesis analysis of clinical psoriasis. J. Dermatol. Sci. 2020, 98, 109–118. [Google Scholar] [CrossRef] [PubMed]
  62. Zhao, J.; Wang, F.; Tian, Q.; Dong, J.; Chen, L.; Hu, R. Involvement of miR-214-3p/FOXM1 axis during the progression of psoriasis. Inflammation 2022, 45, 267–278. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, A.; Zhang, B.; Zhao, W.; Tu, Y.; Wang, Q.; Li, J. MicroRNA-215-5p inhibits the proliferation of keratinocytes and alleviates psoriasis-like inflammation by negatively regulating DYRK1A and its downstream signaling pathways. Exp. Dermatol. 2021, 30, 932–942. [Google Scholar] [CrossRef] [PubMed]
  64. Bian, J.; Liu, R.; Fan, T.; Liao, L.; Wang, S.; Geng, W.; Wang, T.; Shi, W.; Ruan, Q. miR-340 alleviates psoriasis in mice through direct targeting of IL-17A. J. Immunol. 2018, 201, 1412–1420. [Google Scholar] [CrossRef] [PubMed]
  65. Yi, Q.; Xie, W.; Sun, W.; Sun, W.; Liao, Y. A concise review of microRNA-383: Exploring the insights of its function in tumorigenesis. J. Cancer 2022, 13, 313–324. [Google Scholar] [CrossRef]
  66. Wang, H.; Xu, Y.; Jin, M.; Li, H.; Li, S. miR-383 reduces keratinocyte proliferation and induces the apoptosis in psoriasis via disruption of LCN2-dependent JAK/STAT pathway activation. Int. Immunopharmacol. 2021, 96, 107587. [Google Scholar] [CrossRef] [PubMed]
  67. Ye, Y.; Wang, P.; Zhou, F. miR-489-3p inhibits TLR4/NF-κB signaling to prevent inflammation in psoriasis. Exp. Ther. Med. 2021, 22, 744. [Google Scholar] [CrossRef]
  68. Nakahara, T.; Kido-Nakahara, M.; Tsuji, G.; Furue, M. Basics and recent advances in the pathophysiology of atopic dermatitis. J. Dermatol. 2021, 48, 130–139. [Google Scholar] [CrossRef]
  69. Renert-Yuval, Y.; Thyssen, J.P.; Bissonnette, R.; Bieber, T.; Kabashima, K.; Hijnen, D.; Guttman-Yassky, E. Biomarkers in atopic dermatitis–A review on behalf of the International Eczema Council. J. Allergy Clin. Immunol. 2021, 147, 1174–1190. [Google Scholar] [CrossRef]
  70. Yamanaka, K.I.; Mizutani, H. The role of cytokines/chemokines in the pathogenesis of atopic dermatitis. Curr. Probl. Dermatol. 2011, 41, 80–92. [Google Scholar] [PubMed]
  71. Brunner, P.M.; Silverberg, J.I.; Guttman-Yassky, E.; Paller, A.S.; Kabashima, K.; Amagai, M.; Luger, T.A.; Deleuran, M.; Werfel, T.; Eyerich, K.; et al. Increasing comorbidities suggest that atopic dermatitis is a systemic disorder. J. Investig. Dermatol. 2017, 137, 18–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Nygaard, U.; Deleuran, M.; Vestergaard, C. Emerging treatment options in atopic dermatitis: Topical therapies. Dermatology 2017, 233, 333–343. [Google Scholar] [CrossRef] [PubMed]
  73. 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]
  74. Gu, C.; Li, Y.; Wu, J.; Xu, J. IFN-γ-induced microRNA-29b up-regulation contributes to keratinocyte apoptosis in atopic dermatitis through inhibiting Bcl2L2. Int. J. Clin. Exp. Pathol. 2017, 10, 10117–10126. [Google Scholar]
  75. 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]
  76. Chikh, A.; Matin, R.N.; Senatore, V.; Hufbauer, M.; Lavery, D.; Raimondi, C.; Ostano, P.; Mello-Grand, M.; Ghimenti, C.; Bahta, A.; et al. iASPP/p63 autoregulatory feedback loop is required for the homeostasis of stratified epithelia. EMBO J. 2011, 30, 4261–4273. [Google Scholar] [CrossRef] [Green Version]
  77. Nousbeck, J.; McAleer, M.A.; Hurault, G.; Kenny, E.; Harte, K.; Kezic, S.; Tanaka, R.J.; Irvine, A.D. MicroRNA analysis of childhood atopic dermatitis reveals a role for miR-451a. Br. J. Dermatol. 2021, 184, 514–523. [Google Scholar] [CrossRef] [PubMed]
  78. Maeno, M.; Tamagawa-Mineoka, R.; Arakawa, Y.; Nishigaki, H.; Yasuike, R.; Masuda, K.; Katoh, N. Increased plasma miR-24 and miR-191 levels in patients with severe atopic dermatitis: Possible involvement of platelet activation. Clin. Immunol. 2022, 237, 108983. [Google Scholar] [CrossRef]
  79. Yoon, W.S.; Ryu, S.R.; Lee, S.S.; Chae, Y.S.; Kim, E.J.; Choi, J.H.; Oh, S.; Park, S.H.; Choung, J.T.; Yoo, Y.; et al. Suppression of inflammation by recombinant Salmonella typhimurium harboring CCL22 microRNA. DNA Cell Biol. 2012, 31, 290–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. 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] [PubMed]
  81. 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] [PubMed]
  82. Teng, Y.; Zhang, R.; Liu, C.; Zhou, L.; Wang, H.; Zhuang, W.; Huang, Y.; Hong, Z. miR-143 inhibits interleukin-13-induced inflammatory cytokine and mucus production in nasal epithelial cells from allergic rhinitis patients by targeting IL13Rα1. Biochem. Biophys. Res. Commun. 2015, 457, 58–64. [Google Scholar] [CrossRef]
  83. 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]
