Next Article in Journal
Metallodrugs: Mechanisms of Action, Molecular Targets and Biological Activity
Next Article in Special Issue
The Immune Response in Adipocytes and Their Susceptibility to Infection: A Possible Relationship with Infectobesity
Previous Article in Journal
Determinants of Lipid Domain Size
Previous Article in Special Issue
Indole-3-Propionic Acid, a Functional Metabolite of Clostridium sporogenes, Promotes Muscle Tissue Development and Reduces Muscle Cell Inflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Skin Microbiota in Atopic Dermatitis

by
Dora Hrestak
1,
Mario Matijašić
1,*,
Hana Čipčić Paljetak
1,
Daniela Ledić Drvar
2,
Suzana Ljubojević Hadžavdić
2 and
Mihaela Perić
1
1
Department of Intercellular Communication, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Šalata 2, 10000 Zagreb, Croatia
2
Department of Dermatology and Venereology, University Hospital Centre Zagreb, University of Zagreb School of Medicine, Kišpatićeva 12, 10000 Zagreb, Croatia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(7), 3503; https://doi.org/10.3390/ijms23073503
Submission received: 25 February 2022 / Revised: 18 March 2022 / Accepted: 22 March 2022 / Published: 23 March 2022
(This article belongs to the Special Issue Microorganism in Inflammation)

Abstract

:
The skin microbiota represents an ecosystem composed of numerous microbial species interacting with each other, as well as with host epithelial and immune cells. The microbiota provides health benefits to the host by supporting essential functions of the skin and inhibiting colonization with pathogens. However, the disturbance of the microbial balance can result in dysbiosis and promote skin diseases, such as atopic dermatitis (AD). This review provides a current overview of the skin microbiota involvement in AD and its complex interplay with host immune response mechanisms, as well as novel therapeutic strategies for treating AD focused on restoring skin microbial homeostasis.

1. Human Skin Microbiota

All microorganisms residing in a multicellular host form its microbiota. Microbiota helps to maintain the balance (homeostasis) of the host system, contributes to immune responses and promotes tissue repair. Disruption of microbiota balance often results in inflammation or infection that can lead to various pathophysiological conditions and diseases. Additionally, imbalance leads to a microbial shift, often reducing beneficial species, and thereby causing dysbiosis. While the predominant part of the human microbiota is located in the gastrointestinal tract, remaining microorganisms are unequally distributed across the body, including the skin.
The skin is the largest organ of the human body, forming a protective barrier that prevents infections from environmental pathogens, and regulates body temperature, prevents water loss and triggers pain response. The human skin (Figure 1) can also be perceived as an ecosystem, where different parts of the body represent different habitats for bacteria, fungi, viruses and archaea. Microorganisms colonize the skin surface according to the spectrum of preferences, forming units that contribute to the body’s immune system and protect against other pathogenic life forms. Each microbe has adapted to the physicochemical characteristics of its habitat—a behavior similar to that of Earth’s flora and fauna. In addition, because skin has little nutritive value aside from lipids and proteins, the main requirement for microbe survival is their utilization of resources found in the stratum corneum and/or sebum, such as amino acids or urea [1].
Bacterial phyla which can be found on the healthy human skin are Actinobacteria, Firmicutes, Proteobacteria and Bacteriodetes [2], with an emphasis on Staphylococcus, Corynebacterium and Propionibacterium genera that comprise more than 60% of the bacterial skin population [3]. The composition of skin microbiota is highly dependent on the characteristic physiology of the skin site, and specific bacterial taxa were found to inhabit dry, moist and sebaceous microenvironments [4]. Dry areas (forearm, buttock, various parts of the hand) are the most diverse skin sites, reported to contain greater bacterial diversity than the gut or the oral cavity of the individual [5], and harboring numerous phylotypes including β-Proteobacteria, Corynebacteria and Flavobacteriales [2]. Moist skin sites (nostril, armpit, navel, toe webs, inner elbow, groin, cubital and popliteal fossa, palms and soles) provide thermally stable, warm environments, with Corynebacteria and Staphylococci species being the most abundant organisms that can withstand humid conditions [4]. Finally, sebaceous areas (forehead, retroauricular area, lateral sides of the nostrils, back) show the lowest bacterial diversity and are populated mostly by lipophilic Propionibacteria and Staphylococci species [3,4,6]. Although the skin microbiota encounters frequent perturbations due to the constant environmental changes, longitudinal sampling revealed its stability over a 2-year period, with the microbial communities at sebaceous sites being most persistent and those at foot sites the least stable [7,8]. As opposed to bacterial communities, the composition of fungal communities was reported similar across the body sites despite different skin physiology [9]. Studies on fungal communities distinguish the predominance of the genus Malassezia on healthy human skin, specifically on the chest and arms (M. globosa), the trunk (M. sympodialis) and on facial sites (M. restricta) [9,10,11]. Foot sites were colonized by diverse communities of Malassezia, Aspergillus, Cryptococcus, Rhodotorula, Epicoccum and others, which display lower stability over time [9,12]. Several studies show that normal skin microbiota also included Candida [12,13]. However, organisms belonging to the kingdom of fungi represent the least abundant inhabitants of the skin [8]. In contrast to bacterial and fungal communities, no healthy human skin core virome was found conserved among individuals [8]. Foulongne et al. reported a metagenomic study that displayed multiple eukaryotic virus families, namely, Polyomaviridae, Papillomaviridae and Circoviridae [14]. A more recent study by Hannigan et al. demonstrated the presence of Adenoviridae, Anelloviridae, Circoviridae, Herpesviridae, Papillomaviridae and Polyomaviridae [15]. However, a core phageome consisting of Propionibacterium, Staphylococcus, and Streptococcus bacteriophages was recently identified. These phages are obligate partners to the most abundant bacterial species and are therefore site-specific [8].
The stability of skin microbial communities varies between age groups, the diversity being inversely proportional to maturity [16,17]. Initial colonization of the skin in newborn babies depends greatly on the delivery mode and mother’s bacteria to which the neonates are exposed during labor [18]. The skin microbiota of children under 12 years comprises mostly Streptococcus, Granulicatella, Gemella, Rothia and Haemophilus bacterial genera [16], together with highly diverse fungal communities of Ascomycota (Aspergillus, Epicoccum, Phoma), Cladosporium and Cryptococcus, and a low abundance of Malassezia [17]. During puberty, the structure of the skin changes, as the increased hormone levels stimulate additional production of sebum, thus favoring the expansion of lipophilic microbiota. The Streptococcus genus is gradually replaced with Propionibacterium and Corynebacterium, with Malassezia as a predominate fungal constituent of adult skin microbiota [16,17].

2. Atopic Dermatitis

Atopic dermatitis (AD) is one of the most common, chronic, inflammatory skin diseases of the modern world. Additionally known as ‘eczema’, this chronic recurrent disorder is characterized by an intense itching sensation and eczematous lesions. Acute lesions are manifested as bright erythema, oedema and oozing, while chronic lesions present as xerosis, lichenification and residual dyspigmentation. In comparison to healthy controls, both non-lesional and lesional AD skin show higher transepidermal water loss (TEWL) and pH values [19] that corelate with disease severity and predisposition [20,21]. Distributional and morphological characteristics generally relate to age, whilst some phenotypes show predisposition for certain body sites. Although most frequently manifested throughout the first year of life, the disease can occur at any age. Early-onset AD is sub-classified based on age groups as infantile (<2 years), childhood (2–12 years) or adolescent (12–18 years). Additionally, children with AD are more prone to developing other atopic diseases than children without AD [22,23], and AD can be a first step in the sequential development of other atopic manifestations later in life (food allergy, asthma and allergic rhinitis), in the process of the “atopic march” [24,25,26]. Adult-onset occurs after 18 years of age. Recent studies indicate a third class of AD that occurs after the age of 60 and is, therefore, termed the elderly-type AD, sub-labelled as elderly-onset, relapsing and continuous [27,28,29].
Diagnostic criteria established for AD consist of basic features, such as pruritus, lichenification, and chronically relapsing course, and personal and/or family atopic history as well as minor features take into consideration less specific symptoms [30,31,32]. Disease severity can be measured by various tools, estimating the extent of affected areas and multiple types of symptoms to help assess and score clinical signs. Typically preferred are the Scoring of Atopic Dermatitis Index (SCORAD) and the Eczema Area and Severity Index (EASI) [33,34,35].

3. Pathogenesis of Atopic Dermatitis

There are numerous risk factors associated with the development of AD, from genetic predisposition to environmental conditions. Family history of atopic diseases presents the most prominent risk factor for AD. Research shows that the occurrence of any atopic disease in one of the parents increases 1.5-fold the possibility a child developing AD. Moreover, the risk is further increased 3-fold and 5-fold, respectively, if one or both parents suffer from AD [36]. Several environmental risk factors have been proposed to enhance the prevalence of AD, such as living in an urban setting, low UV light exposure, dry climate, Western diet, repeated exposure to antibiotics in early childhood, as well as high level of household education [37].
Pathophysiology of AD is very complex and not yet completely elucidated. Multiple contributing factors, including epidermal barrier impairment, immune dysregulation and alteration of skin microbiota, contribute to the disease. The integration of these factors, with their different intensities and combinations, is thought to cause the varying clinical presentations of AD [38].

