Home Microbial perspective on the skin–gut axis and atopic dermatitis
Article Open Access

Microbial perspective on the skin–gut axis and atopic dermatitis

  • Bo Qu , Xue-er Zhang , Haoyue Feng , Bonan Yan , Yingchun Bai , Shanlin Liu and Yuhua He EMAIL logo
Published/Copyright: April 13, 2024

Abstract

Atopic dermatitis (AD) is a relapsing inflammatory skin condition that has become a global health issue with complex etiology and mounting prevalence. The association of AD with skin and gut microbiota has been revealed by virtue of the continuous development of sequencing technology and genomics analysis. Also, the gut–brain–skin axis and its mutual crosstalk mechanisms have been gradually verified. Accordingly, the microbiota–skin–gut axis also plays an important role in allergic skin inflammation. Herein, we reviewed the relationship between the microbiota–skin–gut axis and AD, explored the underlying signaling molecules and potential pathways, and focused on the potential mechanisms of probiotics, antimicrobial peptides (AMPs), coagulase-negative staphylococci transplantation, fecal microbiota transplantation, AMPs, and addition of essential fatty acids in alleviating AD, with the aim to provide a new perspective for targeting microbiota in the treatment of allergic skin inflammation.

1 Introduction

Atopic dermatitis (AD) is a chronic relapsing inflammatory skin disease mainly manifested as persistent itching of the skin clinically, which predominantly occurs in infants and adolescents but may also affect adults or the elderly. The incidence of AD shows an upward trend in recent decades, impacting approximately 15–20% of children and 1–3% of adults worldwide [1]. Although the pathophysiology of AD has not been completely understood, numerous studies have demonstrated that skin barrier disruption, serine protease disruption, keratinized desmosomes, massive secretion of inflammatory factors, and CD4+ T cell activation contribute to the onset of AD. Among them, T cell-mediated immune responses (Th1/Th2 imbalance and Thl7/Treg imbalance) may play a dominant role [2].

The application of emerging techniques, such as meta-transcriptomics, metagenomics, and metabolomics, has confirmed that local microorganisms can influence immune function at distal sites. For example, the gut microbiota can alter the skin phenotype through immunomodulatory effects, leading to the generation of terms such as the skin–gut axis and the microbiota–skin–gut axis. This article reviews the relationship between the microbiota–skin–gut axis and AD from the perspective of skin and gut microbiota, as well as their interacting signaling molecules and potential pathways, and attempts to elucidate the important roles of these pathways in the pathological mechanisms of AD. In addition, this review focuses on the potential mechanisms of microbe-related treatments such as probiotics, microbiota transplantation, and antimicrobial peptides (AMPs) in alleviating AD.

2 Microbial characteristics in AD

There is a complex ecosystem of microbiota that exist inside and outside the human body, known as human microbial communities, consisting of thousands of microbial species and trillions of microbial cells. Culture-based approaches have traditionally been used for the study of bacterial species, but many bacterial strains cannot be cultured in the laboratory, and bacterial culture-dependent methods under standard laboratory conditions can merely detect less than 1% of bacterial species [3]. Due to the harsh culture conditions for bacteria, the actual species of bacteria existing in the microbiome are often underestimated. The continuous development of molecular biology and bioinformatics has made significant contributions to microbiome research. At present, 16S rRNA gene sequencing, metabolism, and metagenomics are the most commonly used methods for microbial species research. 16S rRNA gene sequencing has allowed species-level and even strain-level resolution of the microbiome. Metagenomics provides a means of assessing the total genetic pool of the microbiome, in a culture-independent manner [4]. In recent years, researchers have regarded microbiota as a complex human trait and used quantitative and statistical genetic approaches to describe the genetic architecture of the microbiome in association with different host genomic loci. These modern molecular and bioinformatic techniques contribute to defining the genetic diversity of microorganisms and clarifying their relationship with commensal and pathogenic microbiomes.