  84. Yan, F.; Meng, W.; Ye, S.; Zhang, X.; Mo, X.; Liu, J.; Chen, D.; Lin, Y. MicroRNA-146a as a potential regulator involved in the pathogenesis of atopic dermatitis. Mol. Med. Rep. 2019, 20, 4645–4653. [Google Scholar] [CrossRef] [PubMed]
  85. Meisgen, F.; Xu Landén, N.; Wang, A.; Réthi, B.; Bouez, C.; Zuccolo, M.; Gueniche, A.; Ståhle, M.; Sonkoly, E.; Breton, L.; et al. MiR-146a negatively regulates TLR2-induced inflammatory responses in keratinocytes. J. Investig. Dermatol. 2014, 134, 1931–1940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. 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, 134, 836–847. [Google Scholar] [CrossRef]
  87. Roedl, D.; Traidl-Hoffmann, C.; Ring, J.; Behrendt, H.; Braun-Falco, M. Serine protease inhibitor lymphoepithelial Kazal type-related inhibitor tends to be decreased in atopic dermatitis. J. Eur. Acad. Dermatol. Venereol. 2009, 23, 1263–1266. [Google Scholar] [CrossRef] [PubMed]
  88. D’Addario, I.; Abbruzzese, C.; Lo Iacono, M.; Teson, M.; Golisano, O.; Barone, V. Overexpression of YAP1 induces immortalization of normal human keratinocytes by blocking clonal evolution. Histochem. Cell. Biol. 2010, 134, 265–276. [Google Scholar] [CrossRef]
  89. 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]
  90. Chen, Y.; Jiang, Y.; Wang, L.; Yang, C.; Qian, Q.; Mao, J.; Sun, D.; Zhu, T. MiR-1294 suppresses ROS-dependent inflammatory response in atopic dermatitis via restraining STAT3/NF-κB pathway. Cell. Immunol. 2022, 371, 104452. [Google Scholar]
  91. Sorg, H.; Tilkorn, D.J.; Hager, S.; Hauser, J.; Mirastschijski, U. Skin wound healing: An update on the current knowledge and concepts. Eur. Surg. Res. 2017, 58, 81–94. [Google Scholar] [CrossRef] [PubMed]
  92. Wilkinson, H.N.; Hardman, M.J. Wound healing: Cellular mechanisms and pathological outcomes. Open Biol. 2020, 10, 200223. [Google Scholar] [CrossRef] [PubMed]
  93. Ross, K. MiR equal than others: MicroRNA enhancement for cutaneous wound healing. J. Cell. Physiol. 2021, 236, 8050–8059. [Google Scholar] [CrossRef] [PubMed]
  94. Mori, R.; Tanaka, K.; Shimokawa, I. Identification and functional analysis of inflammation-related miRNAs in skin wound repair. Dev. Growth Differ. 2018, 60, 306–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Piipponen, M.; Li, D.; Landén, N.X. The immune functions of keratinocytes in skin wound healing. Int. J. Mol. Sci. 2020, 21, 8790. [Google Scholar] [CrossRef]
  96. Ross, K. Towards topical microRNA-directed therapy for epidermal disorders. J. Control. Release 2018, 269, 136–147. [Google Scholar] [CrossRef] [Green Version]
  97. Phillipson, M.; Kubes, P. The healing power of neutrophils. Trends Immunol. 2019, 40, 635–647. [Google Scholar] [CrossRef] [PubMed]
  98. Tanaka, K.; Kim, S.E.; Yano, H.; Matsumoto, G.; Ohuchida, R.; Ishikura, Y.; Araki, M.; Araki, K.; Park, S.; Komatsu, T.; et al. MiR-142 is required for Staphylococcus aureus clearance at skin wound sites via small GTPase-mediated regulation of the neutrophil actin cytoskeleton. J. Investig. Dermatol. 2017, 137, 931–940. [Google Scholar] [CrossRef]
  99. Self-Fordham, J.B.; Naqvi, A.R.; Uttamani, J.R.; Kulkarni, V.; Nares, S. MicroRNA: Dynamic regulators of macrophage polarization and plasticity. Front. Immunol. 2017, 8, 1062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Liechty, C.; Hu, J.; Zhang, L.; Liechty, K.W.; Xu, J. Role of microRNA-21 and its underlying mechanisms in inflammatory responses in diabetic wounds. Int. J. Mol. Sci. 2020, 21, 3328. [Google Scholar] [CrossRef] [PubMed]
  101. Gallant-Behm, C.L.; Piper, J.; Lynch, J.M.; Seto, A.G.; Hong, S.J.; Mustoe, T.A.; Maari, C.; Pestano, L.A.; Dalby, C.M.; Jackson, A.L.; et al. A microRNA-29 mimic (Remlarsen) represses extracellular matrix expression and fibroplasia in the skin. J. Investig. Dermatol. 2019, 139, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
  102. Li, H.; Han, X.; Zuo, K.; Li, L.; Liu, J.; Yuan, X.; Shen, Y.; Shao, M.; Pang, D.; Chu, Y.; et al. miR-23b promotes cutaneous wound healing through inhibition of the inflammatory responses by targeting ASK1. Acta Biochim. Biophys. Sin. 2018, 50, 1104–1113. [Google Scholar] [CrossRef] [PubMed]
  103. Tai, Y.; Woods, E.L.; Dally, J.; Kong, D.; Steadman, R.; Moseley, R.; Midgley, A.C. Myofibroblasts: Function, formation, and scope of molecular therapies for skin fibrosis. Biomolecules 2021, 11, 1095. [Google Scholar] [CrossRef]