3.1. Epithelial Barrier Dysfunction and Immune Dysregulation

The skin barrier plays a major role in the protection from commensal and pathogen penetration, forming a thin line between health and disease. Several factors contribute to epithelial barrier dysfunction in AD, including mutations of genes that encode structural and functional proteins of the epidermis and epigenetic modifications affecting the regulation of immune response and inflammatory processes [39]. Filaggrin is one of the key epidermal proteins for production of a natural moisturizing factor and is essential for maintaining stratum corneum hydration. Mutations in the filaggrin gene are found in about half of patients with moderate to severe disease [39] and can be associated with early-onset AD [40]. Deficit in filaggrin production also results in aberrant keratinocyte differentiation and insufficient skin lipid content [41]. Skin lipids (e.g., ceramides, free fatty acids and cholesterol) are vital for the maintenance of the epidermal barrier function, and thus are responsible for prevention of TEWL and penetration of irritants, allergens and microbes. Disbalance in the composition of skin lipids, as well as the reduction of the lipid content, are associated with the development of AD [19,42].
Epidermal barrier disruption stimulates keratinocytes to release chemokines (thymus and activation-regulated chemokine (TARC), macrophage-derived chemokines (MDC)) and cytokines (IL-1β, IL-25, IL-33 and thymic stromal lymphopoietin (TSLP)). This leads to infiltration of leukocytes, primarily dendritic cells (DC), eosinophils and T-cells [43], initiation of TH2 cell responses, as well as activation of skin-resident group 2 innate lymphoid cells (ILC2s), thus inducing inflammation [44,45,46]. TH2 cells release IL-4, IL-5, IL-13, IL-25 and IL-31, which in turn activate B-cells to produce IgE molecules. ILC2s also release IL-5 and IL-13 and can provide an additional boost to type 2 immunity and IgE production [46]. Together with TH2 cells, IL-22-secreting TH22 cells play an important part in the initiation and acute phase of AD [45], while the switch to TH1/TH17 response characterizes a chronic disease [47]. Keratinocyte-expressed chemokines also recruit different dendritic cell subtypes, including inflammatory dendritic epidermal cells (IDECs) and Langerhans cells, which express high-affinity immunoglobulin-ε receptors (FcεR1s). Binding of IgE antibodies to these receptors facilitates the uptake of allergens and triggers hypersensitivity reactions [48]. Additionally, epithelial cell-derived IL-33 and TSLP and type 2 cytokines can directly activate itch-sensory neurons to induce pruritus [49,50]. The dynamic interplay between epithelial barrier dysfunction, type 2 immunity and pruritus is schematically shown on Figure 2.

3.2. Dysbiosis of Skin Microbiota

The skin of patients with AD reveals considerable anomalies of the microbial communities when compared to the microbiota of healthy subjects; however, it is not certain whether these changes are the cause or the result of epidermal barrier dysfunction and immune dysregulation. Shi et al. showed that 20 genera usually found on healthy individuals also occupied the skin of AD patients [16]. However, the skin in AD is characterized by an expressed microbial imbalance and reduced diversity, specifically manifested as a decrease in the genera Cutibacterium, Streptococcus, Acinetobacter, Corynebacterium and Prevotella, and a marked increase of Staphylococcus, especially of S. aureus [51]. The reduced microbial diversity is particularly evident during severe flares of the disease, with the reported composition of Staphylococcus usually reduced to a single S. aureus strain [18]. The treatment and recovery reverted the microbiota to its pre-flare configuration [51].
The disequilibrium of skin commensals might not sound like an ominous occurrence, however, it instigates another set of problems. Because a balance normally ensures that commensals remain benign and/or beneficial, a microbial shift provides growth space for species whose greater numbers cause more harm than good (Figure 3). Consequently, newly dominant strains can trigger skin inflammation and diseases. S. aureus is the principal bacterial species associated with AD [52]. As an opportunistic pathogen, it is well adapted to adhere to the skin, disrupt the epithelial barrier and trigger the host immune system, in turn inducing skin inflammation [53,54]. While the carriage of S. aureus in healthy subjects is about 20%, the prevalence on the skin of patients with AD is increased and varies from 30% to 100%, depending on patient age, AD severity, as well as sampling and analysis methods [55]. Additionally, the reported abundance of S. aureus in patients with AD was 70% on lesional skin and 39% on non-lesional skin or healthy skin of the same patient, confirming the correlation between S. aureus and disease severity [55,56]. A recent study identified differences in S. aureus strain structure isolated from AD skin lesions from that of non-lesional skin: patients with severe AD tend to carry the clonal complex 1 (CC1) strains, whereas asymptomatic individuals carry the CC30 strains [57]. The chronic persistence of S. aureus on eczematous skin lesions and difficulties of eradicating it using antibiotics were found to be associated with the prevalence of staphylococcal biofilm communities on the skin of patients with AD. Indeed, a study by Di Domenico et al. confirmed the severity of AD can be linked to biofilm formation by S. aureus [56]. A number of studies focused on the interplay between S. aureus, epithelial barrier disruption and the immune system. Unlike healthy skin, skin in AD is more permissive to S. aureus colonization due to its reduced anti-microbial peptide (AMP) levels [58]. The reduced expression of AMPs, particularly defensins and cathelicidins, can be the result of TH2-derived IL-4 and IL-13 cytokines [59]. It was also shown that S. aureus adheres more strongly to AD skin, due to the filaggrin deficiency and deformed corneocytes of the stratum corneum [60]. Additionally, S. aureus produces a number of different proteolytic enzymes and toxins, as well as stimulates the expression of endogenous keratinocyte proteases, which can disrupt the integrity of the skin barrier and enable penetration through stratum corneum [61,62]. Other toxins from S. aureus can directly induce type 2 immune response by activating immune cells and triggering the expression of the inflammatory mediators such as IL-4, IL-13, IL-22 and TSLP [61].
Several reports have demonstrated that Staphylococcus epidermidis overgrowth can also be linked to AD pathogenesis. Usually perceived as a skin commensal essential in coordinating the maturation of the immune system and combating pathogens, S. epidermidis is, in certain conditions, able to contribute to the inflammatory reaction in AD [63]. Byrd et al. found that, unlike single strain S. aureus communities linked to severe AD flares, more heterogenous S. epidermidis strains dominated in patients with less severe symptom manifestations [18]. A number of studies reported that not only Staphylococcus strains incite development of AD. Previously known under the name Propionibacterium acnes, Cutibacterium acnes is one of the most widespread skin commensals, playing a role in the skin defense mechanisms [64]. However, C. acnes can also cause damage to the skin by enhancing S. aureus cytolytic activity, thus inducing proinflammatory cytokine production [65]. The porphyrin molecule coproporphyrin III (CIII) produced by C. acnes was found to induce S. aureus aggregation and biofilm formation, suggesting the cooperation between C. acnes and S. aureus [66].
In contrast to numerous studies on skin bacteria, reports on mycobiota diversity in AD are scarce. As in healthy subjects, Malassezia species (especially M. globosa and M. restricta) were predominant in patients with AD [12]. M. restricta dominated over M. globosa in patients with mild or moderate disease, while the ratio of the two species was equal in patients with severe disease [12,67]. Malassezia species were shown to penetrate the epithelial barrier of AD patients, causing activation of immune cells and skin inflammation [68]. In addition, Malassezia allergens can trigger a specific IgE response, contributing to the disease [68]. As for non-Malassezia fungi, Candida albicans, Cryptococcus diffluens and Cryptococcus liquefaciens were detected more often in patients with AD than in healthy subjects [12]. However, the role of these species in AD pathophysiology needs to be further elucidated.
Contrary to microbial communities being associated with inflammation incitement, there is evidence that the presence of some microbes negatively corelates with AD progression (Figure 4). Scharschmidt et al. confirmed that early exposure to commensal Staphylococci plays a role in antigen-specific tolerance that may prevent AD development in mice [69]. Those findings were corroborated by a study in which decreased susceptibility to AD was associated with early exposure of infants to Staphylococci commensals [70]. Studies reported several species of Staphylococcus genus suppressing S. aureus and its effect on disease progression. The coagulase-negative (CoNS) S. epidermidis, Staphylococcus hominis and Staphylococcus lugdunensis successfully inhibited S. aureus colonization and biofilm formation [71,72]. A similar effect was observed in more recent reports by Zipperer et al. and Nakatsuji et al., who showed that both S. lugdunensis and S. hominis produce lantibiotics that inhibit S. aureus growth [73,74]. Furthermore, an in vitro study demonstrated that a co-infection of S. aureus with the Corynebacterium striatum, in comparison to exclusive S. aureus infections, resulted in an S. aureus shift towards a commensal state [75]. In addition to the C. striatum, byproducts from glycerol fermentation by Cutibacterium acnes also showed S. aureus inhibition, without disrupting the skin microbiome balance [76]. Aside from metabolites of bacterial commensals, the MgSAP1 protease secreted by Malassezia globosa was shown to hydrolyze the S. aureus Protein A, thereby hindering its biofilm formation [77].
These findings provide evidence on the key role of skin microbiota in the pathogenesis of AD. However, the current scientific knowledge is still lacking, and future research efforts need to be directed towards fully understanding the composition of the microbial ecosystem of the human skin, as well as the complex interactions regulating the host–microbiota relationship in health and disease.

4. Treatment of Atopic Dermatitis

The AD management approach is based on disease severity, age and location. A continuous daily emollient application to relieve symptoms and enhance skin hydration represents a baseline therapeutical approach for both children and adults [78]. Preparations such as petrolatum, physiologic lipids or ceramide-based lipids are known to reduce TEWL and decrease bacterial colonization, which improves overall skin barrier function [79]. While standard aqueous creams show positive results in terms of symptom improvement, pH-modified moisturizers significantly alleviate symptoms in AD and serve as a useful treatment adjunct [80]. Emollients can also affect Staphylococcus species abundance and microbiota diversity [81]. However, in the acute phase of the disease, application of potent anti-inflammatory agents is required, with topical corticosteroids (TCS) representing the first-line anti-inflammatory treatment [78]. Although highly effective in improving AD symptoms, long-term use of corticosteroids is discouraged because of their side-effect profile and subsequent patient-compliance issues [78,82]. At the beginning of the 21st century, non-steroid topical calcineurin inhibitors (TCI) were introduced as an alternative to TCS treatments for AD. Macrolide derivatives tacrolimus and pimecrolimus are calcineurin inhibitors that prevent T-cell signal transduction and IL-2 transcription, thus suppressing inflammation [83,84]. Unlike TCS, TCIs are suitable for long-term treatment, and use of tacrolimus is recommended for the maintenance and reduction of relapses, often after initial corticosteroid treatment [78]. In addition, tacrolimus showed a positive impact on the skin microbiome in AD patients [85]. Antibiotics are included in AD treatment in cases of bacterial superinfection, but due to antibiotic resistance and the potential negative effect of antibiotics on commensal bacteria, this treatment method is not a long-term option [86]. Other therapeutic approaches include phototherapy with ultraviolet (UV) light, which can reduce AD recurrence. Narrow band ultraviolet B (nBUVB) phototherapy has been shown to decrease the S. aureus ratio in the skin microbiota [87]. Severe AD may require hospitalization and systemic immunosuppressive treatment with cyclosporine A, a short course of oral glucocorticoids, methotrexate, azathioprine and mycophenolic acid or, as an alternative, biologic therapy [88].