2.1 Overview of skin microbiota in AD

The skin is a complex and dynamic ecosystem inhabited by millions of bacteria, fungi, and viruses. These microbes are collectively referred to as the skin microbiota, which is critical for protecting the skin from pathogens. The bacterial populations on the skin often adapt to the niche they occupy in the skin based on the unique physicochemical properties of the skin surface (temperature, age, sebum content, sweat, etc.) [5]. In accordance with existing literature, regions of the body that are abundant in sebum tend to be primarily colonized by lipokeratinocytes (Propionibacterium) and fungi (Malassezia). Conversely, areas that are moist, such as the axilla, are predominantly inhabited by Corynebacterium spp. and Staphylococcus spp. On the other hand, dry regions like the flexed side of the limbs exhibit varying populations consisting of β-Proteobacteria, Flavobacteria, and Malassezia [6]. The skin microbiota has the ability to influence the immunological response of the host by regulating several factors such as keratinocytes, AMPs, lipid antimicrobials, and cytokines. These regulatory mechanisms contribute to the enhancement of the skin barrier function, ultimately leading to the maintenance of homeostasis [7].

Disruption of the homeostasis of the internal environment in AD patients disrupts the microbial ecology of the skin, which is mainly characterized by significant changes in microbial diversity, number, composition, and metabolism [8]. According to the hygiene hypothesis, there is an inverse relationship between AD and early life exposure to microorganisms [9]. The earliest exposure of infants to microorganisms begins at delivery, and the mode of delivery exerts a significant impact on the newborn’s skin microbial community. For instance, the microbial community of infants delivered through the vagina is dominated by Lactobacilli, whereas that of infants delivered through cesarean section is dominated by Staphylococcus, Corynebacterium, and Propionibacterium [10]. Thus, multiple factors such as the mode of birth, stress, diet, and environmental pollution affect the skin microbiota in AD patients, which mediate the host immune response, induce IgE production and Th2 cell activation, and thus cause the exacerbation of AD symptoms [11]. A recent longitudinal study of the skin microbiota in AD patients has found altered microbial diversity on the AD skin surface (Table 1), as evidenced by a decrease in Streptococcus spp., Acinetobacter spp., Corynebacterium spp., and Prevotella spp., but an increase in Staphylococci such as Staphylococcus epidermidis and haemolyticus spp., among which the colonization density of Staphylococcus aureus is closely related to AD severity [12].

Table 1

Characterization of the skin and gut microbiota in AD

Position Microbial alterations Immune dysregulation
Skin Streptococcus spp. S. epidermidis ↓ AMPS
Acinetobacter spp. Staphylococcus hemolysis ↑ IL-4, ↑ IL-5,
Prevotella spp. Staphylococcus spp. ↑ IL-13, ↑ IL-22,
Corynebacterium S. aureus ↑ IL-31, ↑ IgE,
Propionibacterium acnes ↑ TSLP
Cutibacterium spp.
Acinetobacter
Gut Lactobacillus Bacillus coli ↓ Treg, ↓ IgA,
Bifidobacteria C. difficile ↑ IL-25, ↑ IL-33
S. epidermidis

Note: ↑: increase; ↓: decrease.

2.2 Overview of gut microbiota in AD

The human intestinal tract is home to a large and complex bacterial community, collectively referred to as the gut microbiota [13]. A total of 2,172 gut microbiota species in 12 phyla have been isolated from human individuals, of which 93.5% belong to Bacteroidetes, Firmicutes, Actinobacteria, and Proteobacteria [14]. Recent studies have demonstrated the critical involvement of gut microbiota in the development of allergic diseases such as intestinal inflammatory diseases, asthma, and AD [15]. The metabolites such as amino acid metabolites, short chain fatty acids (SCFAs), and oligosaccharides produced by gut microbiota can form a mucosal layer and constitute the intestinal barrier to avoid translocation of microbiota into the systemic circulation to initiate infection, thereby mediating material metabolism and immune function [16]. The immune balance between the gut microbiota and the host is vulnerable to diet, disease, and other factors. Some food ingredients can disrupt the intestinal barrier and then accelerate pathogenic microbial colonization [17]. Pathogenic microorganisms can enter the blood circulation and activate intestinal mucosal immunity, inducing the production of immune cells and inflammatory cytokines [18].

The imbalance of gut microbiota is also responsible for the pathogenesis of AD. By following high-risk infants for the first year of life and taking stool samples before the onset of atopic symptoms, several studies have found that infants whose mothers have a history of AD have high levels of detectable Escherichia coli in their stools, and a decreased ratio of Bifidobacterium to Clostridium difficile, which directly affects the development of the immune system later in life and increases the risk of developing AD. The gut microbial diversity of AD patients is reduced compared to that of healthy individuals (Table 1), mainly characterized by a significant decrease in the relative abundance of beneficial genera such as Lactobacillus and Bifidobacterium, but a significant increase in the relative abundance of E. coli, C. difficile, and S. aureus [19]. Reduced gut microbial diversity and decreased number of propionate and butyrate-producing bacteria are indicative of aggravated AD, while propionate and butyrate, as components of SCFAs, have anti-inflammatory and immunomodulatory effects and are essential for alleviating chronic inflammatory diseases including AD [20].