  104. Li, D.; Li, X.I.; Wang, A.; Meisgen, F.; Pivarcsi, A.; Sonkoly, E.; Ståhle, M.; Landén, N.X. MicroRNA-31 promotes skin wound healing by enhancing keratinocyte proliferation and migration. J. Investig. Dermatol. 2015, 135, 1676–1685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Hart, M.; Walch-Rückheim, B.; Krammes, L.; Kehl, T.; Rheinheimer, S.; Tänzer, T.; Glombitza, B.; Sester, M.; Lenhof, H.P.; Keller, A.; et al. miR-34a as hub of T cell regulation networks. J. Immunother. Cancer 2019, 7, 187. [Google Scholar]
  106. Zhao, N.; Wang, G.; Long, S.; Hu, M.; Gao, J.; Ran, X.; Wang, J.; Su, Y.; Wang, T. MicroRNA-34a deficiency leads to impaired wound closure by augmented inflammation in mice. Ann. Transl. Med. 2020, 8, 447. [Google Scholar] [CrossRef] [PubMed]
  107. Ban, E.; Jeong, S.; Park, M.; Kwon, H.; Park, J.; Song, E.J.; Kim, A. Accelerated wound healing in diabetic mice by miRNA-497 and its anti-inflammatory activity. Biomed. Pharmacother. 2020, 121, 109613. [Google Scholar] [CrossRef]
  108. Minejima, E.; Bensman, J.; She, R.C.; Mack, W.J.; Tuan Tran, M.; Ny, P.; Lou, M.; Yamaki, J.; Nieberg, P.; Ho, J.; et al. A dysregulated balance of proinflammatory and anti-inflammatory host cytokine response early during therapy predicts persistence and mortality in Staphylococcus aureus bacteremia. Crit. Care Med. 2016, 44, 671–679. [Google Scholar] [CrossRef] [PubMed]
  109. Yang, Y.; Yang, C.; Guo, Y.F.; Liu, P.; Guo, S.; Yang, J.; Zahoor, A.; Shaukat, A.; Deng, G. MiR-142a-3p alleviates Escherichia coli derived lipopolysaccharide-induced acute lung injury by targeting TAB2. Microb. Pathog. 2019, 136, 103721. [Google Scholar] [CrossRef] [PubMed]
  110. Platsidaki, E.; Dessinioti, C. Recent advances in understanding Propionibacterium acnes (Cutibacterium acnes) in acne. F1000Research 2018, 7, 1953. [Google Scholar] [CrossRef] [Green Version]
  111. Zeng, R.; Xu, H.; Liu, Y.; Du, L.; Duan, Z.; Tong, J.; He, Y.; Chen, Q.; Chen, X.; Li, M. miR-146a inhibits biofilm-derived Cutibacterium acnes-induced inflammatory reactions in human keratinocytes. J. Investig. Dermatol. 2019, 139, 2488–2496. [Google Scholar] [CrossRef]
  112. Mata-Martínez, P.; Bergón-Gutiérrez, M.; Del Fresno, C. Dectin-1 signaling update: New perspectives for trained immunity. Front. Immunol. 2022, 13, 812148. [Google Scholar] [CrossRef]
  113. Du, L.; Chen, X.; Duan, Z.; Liu, C.; Zeng, R.; Chen, Q.; Li, M. MiR-146a negatively regulates dectin-1-induced inflammatory responses. Oncotarget 2017, 8, 37355–37366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Achtman, J.C.; Werth, V.P. Pathophysiology of cutaneous lupus erythematosus. Arthritis Res. Ther. 2015, 17, 182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Huang, J.; Xu, X.; Wang, X.; Yang, J.; Xue, M.; Yang, Y.; Zhang, R.; Yang, X.; Yang, J. MicroRNA-590-3p inhibits T helper 17 cells and ameliorates inflammation in lupus mice. Immunology 2022, 165, 260–273. [Google Scholar] [CrossRef]
  116. Lin, C.E.; Kaptein, J.S.; Sheikh, J. Differential expression of microRNAs and their possible roles in patients with chronic idiopathic urticaria and active hives. Allergy Rhinol. 2017, 8, 67–80. [Google Scholar] [CrossRef] [Green Version]
  117. Qu, S.; Yang, L.; Liu, Z. MicroRNA-194 reduces inflammatory response and human dermal microvascular endothelial cells permeability through suppression of TGF-β/SMAD pathway by inhibiting THBS1 in chronic idiopathic urticaria. J. Cell. Biochem. 2020, 121, 111–124. [Google Scholar] [CrossRef]
  118. Zhou, H.; Ni, W.J.; Meng, X.M.; Tang, L.Q. MicroRNAs as regulators of immune and inflammatory responses: Potential therapeutic targets in diabetic nephropathy. Front. Cell Dev. Biol. 2021, 8, 618536. [Google Scholar] [CrossRef]
  119. Cione, E.; La Torre, C.; Cannataro, R.; Caroleo, M.C.; Plastina, P.; Gallelli, L. Quercetin, epigallocatechin gallate, curcumin, and resveratrol: From dietary sources to human microRNA modulation. Molecules 2019, 25, 63. [Google Scholar] [CrossRef] [Green Version]
  120. Lin, M.H.; Hung, C.F.; Sung, H.C.; Yang, S.C.; Yu, H.P.; Fang, J.Y. The bioactivities of resveratrol and its naturally occurring derivatives on skin. J. Food Drug Anal. 2021, 29, 15–38. [Google Scholar] [CrossRef]
  121. Wang, X.; Zhang, Y. Resveratrol alleviates LPS-induced injury in human keratinocyte cell line HaCaT up-regulation of miR-17. Biochem. Biophys. Res. Commun. 2018, 501, 106–112. [Google Scholar] [CrossRef] [PubMed]
  122. Yamauchi, P.S.; Bissonnette, R.; Teixeira, H.D.; Valdecantos, W.C. Systematic review of efficacy of anti-tumor necrosis factor (TNF) therapy in patients with psoriasis previously treated with a different anti-TNF agent. J. Am. Acad. Dermatol. 2016, 75, 612–618. [Google Scholar] [CrossRef] [Green Version]