5. Probiotics and Prebiotics in Treatment of Atopic Dermatitis

Probiotics are live microorganisms with immunomodulatory features which provide beneficial effects on the host’s well-being. Although most probiotic applications have been targeted at gastrointestinal tract disorders, recent reports recognized a vast potential of utilizing probiotics for promoting skin health and managing various skin conditions. Over the last few years there have been scientific breakthroughs with reference to treating AD using oral or topical probiotic cultures. A recent animal study found that orally administered Lactobacillus paracasei KBL382 successfully ameliorates AD symptoms in mice via modulating the immune response and gut microbiota [89]. Similar research was conducted by Kwon et al., in which Lactobacillus sakei WIKIM30 isolated from kimchi and orally delivered to mice resulted in stimulation of Treg cell generation and suppression of TH2 inflammatory response, as well as in restoring the balance of gut microbiota [90]. Another animal study reported that administration of Lactobacillus rhamnosus IDCC 3201 tyndallizate (RHT3201) to mice resulted in less severe AD symptoms in comparison to controls, together with dose-dependent reductions in dermatitis scores [91]. Furthermore, experiments on mouse animal models by Kim et al. suggested oral administration of β-glucan and Lactobacillus plantarum LM1004 inhibited TH2 cell responses and activated Treg immunoregulatory functions, as well as increased relative abundance of butyrate-generating microorganisms in the gut [92].
Along with the research on animal models, clinical trials in humans also showed promising results regarding the oral use of probiotics in treating AD. One of the earliest publications in the field describes oral administration of a mixture containing two Lactobacillus strains (lyophilized Lactobacillus rhamnosus 19070-2 and Lactobacillus reuteri DSM 122460) to children with AD, throughout a period of six weeks in a double-blind placebo-controlled crossover study. The treatment provided a moderate improvement in the clinical severity of eczema [93]. L. rhamnosus [94,95,96] and L. plantarum CJLP133 [97] also displayed a positive treatment efficacy on AD during clinical trials on children. Another study reported successful treatment of AD patients using Lactobacillus fermentum VRI-033 PCC during a double-blind randomized placebo-controlled trial, with reduced SCORAD index and change in AD severity compared to placebo-treated individuals [98]. Moreover, Niccoli et al. efficiently treated pediatric AD patients with a lyophilized form of Lactobacillus salivarious LS01, reporting a significant decrease in SCORAD value and significant improvement in itching intensity when compared to the placebo control group, and both therapy benefits persisting after suspension of treatment [99]. Although typically tested oral probiotic formulations most often consisted of Lactobacillus strains, several studies investigated the positive effects of other bacterial strains or mixtures of different probiotic bacterial strains in management of AD. Matsumoto et al. reported that the administration of Bifidobacterium animalis subsp. lactis LKM512 alleviated itch in AD patients and considerably improved the quality-of-life scores when compared with the controls, suggesting an antipruritic effect of B. animalis [100]. A probiotic mixture of Bifidobacterium lactis CECT 8145, B. longum CECT 7347, and Lactobacillus casei CECT 9104 was reported to reduce the SCORAD index in AD patients compared with the control group [101], while a case report using a mixture of Bifidobacterium lactis HN019, Lactobacillus acidophilus NCFM, Lactobacillus rhamnosus HN001 and Lactobacillus paracasei LPC-37 described an evident response in treating severe AD with significant change in AD severity scores [102]. In fact, a meta-analysis suggested the administration of probiotics has a positive influence on the treatment of AD, with the greatest effect observed in studies using a mixture of different bacterial species [103].
Along with studies on oral probiotics, which indirectly influence skin diseases, a number of topical probiotic formulations have been proposed to ameliorate skin conditions by suppressing inflammation and restoring skin microbiota balance. Nakatsuji et al. demonstrated that the topical application of commensal skin bacteria is effective in protecting against pathogen species, with reduced S. aureus colonization due to selective anti-microbial peptides secreted by commensal coagulase-negative Staphylococci, improvement of clinical symptoms and decreased local inflammation [74,104]. A group of authors also conducted a first-in-human topical microbiome transplantation after collecting the commensal Roseomonas mucosa from healthy subjects. After a six-week therapy, significant decrease in SCORAD and pruritus was noted, as well as reduction in disease severity and no adverse effects or complications, which, consequently, signifies a lesser need for topical steroids [105]. Interestingly, several studies also explored the effects of the topical application of gut commensals for treating AD. A cosmetic lotion containing heat-treated Lactobacillus johnsonii was linked to the reduction of S. aureus load on the skin of adult AD patients and clinical improvement of AD symptoms [106]. Another report describes significant improvement in skin barrier integrity, erythema, scaling and pruritus in patients with AD after a 2-week topical administration of a cream consisting of sonicated Streptococcus thermophilus [107]. Finally, Gueniche et al. found that topical ointment containing lysate of Vitreoscilla filiformis, a Gram-negative bacterium found in thermal springs, which has traditionally been used in treating dermatological diseases, also resulted in clinical improvement in patients with AD [108]. Even though not technically probiotics, the probiotic bacteria preparations evidently have the ability to interact with the skin components and alleviate AD symptoms.
Unlike the studies on probiotics, the research on prebiotics and synbiotics (combination of probiotics and prebiotics) in AD treatment is relatively scarce. Chang et al. published a meta-analysis of six randomized controlled trial studies providing evidence to support the use of synbiotics composed of mixed strains of bacteria for the treatment of AD for children aged 1 year or older [109]. A double-blind randomized study by Passeron et al. performed on children with AD using a prebiotic preparation as well as synbiotic preparation (Lactobacillus rhamnosus Lcr35 plus prebiotic) showed both treatments significantly improved AD symptoms, with no significant difference noted between the two treatments [110]. A similar double-blind study was conducted by Aldaghi et al. on AD infants that were administered either a synbiotic mix containing L. rhamnosus, L. euteri and B. infantis or vitamin D3. The report confirmed both treatments significantly decreased SCORAD scores when compared to the control group [111].
Although further investigation is needed, the results shown in these studies (Table 1) warrant a potential interest in using probiotics and prebiotics as therapeutic treatment in the management of AD. However, it should be noted that these potential therapies are considered as Microbiotic Medicinal Products (MMPs). An MMP is any medicinal product containing living, dead, fragments or components of the microbiota (i.e., bacteria, yeasts, phages, etc.) with the purpose to prevent or treat human diseases through a pharmacological, microbiological, neurological, immunological or metabolic mode of action, or to make a medical diagnosis [112]. Although showing great promise for treating human disease, including AD, the development of these products (especially live biotherapeutic products, LBPs) is faced with many scientific, clinical and regulatory challenges [113]. The current set of requirements for a particular type of the product is not specifically or uniformly defined for LBPs at the global level, so the acceptance criteria for the product quality, efficacy, and safety are often unclear or inappropriate and eventually need to be adjusted to an individual product. The most obvious issue arises when considering the basic requirement for the pharmaceutical product, its sterility, that these therapies evidently cannot achieve. Additionally, the manufacturing of LBPs is complex due to batch-to-batch variations and many factors related to culture conditions and product stability (viability, shelf life, genetic stability) that could influence bacterial properties, consequently changing product efficacy or safety. Determining LBPs’ safety is different from other medicines since the product itself does not reach the systemic circulation, while its activities or metabolites may act directly or indirectly on (systemic) physiological functions of the host, so toxicity is not always directly related to the dosage. Furthermore, the translation of data from animals to humans is almost impossible due to the holobiont concept [114,115], a result of the coevolution of the microbiome and its human host that cannot be reproduced in animal species. Clinical efficacy for LBPs can only be proven in an independent trial of acceptable quality, using well-defined treatment conditions and dosage and with preliminary defined, validated endpoints. As currently no standard clinical trial format exists (products, target populations and application modes differ on a case-by-case basis), proving efficacy in a standardized manner is quite difficult to achieve. Many environmental factors (e.g., transport and storage conditions) as well as host-related factors (e.g., health status, stomach pH, interference with diet, the composition of the recipient microbiota, ethnicity, etc.) can affect the final trial outputs. Nevertheless, despite the many challenges and uncertainties, it seems opportune and scientifically sound to further advance both scientific tools and regulatory frameworks for the development of these therapies, since the future products might offer unique therapeutic opportunities and equip the medical community with additional means for combating major human diseases.

6. Conclusions and Future Perspectives

AD is a complex, multifactorial disease. Although not a life-threatening condition, AD has a severe impact on the patient’s quality of life and is often associated with numerous medical and mental health comorbidities. Our understanding of AD and its pathophysiology has made major advances in the last decade, with detailed insights on the complex interplay between epidermal barrier dysfunction and immune system activation. Moreover, technological advances improved our ability to identify and characterize skin microbial communities, enhancing our knowledge on the disrupted host–microbiota relationship in AD. Recent reports provided evidence for introducing skin microbiota dysbiosis as one of the key features of the disease initiation and progression, paving the way for the development of novel therapeutic interventions. Probiotic and prebiotic preparations, as well as skin microbiota transplantation, are finding their way to clinical applications with promising results in AD management. However, more studies are needed to evaluate the influence of systemically and locally applied therapies to skin microbiota as well as to assess the mechanisms through which the effects are achieved. Additionally, the major challenge will be to translate these research findings into innovative new therapies and to overcome both scientific and regulatory challenges in developing microorganism-based medicinal products with an intended use in patients with AD.