3 Skin–gut axis communication mediated by immune and neuroendocrine systems

There is a two-way relationship between gastrointestinal health and skin homeostasis, which is affected by diet, mood, and many external factors. Scientists have proposed that the skin–gut axis is a part of the gut–brain–skin axis, and the gut, brain, and skin cells are derived from the same germ layer [21]. Both skin and intestinal organs are the keys to immune and neuroendocrine functions, with high similarity in structure and nerve distribution [22]. The host immune and neuroendocrine systems can mediate skin–gut communication. The regulation of the immune system is mainly dominated by gut microbiota, and the immune imbalance is reflected in the skin. Intestinal dysfunction and gut microbiota imbalance may exert an intermediary effect on inflammatory skin diseases (Figure 1).

Figure 1 
               Neuroendocrine-mediated communication in the skin–gut axis. HPA axis: hypothalamic–pituitary–adrenal axis.
Figure 1

Neuroendocrine-mediated communication in the skin–gut axis. HPA axis: hypothalamic–pituitary–adrenal axis.

3.1 Immune-mediated communication in the skin–gut axis

3.1.1 Peyer’s patches

Peyer’s patches are parts of gut-associated lymphoid tissue dispersed throughout the intestine, which can synthesize interleukin-10 (IL-10) and induce T helper cell differentiation to participate in the intestinal mucosal immune system. Cytokines or immune cells from Peyer’s patches can be transported to the skin through circulation, thus regulating the immune state of the skin and improving the defense mechanism [23]. Therefore, it is speculated that Peyer’s patches provide a possible connection for gut–skin communication.

3.1.2 Epithelial cells (ECs)

ECs lining the inner surface of the intestinal tract and the outer surface of the skin play a pivotal role in protecting the human body from microbial infections. Thus, ECs establish a barrier between the external environment and the internal milieu. Alternations in gut microbiota may lead to an increase in intestinal permeability when the integrity of the intestinal epithelial barrier is destroyed to a certain level, causing the leaky gut syndrome. The formation of a leaky gut exacerbates the penetration of immunogenic molecules, including dietary antigens, bacterial toxins, and related pathogens such as tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) [24]. Activation of toll-like receptors (TLR) and zonulin signaling to initiate a series of inflammatory responses exacerbates the loss of barrier integrity of intestinal ECs, leading to bacterial translocation and inflammatory responses [25]. Subsequently, these immunogenic molecules can enter the bloodstream and accumulate in the skin, interfering with the epidermal barrier and eventually leading to skin immune response and skin inflammation [26].

3.1.3 Serum 25(OH)D

Serum 25(OH)D is an important immunomodulator that exists in the human body in the biologically inactive form of vitamin D. Vitamin D in macrophages regulates endogenous cathepsin and inhibin synthesis, cytokine release, and the body’s defense against pathogens. Serum 25(OH)D deficiency has been shown to trigger an inflammatory environment and gut microbiota imbalance. Elevated serum 25(OH)D levels induced by ultraviolet B (UVB) exposure are associated with increased alpha and beta diversity of gut microbiota [26]. Vitamin D can affect both innate and adaptive immune cells, thereby inhibiting pro-inflammatory responses [27]. It is also suggested that skin exposure to UVB may affect the activation of intestinal immune cells and the release of immune mediators, which in turn remodel gut microbiota, but the specific mechanisms remain to be elucidated. Vitamin D has antiproliferative, pro-differentiation, anti-apoptotic, and immunomodulatory effects in the skin. Epidemiologic studies have shown that populations living at higher geographic latitudes have lower sun exposure, lower vitamin D production, and increased prevalence of AD [28]. In addition, studies have observed that vitamin D level deficiency leads to an increased likelihood of developing AD and that the severity of AD is negatively correlated with vitamin D levels [29].