  123. Mensà, E.; Recchioni, R.; Marcheselli, F.; Giuliodori, K.; Consales, V.; Molinelli, E.; Prattichizzo, F.; Rippo, M.R.; Campanati, A.; Procopio, A.D.; et al. MiR-146a-5p correlates with clinical efficacy in patients with psoriasis treated with the tumour necrosis factor-alpha inhibitor adalimumab. Br. J. Dermatol. 2018, 179, 787–789. [Google Scholar] [CrossRef] [PubMed]
  124. Guo, W.; Xu, F.; Zhuang, Z.; Liu, Z.; Xie, J.; Bai, L. Ebosin ameliorates psoriasis-like inflammation of mice via miR-155 targeting tnfaip3 on IL-17 pathway. Front. Immunol. 2021, 12, 662362. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, X.; Zhang, Y.; Zhou, S.; Dain, L.; Mei, L.; Zhu, G. Circular RNA: An emerging frontier in RNA therapeutic targets, RNA therapeutics, and mRNA vaccines. J. Control. Release 2022, 348, 84–94. [Google Scholar] [CrossRef]
  126. Moldovan, L.I.; Tsoi, L.C.; Ranjitha, U.; Hager, H.; Weidinger, S.; Gudjonsson, J.E.; Kjems, J.; Kristensen, L.S. Characterization of circular RNA transcriptomes in psoriasis and atopic dermatitis reveals disease-specific expression profiles. Exp. Dermatol. 2021, 30, 1187–1196. [Google Scholar] [CrossRef]
  127. Lu, J.; Xu, X.; Li, Y.; Yu, N.; Ding, Y.; Shi, Y. CircRAB3B suppresses proliferation, motility, cell cycle progression and promotes the apoptosis of IL-22-induced keratinocytes depending on the regulation of miR-1228-3p/PTEN axis in psoriasis. Autoimmunity 2021, 54, 303–312. [Google Scholar] [CrossRef] [PubMed]
  128. Chowdhari, S.; Saini, N. hsa-miR-4516 mediated downregulation of STAT3/CDK6/UBE2N plays a role in PUVA induced apoptosis in keratinocytes. J. Cell. Physiol. 2014, 229, 1630–1638. [Google Scholar] [CrossRef]
  129. Gaba, S.; Saini, A.; Singh, G.; Monga, V. An insight into the medicinal attributes of berberine derivatives: A review. Bioorg. Med. Chem. 2021, 38, 116143. [Google Scholar] [CrossRef] [PubMed]
  130. Li, W.; Liu, F.; Wang, J.; Long, M.; Wang, Z. MicroRNA-21-mediated inhibition of mast cell degranulation involved in the protective effect of berberine on 2,4-dinitrofluorobenzene-induced allergic contact dermatitis in rats via p38 pathway. Inflammation 2018, 41, 689–699. [Google Scholar] [CrossRef]
  131. Chen, N.; Wang, J.; Mucelli, A.; Zhang, X.; Wang, C. Electro-acupuncture is beneficial for knee osteoarthritis: The evidence from meta-analysis of randomized controlled trials. Am. J. Chin. Med. 2017, 45, 965–985. [Google Scholar] [CrossRef]
  132. Wang, Z.; Yi, T.; Long, M.; Ding, F.; Ouyang, L.; Chen, Z. Involvement of the negative feedback of IL-33 signaling in the anti-inflammatory effect of electro-acupuncture on allergic contact dermatitis via targeting microRNA-155 in mast cells. Inflammation 2018, 41, 859–869. [Google Scholar] [CrossRef]
  133. Lee, Y.S.; Lee, C.H.; Bae, J.T.; Nam, K.T.; Moon, D.B.; Hwang, O.K.; Choi, J.S.; Kim, T.H.; Jun, H.O.; Jung, Y.S.; et al. Inhibition of skin carcinogenesis by suppression of NF-κB dependent ITGAV and TIMP-1 expression in IL-32γ overexpressed condition. J. Exp. Clin. Cancer Res. 2018, 37, 293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lee, Y.S.; Han, S.B.; Ham, H.J.; Park, J.H.; Lee, J.S.; Hwang, D.Y.; Jung, Y.S.; Yoon, D.Y.; Hong, J.T. IL-32γ suppressed atopic dermatitis through inhibition of miR-205 expression via inactivation of nuclear factor-kappa B. J. Allergy Clin. Immunol. 2020, 146, 156–168. [Google Scholar] [CrossRef] [PubMed]
  135. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Zhang, M.; Xu, C.; Liu, D.; Han, M.K.; Wang, L.; Merlin, D. Oral delivery of nanoparticles loaded with ginger active compound, 6-shogaol, attenuates ulcerative colitis and promotes wound healing in a murine model of ulcerative colitis. J. Crohns Colitis 2018, 12, 217–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Al-Rawaf, H.A.; Gabr, S.A.; Alghadir, A.H. Molecular changes in diabetic wound healing following administration of vitamin D and ginger supplements: Biochemical and molecular experimental study. Evid. Based Complement. Alternat. Med. 2019, 2019, 4352470. [Google Scholar] [CrossRef] [Green Version]
  138. Zhao, P.; Sui, B.D.; Liu, N.; Lv, Y.J.; Zheng, C.X.; Lu, Y.B.; Huang, W.T.; Zhou, C.H.; Chen, J.; Pang, D.L.; et al. Anti-aging pharmacology in cutaneous wound healing: Effects of metformin, resveratrol, and rapamycin by local application. Aging Cell 2017, 16, 1083–1093. [Google Scholar] [CrossRef] [PubMed]
  139. Hu, J.; Liu, X.; Chi, J.; Che, K.; Ma, X.; Qiu, M.; Fu, Z.; Wang, Y.; Wang, Y.; Wang, W. Resveratrol enhances wound healing in type 1 diabetes mellitus by promoting the expression of extracellular vesicle-carried microRNA-129 derived from mesenchymal stem cells. J. Proteome Res. 2022, 21, 313–324. [Google Scholar] [CrossRef] [PubMed]