Author Contributions

Conceptualization, M.M. and M.P.; writing—original draft preparation, D.H., M.M. and M.P.; writing—review and editing, H.Č.P., D.L.D., S.L.H. and M.P.; visualization, M.M.; supervision, M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research is a part of the project “Synergistic innovative combination of microbiota components as a basis for the development of innovative topical products for the treatment and prevention of inflammatory conditions of human skin” (acronym PROBITECT), funded by the EU through the ERDF (GA KK.01.2.1.02.0137) and Scientific Center of Excellence for Reproductive and Regenerative Medicine (project “Reproductive and regenerative medicine—exploration of new platforms and potentials”, GA KK01.1.1.01.0008 funded by the EU through the ERDF).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Scharschmidt, T.C.; Fischbach, M.A. What Lives On Our Skin: Ecology, Genomics and Therapeutic Opportunities Of the Skin Microbiome. Drug Discov. Today Dis. Mech. 2013, 10, e83–e89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. SanMiguel, A.; Grice, E.A. Interactions between host factors and the skin microbiome. Cell Mol. Life Sci. 2015, 72, 1499–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Grice, E.A.; Kong, H.H.; Conlan, S.; Deming, C.B.; Davis, J.; Young, A.C.; Program, N.C.S.; Bouffard, G.G.; Blakesley, R.W.; Murray, P.R.; et al. Topographical and temporal diversity of the human skin microbiome. Science 2009, 324, 1190–1192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Grice, E.A.; Segre, J.A. The skin microbiome. Nat. Rev. Microbiol. 2011, 9, 244–253. [Google Scholar] [CrossRef] [PubMed]
  5. Costello, E.K.; Lauber, C.L.; Hamady, M.; Fierer, N.; Gordon, J.I.; Knight, R. Bacterial community variation in human body habitats across space and time. Science 2009, 326, 1694–1697. [Google Scholar] [CrossRef] [Green Version]
  6. Timm, C.M.; Loomis, K.; Stone, W.; Mehoke, T.; Brensinger, B.; Pellicore, M.; Staniczenko, P.P.A.; Charles, C.; Nayak, S.; Karig, D.K. Isolation and characterization of diverse microbial representatives from the human skin microbiome. Microbiome 2020, 8, 58. [Google Scholar] [CrossRef]
  7. Oh, J.; Byrd, A.L.; Deming, C.; Conlan, S.; Program, N.C.S.; Kong, H.H.; Segre, J.A. Biogeography and individuality shape function in the human skin metagenome. Nature 2014, 514, 59–64. [Google Scholar] [CrossRef] [Green Version]
  8. Oh, J.; Byrd, A.L.; Park, M.; Program, N.C.S.; Kong, H.H.; Segre, J.A. Temporal Stability of the Human Skin Microbiome. Cell 2016, 165, 854–866. [Google Scholar] [CrossRef] [Green Version]
  9. Findley, K.; Oh, J.; Yang, J.; Conlan, S.; Deming, C.; Meyer, J.A.; Schoenfeld, D.; Nomicos, E.; Park, M. Topographic diversity of fungal and bacterial communities in human skin. Nature 2013, 498, 367–370. [Google Scholar] [CrossRef]
  10. Prohic, A.; Sadikovic, T.J.; Krupalija-Fazlic, M.; Kuskunovic-Vlahovljak, S. Malassezia species in healthy skin and in dermatological conditions. Int. J. Dermatol. 2016, 55, 494–504. [Google Scholar] [CrossRef]
  11. Abdillah, A.; Khelaifia, S.; Raoult, D.; Bittar, F.; Ranque, S. Comparison of Three Skin Sampling Methods and Two Media for Culturing Malassezia Yeast. J. Fungi 2020, 6, 350. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, E.; Tanaka, T.; Tajima, M.; Tsuboi, R.; Nishikawa, A.; Sugita, T. Characterization of the skin fungal microbiota in patients with atopic dermatitis and in healthy subjects. Microbiol. Immunol. 2011, 55, 625–632. [Google Scholar] [CrossRef] [PubMed]
  13. Rafat, Z.; Hashemi, S.J.; Ahamdikia, K.; Daie Ghazvini, R.; Bazvandi, F. Study of skin and nail Candida species as a normal flora based on age groups in healthy persons in Tehran-Iran. J. Mycol. Med. 2017, 27, 501–505. [Google Scholar] [CrossRef] [PubMed]
  14. Foulongne, V.; Sauvage, V.; Hebert, C.; Dereure, O.; Cheval, J.; Gouilh, M.A.; Pariente, K.; Segondy, M.; Burguiere, A.; Manuguerra, J.C.; et al. Human skin microbiota: High diversity of DNA viruses identified on the human skin by high throughput sequencing. PLoS ONE 2012, 7, e38499. [Google Scholar] [CrossRef] [Green Version]
  15. Hannigan, G.D.; Meisel, J.S.; Tyldsley, A.S.; Zheng, Q.; Hodkinson, B.P.; SanMiguel, A.J.; Minot, S.; Bushman, F.D.; Grice, E.A. The human skin double-stranded DNA virome: Topographical and temporal diversity, genetic enrichment, and dynamic associations with the host microbiome. mBio 2015, 6, e01578-15. [Google Scholar] [CrossRef] [Green Version]
  16. Shi, B.; Bangayan, N.J.; Curd, E.; Taylor, P.A.; Gallo, R.L.; Leung, D.Y.M.; Li, H. The skin microbiome is different in pediatric versus adult atopic dermatitis. J. Allergy Clin. Immunol. 2016, 138, 1233–1236. [Google Scholar] [CrossRef] [Green Version]
  17. Jo, J.H.; Deming, C.; Kennedy, E.A.; Conlan, S.; Polley, E.C.; Ng, W.I.; Program, N.C.S.; Segre, J.A.; Kong, H.H. Diverse Human Skin Fungal Communities in Children Converge in Adulthood. J. Investig. Dermatol. 2016, 136, 2356–2363. [Google Scholar] [CrossRef] [Green Version]
  18. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The human skin microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef]
  19. Jurakic Toncic, R.; Jakasa, I.; Sun, Y.; Hurault, G.; Ljubojevic Hadzavdic, S.; Tanaka, R.J.; Pavicic, B.; Balic, A.; Zuzul, K.; Petkovic, M.; et al. Stratum corneum markers of innate and T helper cell-related immunity and their relation to the disease severity in Croatian patients with atopic dermatitis. J. Eur. Acad. Dermatol. Venereol. 2021, 35, 1186–1196. [Google Scholar] [CrossRef]
  20. Hulpusch, C.; Tremmel, K.; Hammel, G.; Bhattacharyya, M.; de Tomassi, A.; Nussbaumer, T.; Neumann, A.U.; Reiger, M.; Traidl-Hoffmann, C. Skin pH-dependent Staphylococcus aureus abundance as predictor for increasing atopic dermatitis severity. Allergy 2020, 75, 2888–2898. [Google Scholar] [CrossRef]
  21. Rehbinder, E.M.; Advocaat Endre, K.M.; Lodrup Carlsen, K.C.; Asarnoj, A.; Stensby Bains, K.E.; Berents, T.L.; Carlsen, K.H.; Gudmundsdottir, H.K.; Haugen, G.; Hedlin, G.; et al. Predicting Skin Barrier Dysfunction and Atopic Dermatitis in Early Infancy. J. Allergy Clin. Immunol. Pract. 2020, 8, 664–673.e5. [Google Scholar] [CrossRef] [PubMed]
  22. Tsakok, T.; Marrs, T.; Mohsin, M.; Baron, S.; du Toit, G.; Till, S.; Flohr, C. Does atopic dermatitis cause food allergy? A systematic review. J. Allergy Clin. Immunol. 2016, 137, 1071–1078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gustafsson, D.; Sjöberg, O.; Foucard, T. Development of allergies and asthma in infants and young children with atopic dermatitis--a prospective follow-up to 7 years of age. Allergy 2000, 55, 240–245. [Google Scholar] [CrossRef] [PubMed]
  24. Spergel, J.M.; Paller, A.S. Atopic dermatitis and the atopic march. J. Allergy Clin. Immunol. 2003, 112 (Suppl. S6), S118–S127. [Google Scholar] [CrossRef]
  25. Zheng, T.; Yu, J.; Oh, M.H.; Zhu, Z. The atopic march: Progression from atopic dermatitis to allergic rhinitis and asthma. Allergy Asthma Immunol. Res. 2011, 3, 67–73. [Google Scholar] [CrossRef] [Green Version]
  26. Schneider, L.; Hanifin, J.; Boguniewicz, M.; Eichenfield, L.F.; Spergel, J.M.; Dakovic, R.; Paller, A.S. Study of the Atopic March: Development of Atopic Comorbidities. Pediatr. Dermatol. 2016, 33, 388–398. [Google Scholar] [CrossRef] [Green Version]
  27. Bieber, T.; D’Erme, A.M.; Akdis, C.A.; Traidl-Hoffmann, C.; Lauener, R.; Schappi, G.; Schmid-Grendelmeier, P. Clinical phenotypes and endophenotypes of atopic dermatitis: Where are we, and where should we go? J. Allergy Clin. Immunol. 2017, 139 (Suppl. S4), S58–S64. [Google Scholar] [CrossRef] [Green Version]
  28. Williamson, S.; Merritt, J.; De Benedetto, A. Atopic dermatitis in the elderly: A review of clinical and pathophysiological hallmarks. Br. J. Dermatol. 2020, 182, 47–54. [Google Scholar] [CrossRef] [Green Version]
  29. Tanei, R. Atopic Dermatitis in Older Adults: A Review of Treatment Options. Drugs Aging 2020, 37, 149–160. [Google Scholar] [CrossRef] [Green Version]
  30. Hanifin, J.M.; Rajka, G. Diagnostic Features of Atopic-Dermatitis. Acta Derm. Venereol. 1980, 60, 44–47. [Google Scholar]
  31. Andersen, R.M.; Thyssen, J.P.; Maibach, H.I. Qualitative vs. quantitative atopic dermatitis criteria-in historical and present perspectives. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 604–618. [Google Scholar] [CrossRef] [PubMed]
  32. Williams, H.C.; Burney, P.G.; Hay, R.J.; Archer, C.B.; Shipley, M.J.; Hunter, J.J.; Bingham, E.A.; Finlay, A.Y.; Pembroke, A.C.; Graham-Brown, R.A.; et al. The U.K. Working Party’s Diagnostic Criteria for Atopic Dermatitis. I. Derivation of a minimum set of discriminators for atopic dermatitis. Br. J. Dermatol. 1994, 131, 383–396. [Google Scholar] [CrossRef] [PubMed]
  33. Leshem, Y.A.; Hajar, T.; Hanifin, J.M.; Simpson, E.L. What the Eczema Area and Severity Index score tells us about the severity of atopic dermatitis: An interpretability study. Br. J. Dermatol. 2015, 172, 1353–1357. [Google Scholar] [CrossRef] [PubMed]
  34. Chopra, R.; Vakharia, P.P.; Sacotte, R.; Patel, N.; Immaneni, S.; White, T.; Kantor, R.; Hsu, D.Y.; Silverberg, J.I. Relationship between EASI and SCORAD severity assessments for atopic dermatitis. J. Allergy Clin. Immunol. 2017, 140, 1708–1710.e1. [Google Scholar] [CrossRef] [Green Version]
  35. Silverberg, J.I.; Lei, D.; Yousaf, M.; Janmohamed, S.R.; Vakharia, P.P.; Chopra, R.; Chavda, R.; Gabriel, S.; Patel, K.R.; Singam, V.; et al. What are the best endpoints for Eczema Area and Severity Index and Scoring Atopic Dermatitis in clinical practice? A prospective observational study. Br. J. Dermatol. 2021, 184, 888–895. [Google Scholar] [CrossRef]
  36. Torres, T.; Ferreira, E.O.; Goncalo, M.; Mendes-Bastos, P.; Selores, M.; Filipe, P. Update on Atopic Dermatitis. Acta Med. Port 2019, 32, 606–613. [Google Scholar] [CrossRef]
  37. Kantor, R.; Silverberg, J.I. Environmental risk factors and their role in the management of atopic dermatitis. Expert Rev. Clin. Immunol. 2017, 13, 15–26. [Google Scholar] [CrossRef]
  38. Weidinger, S.; Beck, L.A.; Bieber, T.; Kabashima, K.; Irvine, A.D. Atopic dermatitis. Nat. Rev. Dis. Primers 2018, 4, 1. [Google Scholar] [CrossRef]
  39. Nedoszytko, B.; Reszka, E.; Gutowska-Owsiak, D.; Trzeciak, M.; Lange, M.; Jarczak, J.; Niedoszytko, M.; Jablonska, E.; Romantowski, J.; Strapagiel, D.; et al. Genetic and Epigenetic Aspects of Atopic Dermatitis. Int. J. Mol. Sci. 2020, 21, 6484. [Google Scholar] [CrossRef]
  40. Weidinger, S.; Illig, T.; Baurecht, H.; Irvine, A.D.; Rodriguez, E.; Diaz-Lacava, A.; Klopp, N.; Wagenpfeil, S.; Zhao, Y.; Liao, H.; et al. Loss-of-function variations within the filaggrin gene predispose for atopic dermatitis with allergic sensitizations. J. Allergy Clin. Immunol. 2006, 118, 214–219. [Google Scholar] [CrossRef]
  41. Kawasaki, H.; Nagao, K.; Kubo, A.; Hata, T.; Shimizu, A.; Mizuno, H.; Yamada, T.; Amagai, M. Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrin-null mice. J. Allergy Clin. Immunol. 2012, 129, 1538–1546.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. van Smeden, J.; Bouwstra, J.A. Stratum Corneum Lipids: Their Role for the Skin Barrier Function in Healthy Subjects and Atopic Dermatitis Patients. Curr. Probl. Dermatol. 2016, 49, 8–26. [Google Scholar]
  43. Salimi, M.; Barlow, J.L.; Saunders, S.P.; Xue, L.; Gutowska-Owsiak, D.; Wang, X.; Huang, L.C.; Johnson, D.; Scanlon, S.T.; McKenzie, A.N.; et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J. Exp. Med. 2013, 210, 2939–2950. [Google Scholar] [CrossRef] [PubMed]
  44. Tsakok, T.; Woolf, R.; Smith, C.H.; Weidinger, S.; Flohr, C. Atopic dermatitis: The skin barrier and beyond. Br. J. Dermatol. 2019, 180, 464–474. [Google Scholar] [CrossRef] [PubMed]
  45. Malik, K.; Heitmiller, K.D.; Czarnowicki, T. An Update on the Pathophysiology of Atopic Dermatitis. Dermatol. Clin. 2017, 35, 317–326. [Google Scholar] [CrossRef] [PubMed]
  46. Mashiko, S.; Mehta, H.; Bissonnette, R.; Sarfati, M. Increased frequencies of basophils, type 2 innate lymphoid cells and Th2 cells in skin of patients with atopic dermatitis but not psoriasis. J. Dermatol. Sci. 2017, 88, 167–174. [Google Scholar] [CrossRef] [Green Version]
  47. Su, C.; Yang, T.; Wu, Z.; Zhong, J.; Huang, Y.; Huang, T.; Zheng, E. Differentiation of T-helper cells in distinct phases of atopic dermatitis involves Th1/Th2 and Th17/Treg. Eur. J. Inflamm. 2017, 15, 46–52. [Google Scholar] [CrossRef] [Green Version]
  48. Novak, N.; Bieber, T.; Hoffmann, M.; Folster-Holst, R.; Homey, B.; Werfel, T.; Sager, A.; Zuberbier, T. Efficacy and safety of subcutaneous allergen-specific immunotherapy with depigmented polymerized mite extract in atopic dermatitis. J. Allergy Clin. Immunol. 2012, 130, 925–931.e4. [Google Scholar] [CrossRef]
  49. Wilson, S.R.; The, L.; Batia, L.M.; Beattie, K.; Katibah, G.E.; McClain, S.P.; Pellegrino, M.; Estandian, D.M.; Bautista, D.M. The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell 2013, 155, 285–295. [Google Scholar] [CrossRef] [Green Version]
  50. Oetjen, L.K.; Mack, M.R.; Feng, J.; Whelan, T.M.; Niu, H.; Guo, C.J.; Chen, S.; Trier, A.M.; Xu, A.Z.; Tripathi, S.V.; et al. Sensory Neurons Co-opt Classical Immune Signaling Pathways to Mediate Chronic Itch. Cell 2017, 171, 217–228.e13. [Google Scholar] [CrossRef] [Green Version]
  51. Kong, H.H.; Oh, J.; Deming, C.; Conlan, S.; Grice, E.A.; Beatson, M.A.; Nomicos, E.; Polley, E.C.; Komarow, H.D.; Program, N.C.S.; et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012, 22, 850–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Geoghegan, J.A.; Irvine, A.D.; Foster, T.J. Staphylococcus aureus and Atopic Dermatitis: A Complex and Evolving Relationship. Trends Microbiol. 2018, 26, 484–497. [Google Scholar] [CrossRef] [PubMed]
  53. Williams, M.R.; Nakatsuji, T.; Gallo, R.L. Staphylococcus aureus: Master Manipulator of the Skin. Cell Host Microbe 2017, 22, 579–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Fyhrquist, N.; Muirhead, G.; Prast-Nielsen, S.; Jeanmougin, M.; Olah, P.; Skoog, T.; Jules-Clement, G.; Feld, M.; Barrientos-Somarribas, M.; Sinkko, H.; et al. Microbe-host interplay in atopic dermatitis and psoriasis. Nat. Commun. 2019, 10, 4703. [Google Scholar] [CrossRef] [Green Version]
  55. Totte, J.E.; van der Feltz, W.T.; Hennekam, M.; van Belkum, A.; van Zuuren, E.J.; Pasmans, S.G. Prevalence and odds of Staphylococcus aureus carriage in atopic dermatitis: A systematic review and meta-analysis. Br. J. Dermatol. 2016, 175, 687–695. [Google Scholar] [CrossRef]
  56. Di Domenico, E.G.; Cavallo, I.; Bordignon, V.; Prignano, G.; Sperduti, I.; Gurtner, A.; Trento, E.; Toma, L.; Pimpinelli, F.; Capitanio, B.; et al. Inflammatory cytokines and biofilm production sustain Staphylococcus aureus outgrowth and persistence: A pivotal interplay in the pathogenesis of Atopic Dermatitis. Sci. Rep. 2018, 8, 9573. [Google Scholar] [CrossRef]
  57. Fleury, O.M.; McAleer, M.A.; Feuillie, C.; Formosa-Dague, C.; Sansevere, E.; Bennett, D.E.; Towell, A.M.; McLean, W.H.I.; Kezic, S.; Robinson, D.A.; et al. Clumping Factor B Promotes Adherence of Staphylococcus aureus to Corneocytes in Atopic Dermatitis. Infect. Immun. 2017, 85, e00994-16. [Google Scholar] [CrossRef] [Green Version]
  58. Ong, P.Y.; Ohtake, T.; Brandt, C.; Strickland, I.; Boguniewicz, M.; Ganz, T.; Gallo, R.L.; Leung, D.Y.M. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N. Engl. J. Med. 2002, 347, 1151–1160. [Google Scholar] [CrossRef] [Green Version]
  59. Hata, T.R.; Kotol, P.; Boguniewicz, M.; Taylor, P.; Paik, A.; Jackson, M.; Nguyen, M.; Kabigting, F.; Miller, J.; Gerber, M.; et al. History of eczema herpeticum is associated with the inability to induce human beta-defensin (HBD)-2, HBD-3 and cathelicidin in the skin of patients with atopic dermatitis. Br. J. Dermatol. 2010, 163, 659–661. [Google Scholar] [CrossRef] [Green Version]
  60. Feuillie, C.; Vitry, P.; McAleer, M.A.; Kezic, S.; Irvine, A.D.; Geoghegan, J.A.; Dufrene, Y.F. Adhesion of Staphylococcus aureus to Corneocytes from Atopic Dermatitis Patients Is Controlled by Natural Moisturizing Factor Levels. mBio 2018, 9, e01184-18. [Google Scholar] [CrossRef] [Green Version]
  61. Nakatsuji, T.; Chen, T.H.; Two, A.M.; Chun, K.A.; Narala, S.; Geha, R.S.; Hata, T.R.; Gallo, R.L. Staphylococcus aureus Exploits Epidermal Barrier Defects in Atopic Dermatitis to Trigger Cytokine Expression. J. Investig. Dermatol. 2016, 136, 2192–2200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Williams, M.R.; Nakatsuji, T.; Sanford, J.A.; Vrbanac, A.F.; Gallo, R.L. Staphylococcus aureus Induces Increased Serine Protease Activity in Keratinocytes. J. Investig. Dermatol. 2017, 137, 377–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Hon, K.L.; Tsang, Y.C.; Pong, N.H.; Leung, T.F.; Ip, M. Exploring Staphylococcus epidermidis in atopic eczema: Friend or foe? Clin. Exp. Dermatol. 2016, 41, 659–663. [Google Scholar] [CrossRef] [PubMed]