  140. Li, Y.; Zhao, S.; der Merwe, L.V.; Dai, W.; Lin, C. Efficacy of curcumin for wound repair in diabetic rats/mice: A systematic review and meta-analysis of preclinical studies. Curr. Pharm. Des. 2022, 28, 187–197. [Google Scholar] [CrossRef] [PubMed]
  141. Huang, J.; Fu, J.; Liu, B.; Wang, R.; You, T. A Synthetic curcuminoid analog, (2E,6E)-2,6-bis(2-(trifluoromethyl)benzylidene)cyclohexanone, ameliorates impaired wound healing in streptozotocin-induced diabetic mice by increasing miR-146a. Molecules 2020, 25, 920. [Google Scholar] [CrossRef] [Green Version]
  142. Mo, F.F.; An, T.; Zhang, Z.J.; Liu, Y.F.; Liu, H.X.; Pan, Y.Y.; Miao, J.N.; Zhao, D.D.; Yang, X.Y.; Zhang, D.W.; et al. Jiang Tang Xiao Ke granule play an anti-diabetic role in diabetic mice pancreatic tissue by regulating the mRNAs and microRNAs associated with PI3K-Akt signaling pathway. Front. Pharmacol. 2017, 8, 795. [Google Scholar] [CrossRef] [Green Version]
  143. Zhang, W.; Qu, X.; Zhu, Z.; Wang, L.; Qi, Q.; Zhou, P.; Wang, X.; Li, W. Inhibition of miR-139-5p by topical JTXK gel promotes healing of Staphylococcus aureus-infected skin wounds. Cells Dev. 2021, 166, 203658. [Google Scholar] [CrossRef]
  144. Xia, X.; Li, Z.; Liu, K.; Wu, Y.; Jiang, D.; Lai, Y. Staphylococcal LTA-induced miR-143 inhibits Propionibacterium acnes-mediated inflammatory response in skin. J. Investig. Dermatol. 2016, 136, 621–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Lee, K.S.; Cha, H.J.; Lee, G.T.; Lee, K.K.; Hong, J.T.; Ahn, K.J.; An, I.S.; An, S.; Bae, S. Troxerutin induces protective effects against ultraviolet B radiation through the alteration of microRNA expression in human HaCaT keratinocyte cells. Int. J. Mol. Med. 2014, 33, 934–942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Zamanian, M.; Bazmandegan, G.; Sureda, A.; Sobarzo-Sanchez, E.; Yousefi-Manesh, H.; Shirooie, S. The protective roles and molecular mechanisms of troxerutin (vitamin P4) for the treatment of chronic diseases: A mechanistic review. Curr. Neuropharmacol. 2021, 19, 97–110. [Google Scholar] [CrossRef] [PubMed]
  147. Zakrewsky, M.; Mitragotri, S. Therapeutic RNAi robed with ionic liquid moieties as a simple, scalable prodrug platform for treating skin disease. J. Control. Release 2016, 242, 80–88. [Google Scholar] [CrossRef] [Green Version]
  148. Kapadia, C.H.; Luo, B.; Dang, M.N.; Irvin-Choy, N.; Valcourt, D.M.; Day, E.S. Polymer nanocarriers for MicroRNA delivery. J. Appl. Polym. Sci. 2020, 137, 48651. [Google Scholar] [CrossRef] [PubMed]
  149. Laanesoo, A.; Periyasamy, K.; Pooga, M.; Rebane, A. Development of CPP-based methods for delivery of miRNAs into the skin and airways: Lessons from cell culture and mouse models. Methods Mol. Biol. 2022, 2383, 515–528. [Google Scholar]
  150. Taylor, R.E.; Zahid, M. Cell penetrating peptides, novel vectors for gene therapy. Pharmaceutics 2020, 12, 225. [Google Scholar] [CrossRef] [Green Version]
  151. Wang, K.; Zhao, X.; Yang, F.; Liu, P.; Xing, J. Percutaneous delivery application of acylated steric acid-9-P(arginine) cell penetrating peptides used as transdermal penetration enhancer. J. Biomed. Nanotechnol. 2019, 15, 417–430. [Google Scholar] [CrossRef]
  152. Urgard, E.; Lorents, A.; Klaas, M.; Padari, K.; Viil, J.; Runnel, T.; Langel, K.; Kingo, K.; Tkaczyk, E.; Langel, Ü.; et al. Pre-administration of PepFect6-microRNA-146a nanocomplexes inhibits inflammatory responses in keratinocytes and in a mouse model of irritant contact dermatitis. J. Control. Release 2016, 235, 195–204. [Google Scholar] [CrossRef]
  153. Mulholland, E.J.; McErlean, E.M.; Dunne, N.; McCarthy, H.O. A peptide/microRNA-31 nanomedicine within an electrospun biomaterial designed to regenerate wounds in vivo. Acta Biomater. 2022, 138, 285–300. [Google Scholar]
  154. McErlean, E.M.; Ziminska, M.; McCrudden, C.M.; McBride, J.W.; Loughran, S.P.; Cole, G.; Mulholland, E.J.; Kett, V.; Buckley, N.E.; Robson, T.; et al. Rational design and characterization of a linear cell penetrating peptide for non-viral gene delivery. J. Control. Release 2021, 330, 1288–1299. [Google Scholar] [CrossRef]
  155. Behere, I.; Ingavle, G. In vitro and in vivo advancement of multifunctional electrospun nanofiber scaffolds in wound healing applications: Innovative nanofiber designs, stem cell approaches, and future perspectives. J. Biomed. Mater. Res. A 2022, 110, 443–461. [Google Scholar] [CrossRef]
  156. Zgheib, C.; Hilton, S.A.; Dewberry, L.C.; Hodges, M.M.; Ghatak, S.; Xu, J.; Singh, S.; Roy, S.; Sen, C.K.; Seal, S.; et al. Use of cerium oxide nanoparticles conjugated with microRNA-146a to correct the diabetic wound healing impairment. J. Am. Coll. Surg. 2019, 228, 107–115. [Google Scholar] [CrossRef]