  64. Fitz-Gibbon, S.; Tomida, S.; Chiu, B.H.; Nguyen, L.; Du, C.; Liu, M.; Elashoff, D.; Erfe, M.C.; Loncaric, A.; Kim, J.; et al. Propionibacterium acnes strain populations in the human skin microbiome associated with acne. J. Investig. Dermatol. 2013, 133, 2152–2160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lo, C.W.; Lai, Y.K.; Liu, Y.T.; Gallo, R.L.; Huang, C.M. Staphylococcus aureus hijacks a skin commensal to intensify its virulence: Immunization targeting beta-hemolysin and CAMP factor. J. Invest. Dermatol. 2011, 131, 401–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wollenberg, M.S.; Claesen, J.; Escapa, I.F.; Aldridge, K.L.; Fischbach, M.A.; Lemon, K.P. Propionibacterium-produced coproporphyrin III induces Staphylococcus aureus aggregation and biofilm formation. mBio 2014, 5, e01286-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Kim, J.E.; Kim, H.S. Microbiome of the Skin and Gut in Atopic Dermatitis (AD): Understanding the Pathophysiology and Finding Novel Management Strategies. J. Clin. Med. 2019, 8, 444. [Google Scholar] [CrossRef] [Green Version]
  68. Nowicka, D.; Nawrot, U. Contribution of Malassezia spp. to the development of atopic dermatitis. Mycoses 2019, 62, 588–596. [Google Scholar] [CrossRef]
  69. Scharschmidt, T.C.; Vasquez, K.S.; Truong, H.A.; Gearty, S.V.; Pauli, M.L.; Nosbaum, A.; Gratz, I.K.; Otto, M.; Moon, J.J.; Liese, J.; et al. A Wave of Regulatory T Cells into Neonatal Skin Mediates Tolerance to Commensal Microbes. Immunity 2015, 43, 1011–1021. [Google Scholar] [CrossRef] [Green Version]
  70. Kennedy, E.A.; Connolly, J.; Hourihane, J.O.; Fallon, P.G.; McLean, W.H.I.; Murray, D.; Jo, J.H.; Segre, J.A.; Kong, H.H.; Irvine, A.D. Skin microbiome before development of atopic dermatitis: Early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year. J. Allergy Clin. Immunol. 2017, 139, 166–172. [Google Scholar] [CrossRef] [Green Version]
  71. Iwase, T.; Uehara, Y.; Shinji, H.; Tajima, A.; Seo, H.; Takada, K.; Agata, T.; Mizunoe, Y. Staphylococcus epidermidis Esp inhibits Staphylococcus aureus biofilm formation and nasal colonization. Nature 2010, 465, 346–349. [Google Scholar] [CrossRef] [PubMed]
  72. Sugimoto, S.; Iwamoto, T.; Takada, K.; Okuda, K.; Tajima, A.; Iwase, T.; Mizunoe, Y. Staphylococcus epidermidis Esp degrades specific proteins associated with Staphylococcus aureus biofilm formation and host-pathogen interaction. J. Bacteriol. 2013, 195, 1645–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zipperer, A.; Konnerth, M.C.; Laux, C.; Berscheid, A.; Janek, D.; Weidenmaier, C.; Burian, M.; Schilling, N.A.; Slavetinsky, C.; Marschal, M.; et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature 2016, 535, 511–516. [Google Scholar] [CrossRef] [PubMed]
  74. Nakatsuji, T.; Hata, T.R.; Tong, Y.; Cheng, J.Y.; Shafiq, F.; Butcher, A.M.; Salem, S.S.; Brinton, S.L.; Rudman Spergel, A.K.; Johnson, K.; et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat. Med. 2021, 27, 700–709. [Google Scholar] [CrossRef]
  75. Ramsey, M.M.; Freire, M.O.; Gabrilska, R.A.; Rumbaugh, K.P.; Lemon, K.P. Staphylococcus aureus Shifts toward Commensalism in Response to Corynebacterium Species. Front. Microbiol. 2016, 7, 1230. [Google Scholar] [CrossRef] [Green Version]
  76. Shu, M.; Wang, Y.; Yu, J.; Kuo, S.; Coda, A.; Jiang, Y.; Gallo, R.L.; Huang, C.M. Fermentation of Propionibacterium acnes, a commensal bacterium in the human skin microbiome, as skin probiotics against methicillin-resistant Staphylococcus aureus. PLoS ONE 2013, 8, e55380. [Google Scholar] [CrossRef] [Green Version]
  77. Li, H.; Goh, B.N.; Teh, W.K.; Jiang, Z.; Goh, J.P.Z.; Goh, A.; Wu, G.; Hoon, S.S.; Raida, M.; Camattari, A.; et al. Skin Commensal Malassezia globosa Secreted Protease Attenuates Staphylococcus aureus Biofilm Formation. J. Investig. Dermatol. 2018, 138, 1137–1145. [Google Scholar] [CrossRef]
  78. Wollenberg, A.; Barbarot, S.; Bieber, T.; Christen-Zaech, S.; Deleuran, M.; Fink-Wagner, A.; Gieler, U.; Girolomoni, G.; Lau, S.; Muraro, A.; et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: Part I. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 657–682. [Google Scholar] [CrossRef] [Green Version]
  79. Kim, B.E.; Leung, D.Y.M. Significance of Skin Barrier Dysfunction in Atopic Dermatitis. Allergy Asthma Immunol. Res. 2018, 10, 207–215. [Google Scholar] [CrossRef] [Green Version]
  80. Goh, S.W.; Jamil, A.; Safian, N.; Md Nor, N.; Muhammad, N.; Saharudin, N.L. A randomized half-body, double blind, controlled trial on the effects of a pH-modified moisturizer vs. standard moisturizer in mild to moderate atopic dermatitis. An. Bras. Dermatol. 2020, 95, 320–325. [Google Scholar] [CrossRef]
  81. Flores, G.E.; Seite, S.; Henley, J.B.; Martin, R.; Zelenkova, H.; Aguilar, L.; Fierer, N. Microbiome of Affected and Unaffected Skin of Patients With Atopic Dermatitis Before and After Emollient Treatment. J. Drugs Dermatol. 2014, 13, 1365–1372. [Google Scholar]
  82. Drucker, A.M.; Eyerich, K.; de Bruin-Weller, M.S.; Thyssen, J.P.; Spuls, P.I.; Irvine, A.D.; Girolomoni, G.; Dhar, S.; Flohr, C.; Murrell, D.F.; et al. Use of systemic corticosteroids for atopic dermatitis: International Eczema Council consensus statement. Br. J. Dermatol. 2018, 178, 768–775. [Google Scholar] [CrossRef]
  83. Norris, D.A. Mechanisms of action of topical therapies and the rationale for combination therapy. J. Am. Acad. Dermatol. 2005, 53 (Suppl. S1), S17–S25. [Google Scholar] [CrossRef] [PubMed]
  84. Luger, T.; Paller, A.S.; Irvine, A.D.; Sidbury, R.; Eichenfield, L.F.; Werfel, T.; Bieber, T. Topical therapy of atopic dermatitis with a focus on pimecrolimus. J. Eur. Acad. Dermatol. Venereol. 2021, 35, 1505–1518. [Google Scholar] [CrossRef] [PubMed]
  85. Wongpiyabovorn, J.; Soonthornchai, W.; Wilantho, A.; Palasuk, M.; Payungporn, S.; Sodsai, P.; Poomipak, W.; Weschawalit, S.; Ruchusatsawat, K.; Baillie, G.S.; et al. Effect of tacrolimus on skin microbiome in atopic dermatitis. Allergy 2019, 74, 1400–1406. [Google Scholar] [CrossRef] [Green Version]
  86. Bessa, G.R.; Quinto, V.P.; Machado, D.C.; Lipnharski, C.; Weber, M.B.; Bonamigo, R.R.; D’Azevedo, P.A. Staphylococcus aureus resistance to topical antimicrobials in atopic dermatitis. An. Bras. Dermatol. 2016, 91, 604–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Kwon, S.; Choi, J.Y.; Shin, J.W.; Huh, C.H.; Park, K.C.; Du, M.H.; Yoon, S.; Na, J.I. Changes in Lesional and Non-lesional Skin Microbiome During Treatment of Atopic Dermatitis. Acta Derm. Venereol. 2019, 99, 284–290. [Google Scholar] [CrossRef] [Green Version]
  88. Wollenberg, A.; Barbarot, S.; Bieber, T.; Christen-Zaech, S.; Deleuran, M.; Fink-Wagner, A.; Gieler, U.; Girolomoni, G.; Lau, S.; Muraro, A.; et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: Part II. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 850–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Kim, W.K.; Jang, Y.J.; Han, D.H.; Jeon, K.; Lee, C.; Han, H.S.; Ko, G. Lactobacillus paracasei KBL382 administration attenuates atopic dermatitis by modulating immune response and gut microbiota. Gut Microbes 2020, 12, 1819156. [Google Scholar] [CrossRef]
  90. Kwon, M.S.; Lim, S.K.; Jang, J.Y.; Lee, J.; Park, H.K.; Kim, N.; Yun, M.; Shin, M.Y.; Jo, H.E.; Oh, Y.J.; et al. Lactobacillus sakei WIKIM30 Ameliorates Atopic Dermatitis-Like Skin Lesions by Inducing Regulatory T Cells and Altering Gut Microbiota Structure in Mice. Front. Immunol. 2018, 9, 1905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Lee, S.H.; Yoon, J.M.; Kim, Y.H.; Jeong, D.G.; Park, S.; Kang, D.J. Therapeutic effect of tyndallized Lactobacillus rhamnosus IDCC 3201 on atopic dermatitis mediated by down-regulation of immunoglobulin E in NC/Nga mice. Microbiol. Immunol. 2016, 60, 468–476. [Google Scholar] [CrossRef] [PubMed]
  92. Kim, I.S.; Lee, S.H.; Kwon, Y.M.; Adhikari, B.; Kim, J.A.; Yu, D.Y.; Kim, G.I.; Lim, J.M.; Kim, S.H.; Lee, S.S.; et al. Oral Administration of beta-Glucan and Lactobacillus plantarum Alleviates Atopic Dermatitis-Like Symptoms. J. Microbiol. Biotechnol. 2019, 29, 1693–1706. [Google Scholar] [CrossRef] [PubMed]
  93. Rosenfeldt, V.; Benfeldt, E.; Nielsen, S.D.; Michaelsen, K.F.; Jeppesen, D.L.; Valerius, N.H.; Paerregaard, A. Effect of probiotic Lactobacillus strains in children with atopic dermatitis. J. Allergy Clin. Immunol. 2003, 111, 389–395. [Google Scholar] [CrossRef]
  94. Wickens, K.; Black, P.; Stanley, T.V.; Mitchell, E.; Barthow, C.; Fitzharris, P.; Purdie, G.; Crane, J. A protective effect of Lactobacillus rhamnosus HN001 against eczema in the first 2 years of life persists to age 4 years. Clin. Exp. Allergy 2012, 42, 1071–1079. [Google Scholar] [CrossRef] [PubMed]