  157. Augustine, R.; Hasan, A.; Patan, N.K.; Dalvi, Y.B.; Varghese, R.; Antony, A.; Unni, R.N.; Sandhyarani, N.; Moustafa, A.A. Cerium oxide oanoparticle incorporated electrospun poly(3-hydroxybutyrate-co-3-hydroxyvalerate) membranes for diabetic wound healing applications. ACS Biomater. Sci. Eng. 2020, 6, 58–70. [Google Scholar] [CrossRef]
  158. Niemiec, S.M.; Louiselle, A.E.; Hilton, S.A.; Dewberry, L.C.; Zhang, L.; Azeltine, M.; Xu, J.; Singh, S.; Sakthivel, T.S.; Seal, S.; et al. Nanosilk increases the strength of diabetic skin and delivers CNP-miR146a to improve wound healing. Front. Immunol. 2020, 11, 590285. [Google Scholar] [CrossRef]
  159. Lehmann, T.; Vaughn, A.E.; Seal, S.; Liechty, K.W.; Zgheib, C. Silk fibroin-based therapeutics for impaired wound healing. Pharmaceutics 2022, 14, 651. [Google Scholar] [CrossRef]
  160. Moon, H.H.; Joo, M.K.; Mok, H.; Lee, M.; Hwang, K.C.; Kim, S.W.; Jeong, J.H.; Choi, D.; Kim, S.H. MSC-based VEGF gene therapy in rat myocardial infarction model model using facial amphipathic bile acid-conjugated polyethyleneimine. Biomaterials 2014, 35, 1744–1754. [Google Scholar] [CrossRef]
  161. Wang, S.Y.; Kim, H.; Kwak, G.; Jo, S.D.; Cho, D.; Yang, Y.; Kwon, I.C.; Jeong, J.H.; Kim, S.H. Development of microRNA-21 mimic nanocarriers for the treatment of cutaneous wounds. Theranostics 2020, 10, 3240–3253. [Google Scholar] [CrossRef]
  162. Saleh, B.; Dhaliwal, H.K.; Portillo-Lara, R.; Shirzaei Sani, E.; Abdi, R.; Amiji, M.M.; Annabi, N. Local immunomodulation using an adhesive hydrogel loaded with miRNA-laden nanoparticles promotes wound healing. Small 2019, 15, e1902232. [Google Scholar] [CrossRef]
  163. Sakr, M.A.; Sakthivel, K.; Hossain, T.; Shin, S.R.; Siddiqua, S.; Kim, J.; Kim, K. Recent trens in gelatin methacryloyl nanocomposite hydrogels for tissue engineering. J. Biomed. Mater. Res. A 2022, 110, 708–724. [Google Scholar] [CrossRef]
  164. Feng, H.; Wu, R.; Zhang, S.; Kong, Y.; Liu, Z.; Wu, H.; Wang, H.; Su, Y.; Zhao, M.; Lu, Q. Topical administration of nanocarrier miRNA-210 antisense ameliorates imiquimod-induced psoriasis-like dermatitis in mice. J. Dermatol. 2020, 47, 147–154. [Google Scholar] [CrossRef] [PubMed]
  165. Ratajczak, M.Z.; Ratajczak, J. Extracellular microvesicles/exosomes: Discovery, disbelief, acceptance, and the future? Leukemia 2020, 34, 3126–3135. [Google Scholar] [CrossRef]
  166. Massey, A.E.; Malik, S.; Sikander, M.; Doxtater, K.A.; Tripathi, M.K.; Khan, S.; Yallapu, M.M.; Jaggi, M.; Chauhan, S.C.; Hafeez, B.B. Clinical implications of exosomes: Targeted drug delivery for cancer treatment. Int. J. Mol. Sci. 2021, 22, 5278. [Google Scholar] [CrossRef]
  167. Xia, W.; Li, M.; Jiang, X.; Huang, X.; Gu, S.; Ye, J.; Zhu, L.; Hou, M.; Zan, T. Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J. Nanobiotechnol. 2022, 20, 144. [Google Scholar] [CrossRef]
  168. Amar-Lewis, E.; Azagury, A.; Chintakunta, R.; Goldbart, R.; Traitel, T.; Prestwood, J.; Landesman-Milo, D.; Peer, D.; Kost, J. Quaternized starch-based carrier for siRNA delivery: From cellular uptake to gene silencing. J. Control. Release 2014, 185, 109–120. [Google Scholar] [CrossRef]
  169. Li, L.; He, S.; Yu, L.; Elshazly, E.H.; Wang, H.; Chen, K.; Zhang, S.; Ke, L.; Gong, R. Codelivery of DOX and siRNA by folate-biotin-quaternized starch nanoparticles for promoting synergistic suppression of human lung cancer cells. Drug Deliv. 2019, 26, 499–508. [Google Scholar] [CrossRef]
  170. Lifshiz Zimon, R.; Lerman, G.; Elharrar, E.; Meningher, T.; Barzilai, A.; Masalha, M.; Chintakunta, R.; Hollander, E.; Goldbart, R.; Traitel, T.; et al. Ultrasound targeting of Q-starch/miR-197 complexes for topical treatment of psoriasis. J. Control. Release 2018, 284, 103–111. [Google Scholar] [CrossRef]
  171. Polat, B.E.; Hart, D.; Langer, R.; Blankschtein, D. Ultrasound-mediated transdermal drug delivery: Mechanisms, scope, and emerging trends. J. Control. Release 2011, 152, 330–348. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The biogenesis and gene expression of miRNA in cells.
Figure 1. The biogenesis and gene expression of miRNA in cells.
Biomolecules 12 01072 g001
Figure 2. The function of miRNA in the regulation of innate or adaptive immune responses in different cells.
Figure 2. The function of miRNA in the regulation of innate or adaptive immune responses in different cells.
Biomolecules 12 01072 g002
Figure 3. The role of miRNA in the regulation of T cell differentiation and cytokine production in psoriasis.
Figure 3. The role of miRNA in the regulation of T cell differentiation and cytokine production in psoriasis.
Biomolecules 12 01072 g003
Figure 4. The dysregulation of miRNAs involved in atopic dermatitis and their effect on pathogenesis.