  95. Wickens, K.; Stanley, T.V.; Mitchell, E.A.; Barthow, C.; Fitzharris, P.; Purdie, G.; Siebers, R.; Black, P.N.; Crane, J. Early supplementation with Lactobacillus rhamnosus HN001 reduces eczema prevalence to 6 years: Does it also reduce atopic sensitization? Clin. Exp. Allergy 2013, 43, 1048–1057. [Google Scholar] [CrossRef] [PubMed]
  96. Wu, Y.J.; Wu, W.F.; Hung, C.W.; Ku, M.S.; Liao, P.F.; Sun, H.L.; Lu, K.H.; Sheu, J.N.; Lue, K.H. Evaluation of efficacy and safety of Lactobacillus rhamnosus in children aged 4-48 months with atopic dermatitis: An 8-week, double-blind, randomized, placebo-controlled study. J. Microbiol. Immunol. Infect. 2017, 50, 684–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Han, Y.; Kim, B.; Ban, J.; Lee, J.; Kim, B.J.; Choi, B.S.; Hwang, S.; Ahn, K.; Kim, J. A randomized trial of Lactobacillus plantarum CJLP133 for the treatment of atopic dermatitis. Pediatr. Allergy Immunol. 2012, 23, 667–673. [Google Scholar] [CrossRef]
  98. Weston, S.; Halbert, A.; Richmond, P.; Prescott, S.L. Effects of probiotics on atopic dermatitis: A randomised controlled trial. Arch. Dis. Child. 2005, 90, 892–897. [Google Scholar] [CrossRef]
  99. Niccoli, A.A.; Artesi, A.L.; Candio, F.; Ceccarelli, S.; Cozzali, R.; Ferraro, L.; Fiumana, D.; Mencacci, M.; Morlupo, M.; Pazzelli, P.; et al. Preliminary Results on Clinical Effects of Probiotic Lactobacillus salivarius LS01 in Children Affected by Atopic Dermatitis. J. Clin. Gastroenterol. 2014, 48, S34–S36. [Google Scholar] [CrossRef]
  100. Matsumoto, M.; Ebata, T.; Hirooka, J.; Hosoya, R.; Inoue, N.; Itami, S.; Tsuji, K.; Yaginuma, T.; Muramatsu, K.; Nakamura, A.; et al. Antipruritic effects of the probiotic strain LKM512 in adults with atopic dermatitis. Ann. Allergy Asthma Immunol. 2014, 113, 209–216.e7. [Google Scholar] [CrossRef]
  101. Navarro-Lopez, V.; Ramirez-Bosca, A.; Ramon-Vidal, D.; Ruzafa-Costas, B.; Genoves-Martinez, S.; Chenoll-Cuadros, E.; Carrion-Gutierrez, M.; Horga de la Parte, J.; Prieto-Merino, D.; Codoner-Cortes, F.M. Effect of Oral Administration of a Mixture of Probiotic Strains on SCORAD Index and Use of Topical Steroids in Young Patients With Moderate Atopic Dermatitis: A Randomized Clinical Trial. JAMA Dermatol. 2018, 154, 37–43. [Google Scholar] [CrossRef] [PubMed]
  102. Lise, M.; Mayer, I.; Silveira, M. Use of probiotics in atopic dermatitis. Rev. Assoc. Med. Bras. 2018, 64, 997–1001. [Google Scholar] [CrossRef] [PubMed]
  103. Kim, S.O.; Ah, Y.M.; Yu, Y.M.; Choi, K.H.; Shin, W.G.; Lee, J.Y. Effects of probiotics for the treatment of atopic dermatitis: A meta-analysis of randomized controlled trials. Ann. Allergy Asthma Immunol. 2014, 113, 217–226. [Google Scholar] [CrossRef]
  104. Nakatsuji, T.; Chen, T.H.; Narala, S.; Chun, K.A.; Two, A.M.; Yun, T.; Shafiq, F.; Kotol, P.F.; Bouslimani, A.; Melnik, A.V.; et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci. Transl. Med. 2017, 9, eaah4680. [Google Scholar] [CrossRef] [Green Version]
  105. Myles, I.A.; Earland, N.J.; Anderson, E.D.; Moore, I.N.; Kieh, M.D.; Williams, K.W.; Saleem, A.; Fontecilla, N.M.; Welch, P.A.; Darnell, D.A.; et al. First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis. JCI Insight 2018, 3, e120608. [Google Scholar] [CrossRef] [Green Version]
  106. Blanchet-Rethore, S.; Bourdes, V.; Mercenier, A.; Haddar, C.H.; Verhoeven, P.O.; Andres, P. Effect of a lotion containing the heat-treated probiotic strain Lactobacillus johnsonii NCC 533 on Staphylococcus aureus colonization in atopic dermatitis. Clin. Cosmet Investig. Dermatol. 2017, 10, 249–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Di Marzio, L.; Centi, C.; Cinque, B.; Masci, S.; Giuliani, M.; Arcieri, A.; Zicari, L.; De Simone, C.; Cifone, M.G. Effect of the lactic acid bacterium Streptococcus thermophilus on stratum corneum ceramide levels and signs and symptoms of atopic dermatitis patients. Exp. Dermatol. 2003, 12, 615–620. [Google Scholar] [CrossRef]
  108. Gueniche, A.; Knaudt, B.; Schuck, E.; Volz, T.; Bastien, P.; Martin, R.; Rocken, M.; Breton, L.; Biedermann, T. Effects of nonpathogenic gram-negative bacterium Vitreoscilla filiformis lysate on atopic dermatitis: A prospective, randomized, double-blind, placebo-controlled clinical study. Br. J. Dermatol. 2008, 159, 1357–1363. [Google Scholar] [CrossRef]
  109. Chang, Y.S.; Trivedi, M.K.; Jha, A.; Lin, Y.F.; Dimaano, L.; Garcia-Romero, M.T. Synbiotics for Prevention and Treatment of Atopic Dermatitis: A Meta-analysis of Randomized Clinical Trials. JAMA Pediatr. 2016, 170, 236–242. [Google Scholar] [CrossRef]
  110. Passeron, T.; Lacour, J.P.; Fontas, E.; Ortonne, J.P. Prebiotics and synbiotics: Two promising approaches for the treatment of atopic dermatitis in children above 2 years. Allergy 2006, 61, 431–437. [Google Scholar] [CrossRef]
  111. Aldaghi, M.; Tehrani, H.; Karrabi, M.; Abadi, F.S.; Sahebkar, M. The effect of multistrain synbiotic and vitamin D3 supplements on the severity of atopic dermatitis among infants under 1 year of age: A double-blind, randomized clinical trial study. J. Dermatol. Treat. 2020. [Google Scholar] [CrossRef]
  112. Pharmabiotic Research Institute. Available online: https://www.pharmabiotic.org/#mmps (accessed on 17 March 2022).
  113. Cordaillat-Simmons, M.; Rouanet, A.; Pot, B. Live biotherapeutic products: The importance of a defined regulatory framework. Exp. Mol. Med. 2020, 52, 1397–1406. [Google Scholar] [CrossRef] [PubMed]
  114. Margulis, L.S.; Fester, R. Symbiosis as a Source of Evolutionary Innovation: Speciation and Morphogenesis; MIT Press: Cambridge, MA, USA, 1991; 454p. [Google Scholar]
  115. Theis, K.R.; Dheilly, N.M.; Klassen, J.L.; Brucker, R.M.; Baines, J.F.; Bosch, T.C.; Cryan, J.F.; Gilbert, S.F.; Goodnight, C.J.; Lloyd, E.A.; et al. Getting the Hologenome Concept Right: An Eco-Evolutionary Framework for Hosts and Their Microbiomes. mSystems 2016, 1, e00028-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation of human skin structure consisting of three main layers: epidermis, dermis and subcutis. The outermost skin layer, epidermis, is composed of terminally differentiated keratinocytes that enable continuous skin renewing, held together by corneodesmosomes and mortar lipids. Epidermis is supported by the collagen-bound dermis that provides a home for nerves, blood, lymph vessels, mast cells and other structures (i.e., sweat glands, hair follicles), and subcutis consisting of adipose tissue. Skin regions vary in terms of topography, temperature, salt content and acidity (pH) and are, based on their features, categorized into three major groups: moist sebaceous and dry.
Figure 1. Schematic representation of human skin structure consisting of three main layers: epidermis, dermis and subcutis. The outermost skin layer, epidermis, is composed of terminally differentiated keratinocytes that enable continuous skin renewing, held together by corneodesmosomes and mortar lipids. Epidermis is supported by the collagen-bound dermis that provides a home for nerves, blood, lymph vessels, mast cells and other structures (i.e., sweat glands, hair follicles), and subcutis consisting of adipose tissue. Skin regions vary in terms of topography, temperature, salt content and acidity (pH) and are, based on their features, categorized into three major groups: moist sebaceous and dry.
Ijms 23 03503 g001
Figure 2. Epithelial barrier dysfunction, immune dysregulation and skin microbiota dysbiosis in initiation and progression of AD. Epithelial barrier dysfunction and stress from environmental and mechanical factors lead to skin barrier damage and enhanced epidermal permeability, which in turn increases microbial and allergen contact with the cutaneous immune system. Damaged keratinocytes activate immune mechanisms by releasing proinflammatory cytokines (IL-1β, TSLP, IL-25, IL-33) and chemokines, mobilizing innate lymphocyte subsets and skin-resident dendritic cells (DCs). DCs attract and prime naive T-cells, promoting TH2/TH22 cell responses and inducing inflammation process. Type 2 cytokines (IL-4, IL-5, IL-13, IL-25) drive the inflammation, recruiting and activating other types of immune cells, such as eosinophils, mast cells and B-cells. The secreted molecules and proinflammatory cytokines act directly on cutaneous nerves and contribute to pruritus. Moreover, the inflammation further disrupts skin barrier and favors colonization by pathogens (S. aureus), additionally inducing keratinocyte damage and boosting TH2-type response, thus supporting the disease cycle. The activation of TH1/TH17 cell responses in chronic disease induce tissue remodeling, increasing skin thickness and lichenification. IDEC—inflammatory dendritic epidermal cell, ILC2—group 2 innate lymphoid cell, LC—Langerhans cell, TH1—TH1 cell, TH2—TH2 cell, TH17—TH17 cell, TH22—TH22 cell.
Figure 2. Epithelial barrier dysfunction, immune dysregulation and skin microbiota dysbiosis in initiation and progression of AD. Epithelial barrier dysfunction and stress from environmental and mechanical factors lead to skin barrier damage and enhanced epidermal permeability, which in turn increases microbial and allergen contact with the cutaneous immune system. Damaged keratinocytes activate immune mechanisms by releasing proinflammatory cytokines (IL-1β, TSLP, IL-25, IL-33) and chemokines, mobilizing innate lymphocyte subsets and skin-resident dendritic cells (DCs). DCs attract and prime naive T-cells, promoting TH2/TH22 cell responses and inducing inflammation process. Type 2 cytokines (IL-4, IL-5, IL-13, IL-25) drive the inflammation, recruiting and activating other types of immune cells, such as eosinophils, mast cells and B-cells. The secreted molecules and proinflammatory cytokines act directly on cutaneous nerves and contribute to pruritus. Moreover, the inflammation further disrupts skin barrier and favors colonization by pathogens (S. aureus), additionally inducing keratinocyte damage and boosting TH2-type response, thus supporting the disease cycle. The activation of TH1/TH17 cell responses in chronic disease induce tissue remodeling, increasing skin thickness and lichenification. IDEC—inflammatory dendritic epidermal cell, ILC2—group 2 innate lymphoid cell, LC—Langerhans cell, TH1—TH1 cell, TH2—TH2 cell, TH17—TH17 cell, TH22—TH22 cell.
Ijms 23 03503 g002
Figure 3. Comparison of microbiota composition between healthy skin and skin affected by AD.
Figure 3. Comparison of microbiota composition between healthy skin and skin affected by AD.
Ijms 23 03503 g003
Figure 4. Interplay between Staphylococcus aureus and skin microbiota. Several members of Staphylococcus genus inhibit S. aureus growth and biofilm formation. Staphylococcus lugdunensis and S. hominis suppress colonization of S. aureus by secreting antibiotics and lantibiotics. S. epidermidis induces keratinocytes to produce anti-microbial peptides (AMPs) to eradicate S. aureus, as well as produces protease glutamyl endopeptidase (Esp) which inhibits formation of S. aureus biofilm. The MgSAP1 protease secreted by Malassezia globosa was shown to have a similar effect of disrupting S. aureus biofilm. In contrast, Cutibacterium acnes and its corpoporphyrin III molecule promote S. aureus activity and aggregation, thus inducing S. aureus biofilm formation.
Figure 4. Interplay between Staphylococcus aureus and skin microbiota. Several members of Staphylococcus genus inhibit S. aureus growth and biofilm formation. Staphylococcus lugdunensis and S. hominis suppress colonization of S. aureus by secreting antibiotics and lantibiotics. S. epidermidis induces keratinocytes to produce anti-microbial peptides (AMPs) to eradicate S. aureus, as well as produces protease glutamyl endopeptidase (Esp) which inhibits formation of S. aureus biofilm. The MgSAP1 protease secreted by Malassezia globosa was shown to have a similar effect of disrupting S. aureus biofilm. In contrast, Cutibacterium acnes and its corpoporphyrin III molecule promote S. aureus activity and aggregation, thus inducing S. aureus biofilm formation.
Ijms 23 03503 g004
Table 1. Studies using probiotics and prebiotics in management of AD.
Table 1. Studies using probiotics and prebiotics in management of AD.
Study, YearStudy TypeBacterial StrainAdministrationAnimal ModelOutcome Summary
Kim et al., 2020 [93]mouse modelL. paracasei KBL382oralNC/Nga mice
DFE- and DNCB-induced AD
modulation of the immune response and gut microbiota
Kwon et al., 2018 [94]mouse modelL. sakei WIKIM30oralBALB/c mice
DNCB-induced AD
stimulation of Treg cell generation and suppression of TH2 inflammatory response, restoring the balance of gut microbiota
Lee et al., 2016 [95]mouse modelL. rhamnosus IDCC 3201 tyndallizate (RHT3201)oralNC/Nga mice
DFE-induced AD
less severe AD symptoms in comparison to controls, dose-dependent reductions in dermatitis scores
Kim et al., 2019 [96]rat/mouse modelsL. plantarum LM1004oralSprague-Dawley rats, ddY mice
histamine-induced AD
DNFB-induced AD
inhibition of TH2 cell responses and activation of Treg immunoregulatory functions, increase of relative abundance of butyrate-generating microorganisms in the gut
Study, YearStudy TypeBacterial StrainAdministrationParticipants (Age)Outcome Summary
Rosenfeldt et al., 2003 [97]humanlyophilized L. rhamnosus 19070-2
and L. reuteri DSM 122460
oralchildren
(1–13 y)
moderate improvement in the clinical severity of eczema
Wickens et al., 2012 [98]humanL. rhamnosusoralinfantsreduced eczema prevalence
Wickens et al., 2013 [99]humanL. rhamnosusoralchildren
(<6 y)
significantly reduced cumulative eczema prevalence, decrease in SCORAD values and atopic sensitization
Wu et al., 2015 [100]humanL. rhamnosusoralchildren
(4–48 mos.)
decrease of SCORAD values and disease intensity
Han et al., 2012 [101]humanL. plantarum CJLP133oralchildren
(12 mos.–13 y)
decrease of SCORAD values, IFN-γ and IL-4
Weston et al., 2005 [102]humanL. fermentum VRI-033 PCCoralchildren
(6–18 mos.)
change in AD severity compared to placebo-treated individuals
Niccoli et al., 2014 [103]humanL. salivarious LS01oralchildrendecrease of SCORAD values and significant improvement in itching intensity, both therapy benefits persisting after suspension of treatment
Matsumoto et al., 2014 [104]humanB. animalis subsp. lactis LKM512oraladultsalleviated itch in AD patients and considerably improved the quality-of-life scores
Navarro-Lopez et al., 2018 [105]humanB. lactis CECT 8145, B. longum CECT 7347, and L. casei CECT 9104oralchildren
(4–17 y)
decrease of SCORAD values in patients with moderate AD
Lise et al., 2018 [106]humanB. lactis HN019, L. acidophilus NCFM, L. rhamnosus HN001
and L. paracasei LPC-37
oralchildrenevident response in treating severe AD with significant change in AD severity scores
Kim et al., 2014 [107]humanLactobacillus and Bifidobacterium
species
oralchildren and adults
(1 mo.–65 y)
decrease of SCORAD values
Nakatsuji et al., 2017 [108]humantopical application of commensal skin bacteriatopicaladultsprotective effect against pathogen species (reduced S. aureus colonization due to selective AMPs secreted by commensal CoNS), improvement of clinical symptoms and decreased inflammation
Nakatsuji et al., 2021 [80]humanS. hominis A9 (ShA9)topicaladultsfewer adverse events associated with AD, inhibited expression of mRNA for psmα
Myles et al., 2018 [109]humanR. mucosatopicalchildren and adultssignificant decrease in SCORAD and pruritus, reduction in disease severity and no adverse effects or complications
Blanchet-Rethoré et al., 2017 [110]humanheat-treated
L. johnsonii
topicaladultsclinical improvement of AD symptoms in patients with moderate AD
Di Marzio et al., 2003 [111]humansonicated S. thermophilustopicaladultssignificant improvement in skin barrier integrity, erythema, scaling and pruritus
Gueniche et al., 2008 [112]humanlysate of V. filiformistopicalchildren and adultsclinical improvement in patients with AD, decreased SCORAD values and pruritus
Chang et al., 2016 [113]humanmultiple strains of bacteriatopicalchildren
(>1 y)
decrease of SCORAD values
Passeron et al., 2006 [114]humanL. rhamnosus Lcr35 plus prebioticstopicalchildren
(>2 y)
improved AD symptoms and decreased SCORAD values
Aldaghi et al., 2020 [115]humanL. rhamnosus, L. euteri and B. infantis or vitamin D3topicalinfantssignificantly decreased SCORAD values
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hrestak, D.; Matijašić, M.; Čipčić Paljetak, H.; Ledić Drvar, D.; Ljubojević Hadžavdić, S.; Perić, M. Skin Microbiota in Atopic Dermatitis. Int. J. Mol. Sci. 2022, 23, 3503. https://doi.org/10.3390/ijms23073503

AMA Style

Hrestak D, Matijašić M, Čipčić Paljetak H, Ledić Drvar D, Ljubojević Hadžavdić S, Perić M. Skin Microbiota in Atopic Dermatitis. International Journal of Molecular Sciences. 2022; 23(7):3503. https://doi.org/10.3390/ijms23073503

Chicago/Turabian Style

Hrestak, Dora, Mario Matijašić, Hana Čipčić Paljetak, Daniela Ledić Drvar, Suzana Ljubojević Hadžavdić, and Mihaela Perić. 2022. "Skin Microbiota in Atopic Dermatitis" International Journal of Molecular Sciences 23, no. 7: 3503. https://doi.org/10.3390/ijms23073503

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