Figure 4. The dysregulation of miRNAs involved in atopic dermatitis and their effect on pathogenesis.
Biomolecules 12 01072 g004
Table 1. The targets and biological mechanisms of anti-inflammatory miRNAs for treating psoriasis.
Table 1. The targets and biological mechanisms of anti-inflammatory miRNAs for treating psoriasis.
miRNA CodeTargetsExperimental ModelsOutcomeReference
miR-99aFZD5 and FZD8HaCaT cells and patientsKeratinocyte proliferation inhibition through β-catenin signalingShen et al. [47]
miR-125aCAMK4HaCaT cells and patientsKeratinocyte proliferation inhibition and apoptosis enhancementSu et al. [49]
miR-146aCARD10, FERMT1, IRAK1 and TRAF6miR-146a−/− and wild-type mice and patientsInhibited psoriasiform inflammation, hyperplasia, and neutrophil infiltrationSrivastava et al. [51]
miR-146bCARD10, FERMT1, IRAK1 and TRAF6Normal human epidermal keratinocytes and miR-146a−/− or miR-146b−/− miceModulation of inflammatory response and keratinocyte proliferationHermann et al. [53]
miR-146a/bCARD10, FERMT1, IRAK1 and TRAF6Normal human epidermal keratinocytesSERPINB2 is coordinately regulated in the psoriatic inflammation with miR-146a/bVaher et al. [55]
miR-187CD276HaCaT, wild-type mice, and patientsInhibition of keratinocyte hyperproliferationTang et al. [56]
miR-193b-3pERBB4HaCaT and wild-type miceBlockade of psoriasis-like inflammation through NF-κB/STAT3 signalingHuang et al. [58]
miR-203aKynureninaseHaCaT and wild-type miceReduction of IL-1β in cytokine-activated keratinocytesWang et al. [60]
miR-214-3pFOXM1HaCaT, wild-type mice, and patientsInhibition of keratinocyte hyperproliferation and psoriasiform inflammationZhao et al. [62]
miR-215-5pDYRK1AHaCaT and wild-type miceSuppression of proliferation and cell cycle progression of keratinocytesLiu et al. [63]
miR-340IL-17A293T cells and wild-type miceReduction of psoriasiform symptomsBian et al. [64]
miR-383LCN2Cells from the skin of imiquimod-treated ratsReduced cell proliferation and increased cell apoptosisWang et al. [66]
miR-489-3pTLR4HaCaTInhibition of keratinocyte proliferation and TLR4/NF-κB signalingYe et al. [67]
CAMK4, calmodulin-dependent protein kinase IV; CARD10, caspase recruitment domain family member 10; CD276, cluster of differentiation 276; DYRK1A, dual-specificity tyrosine phosphorylation regulated kinase 1A; ERBB4, Erb-B2 receptor tyrosine kinase 4; FERMT1, fermitin family homolog 1; FOXM1, forkhead box M1; FZD, frizzled; IRAK1, IL-1 receptor-associated kinase 1; LCN2, lipocalin 2; NF-κB, nuclear factor-κB; STAT3, signal transducer and activator of transcription 3; TLR, Toll-like receptor; TRAF6, tumor necrosis factor receptor-associated factor 6.
Table 2. The targets and biological mechanisms of anti-inflammatory miRNAs for treating atopic dermatitis.
Table 2. The targets and biological mechanisms of anti-inflammatory miRNAs for treating atopic dermatitis.
miRNA CodeTargetsExperimental ModelsOutcomeReference
ST-miRCCL22CCL22RAW264.7 macrophages and wild-type miceReduction of IgE, IL-4, and Th17 cellsYoon et al. [79]
miR-10a-5pHyaluronan synthase 3Keratinocytes and patientsInhibition of keratinocyte proliferation and cytokines/chemokinesVaher et al. [80]
miR-124RELA (the gene name of p65)Keratinocytes and patientsDownregulation of IL-8, CCL5, and CCL8Yang et al. [81]
miR-143IL-13Rα1KeratinocytesEnhancement of the synthesis of filaggrin, loricrin, and involucrinZeng et al. [83]
miR-146aIRAK1 and TRAF6KeratinocytesSuppression of TLR2-induced production of TNF-α, IL-8, and CCL20.Meisgen et al. [85]
miR-146aCARD10 and IRAK1Keratinocytes and wild-type and miR-146a−/− miceAlleviation of chronic skin inflammation through innate immune response suppression in keratinocytesRebane et al. [86]
miR-375-3pYAP1HaCaTInhibition of IL-1β and IL-6 accompanied by a reduction in NF-κ nuclear translocationCheng et al. [89]
miR-1294STAT3HaCaT, 3D skin equivalent, and wild-type miceDecrease in inflammation and skin barrier destructionChen et al. [90]
CARD10, caspase recruitment domain family member 10; CCL, chemokine (C-C motif) ligand; IgE, immunoglobulin E; IL, interleukin; IRAK1, IL-1 receptor-associated kinase 1; STAT3, signal transducer and activator of transcription 3; ST, Salmonella typhimurium; TLR, Toll-like receptor; TNF-α, tumor necrosis factor-α; TRAF6, tumor necrosis factor receptor-associated factor 6; YAP1, Yes-associated protein 1.
Table 3. The targets and biological mechanisms of anti-inflammatory miRNAs for skin wound healing.
Table 3. The targets and biological mechanisms of anti-inflammatory miRNAs for skin wound healing.
miRNA CodeTargetsExperimental ModelsOutcomeReference
miR-23bASK1HaCaT and wild-type miceInhibition of cytokines and enhancement of α-SMA expressionLi et al. [102]
miR-31EMP-1Keratinocytes and healthy volunteersEnhancement of wound healing via increased keratinocyte migrationLi et al. [104]
miR-34aBcl-2 and CCND1Wild-type and miR-34a−/− micemiR-34a deficiency leads to impaired wound closureZhao et al. [106]
miR-497AKT2 and E2F3Human dermal fibroblasts and wild-type miceInhibition of cytokines and acceleration of diabetic wound healingBan et al. [107]
AKT2, RAC-β serine/threonine-protein kinase; ASK1, apoptotic signal-regulating kinase 1; α-SMA, α-smooth-muscle actin; Bcl-2, B-cell lymphoma 2; CCND1, cyclin D1; E2F3, E2F transcription factor 3; EMP-1, epithelial membrane protein 1.
Table 4. miRNAs as the targets of bioactive molecules for inhibiting skin inflammation.
Table 4. miRNAs as the targets of bioactive molecules for inhibiting skin inflammation.
Bioactive MoleculeTarget miRNAsExperimental ModelsOutcomeReference
ResveratrolmiR-17HaCaTResveratrol upregulates miR-17 for alleviated lipopolysaccharide-induced inflammationWang and Zhang [121]
AdalimumabmiR-146a-5pPsoriasis patientsReduction of miR-146a-5p is associated with the improvement of psoriasisMensà et al. [123]
EbosinmiR-155-3pHaCaT and wild-type miceEbosin reduces psoriatic inflammation through miR-155-3p/IL-17 axisGuo et al. [124]
CircRAB3BmiR-1228-3pHaCaTCircRAB3B negatively regulates the expression of miR-1228-3pLu et al. [127]
PUVAhsa-miR-4516HaCaThas-miR-4516 mediates PUVA-induced apoptosis in keratinocytesChowdhari and Saini [128]
BerberinemiR-21Mast cells and wild-type miceBerberine mitigates allergic dermatitis via miRNA/p38 signalingLi et al. [130]
Electro-acupuncturemiR-155Mast cells and wild-type ratsAcupuncture lowered the expression of miR-155 through the signaling of IL-33Wang et al. [132]
IL-32γmiR-205HaCaT and wild-type and IL-32γ transgenic miceIL-32γ inhibited AD through downregulation of miR-205Lee et al. [134]
BelinostatmiR-335Keratinocytes and AD patients Liew et al. [135]
Vitamin D and gingermiR-15a, miR-146a, and miR-155Wild-type ratsCombined treatment of vitamin D and ginger decreased miR-155 and increased miR-15a and miR-146aAl-Rawaf et al. [137]
ResveratrolmiR-129HUVEC and wild-type ratsResveratrol promoted wound healing through TRAF downregulation via MSC-EV-carried miR-219Hu et al. [139]
C66miR-146aHUVEC and wild-type miceDecreased miR-146a level in diabetic wounds was upregulated after C66 treatment.Huang et al. [141]
Jiang Tang Xiao KemiR-139-5pWild-type and miR-139−/− miceDecreased miR-139-5p expression in the infected wound after topical JTXK administrationZhang et al. [143]
Lipoteichoic acidmiR-143Keratinocytes and wild-type miceLipoteichoic acid activated TLR2 to upregulate miR-143Xia et al. [144]
TroxerutinmiR-205-3p, miR-483-5p, miR-513b, and miR-3648HaCaTmiR-205-3p expression was elevated, while miR-483-5p, miR-513b, and miR-3648 expressions were decreased by troxerutinLee et al. [145]
AD, atopic dermatitis; C66, (2E,6E)-2,6-bis(2-(trifluoromethyl)benzylidene)cyclohexanone; HUVEC, human umbilical vein endothelial cell; PUVA, psoralen and ultraviolet A; TLR, Toll-like receptor.
Table 5. The approaches for facile delivery of miRNAs into target cells and skin.
Table 5. The approaches for facile delivery of miRNAs into target cells and skin.
miRNA CodeApproachInflammation ModelsOutcomeReference
miR-146aCPPsIrritant contact dermatitisFacile internalization of miR-146a/CPP nanocomplex into keratinocytes inhibits inflammation responseUrgard et al. [152]
miR-31CPPsExcisional woundmiRNA-31/CPP nanocomplex within an electrospun nanofiber facilely regenerates woundsMulholland et al. [153]
miR-146aCerium oxide nanoparticlesDiabetic woundmiR-146a-conjugated nanoparticles correct wound-healing impairmentZgheib et al. [156]
miR-146aCerium oxide nanoparticles in silk fibroinDiabetic woundIncorporation of miR-146a-conjugated nanoparticles into silk fibroin improves the diabetic wound repairNiemiec et al. [158]
miR-21BA-PEI nanoparticlesExcisional woundBA-PEI nanoparticles enhance the effect of miR-21 on wound healingWang et al. [161]
miR-223-5pHyaluronic acid nanoparticles in hydrogelsExcisional woundNanoparticle-loaded hydrogels control macrophage polarization during wound healingSaleh et al. [162]
miR-210 antisenseReconstituted high-density lipoprotein nanogelsPsoriasiform lesionTopical application of the nanogels significantly reduces immune cell accumulation in lesionsFeng et al. [164]
miR-125bExosomesExcisional woundExosomal transfer of miR-125b to fibroblasts suppresses sirtuin 7 to accelerate wound healingXia et al. [167]
miR-197Ultrasound-mediated nanocomplex deliveryXenograft transplantation miceUltrasound-assisted delivery enhances miR-197-loaded nanocomplexLifshiz Zimon et al. [170]
BA-PEI, bile acid-attached polyethyleneimine; CPPs, cell-penetrating peptides.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/biom12081072

AMA Style

Yang S-C, Alalaiwe A, Lin Z-C, Lin Y-C, Aljuffali IA, Fang J-Y. Anti-Inflammatory microRNAs for Treating Inflammatory Skin Diseases. Biomolecules. 2022; 12(8):1072. https://doi.org/10.3390/biom12081072

Chicago/Turabian Style

Yang, Shih-Chun, Ahmed Alalaiwe, Zih-Chan Lin, Yu-Chih Lin, Ibrahim A. Aljuffali, and Jia-You Fang. 2022. "Anti-Inflammatory microRNAs for Treating Inflammatory Skin Diseases" Biomolecules 12, no. 8: 1072. https://doi.org/10.3390/biom12081072

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