3.2 Neuroendocrine-mediated communication in the skin–gut axis

3.2.1 Neuropeptides

From the perspective of the neuroendocrine system, the activities of the central nervous system such as depression and anxiety, brain cognitive development, and stress response are related to gut microbiota. Mental stress can accelerate the excessive growth of intestinal microorganisms, increase intestinal permeability, destroy the intestinal immune barrier, and promote the release of neuropeptides from the nervous system [30]. Neuropeptides can stimulate keratinocytes to produce pro-inflammatory cytokines (such as IL-1α, IL-6, and IL-8), enhance the migration, antigen expression, and sensitization of Langerhans cells, induce mast cell degranulation and release of histamine and other mediators, and trigger peripheral blood mononuclear leukocytes to release IFN-γ, IL-4, TNF-α, and IL-10, thus eventually initiating inflammatory skin diseases (such as AD, psoriasis, and rosacea) [31,32].

3.2.2 Secretion of hormone-like compounds

Hormone-like compounds are a variety of metabolites secreted by gut microbiota (such as SCFAs and cortisol) and neurotransmitters (gamma-amino butyric acid [GABA], 5-hydroxytryptamine [5-HT], dopamine, and tryptophan). These hormone-like pleiotropic compounds are released into the blood and act on distant skin [33]. The skin–gut axis has a two-way effect, and changes in skin-related structural components also cause changes in gut microbiota as illustrated in Figure 1.

Both the skin and the gut are key to the immune and neuroendocrine systems, and the two may communicate through (1) the releasing effects of neural signaling molecules and (2) altered characterization of the skin and gut microbiota, and interactions between microbiota metabolites and associated immune cells.

4 Pathways of skin and gut microbiota affecting AD

4.1 Skin barrier pathways

The dysbacteriosis of microbiota in the AD skin surface can be attributed to the destruction of skin barrier function, the reduced contents of natural moisturizing factors, the increase in the surface pH value, and the changes in surface lipid compositions [34]. The latest research has shown that the colonization of S. aureus is related to the pathogenesis, disease flare, and disease phenotype of AD. S. aureus has a variety of highly evolved cell wall proteins and secretory factors, which can adhere to human skin and disturb the skin barrier through physical, chemical, and inflammatory mechanisms (Figure 2). The following aspects are involved.

Figure 2 
                  Crosstalk between the skin and the microbiome under conditional pathogenic bacteria. IL: interleukin; TNF-α: tumor necrosis factor-α; IFN-γ: interferon-γ; SCFAs: short chain fatty acids; SEA/B: staphylococcal enterotoxin A/B; TSST: toxic shock syndrome toxin.
Figure 2

Crosstalk between the skin and the microbiome under conditional pathogenic bacteria. IL: interleukin; TNF-α: tumor necrosis factor-α; IFN-γ: interferon-γ; SCFAs: short chain fatty acids; SEA/B: staphylococcal enterotoxin A/B; TSST: toxic shock syndrome toxin.

4.1.1 Epidermal barrier damage

S. aureus colonized on the surface of AD skin can express a variety of toxin factors and proteases, damaging the skin barrier and causing superficial and invasive skin infections. For example, S. aureus alpha-toxin (also known as pore-forming toxin) can penetrate the host cell membrane and dissolve the cell membrane [35]. Many proteases (serine protease, kallikrein KLK6, 13, and 14) can dissolve the stratum corneum, resulting in impaired skin barrier function [36]. Lipoteichoic acid (LTA), a product of the S. aureus cell wall, leads to skin barrier damage by inhibiting the expression of epidermal barrier proteins including filament polyprotein and cucumber protein [37].

4.1.2 Increase of bacterial adhesion

S. aureus can produce several surface molecules such as clumping factors A and B (ClfA and ClfB), fibronectin-binding protein (FnBP), and iron-regulated surface determinant A (IsdA) to adhere to the human stratum corneum [38]. In addition, the lack of skin lipids and the release of Th2 cytokines such as IL-4 in AD patients increase the expression of fibronectin and fibrinogen, conferring a favorable environment for S. aureus to adhere closely to keratinocytes [39].

4.1.3 Defect of bacterial clearance

The average pH value of the skin of AD patients is slightly alkaline, causing a decrease in the sphingosine level in the stratum corneum [39]. Sphingosine belongs to sphingolipids (SPL). SPL is an important component of cell membrane structure, and pharmacological SPL antagonism exacerbates vascular permeability and inflammation [40]. The instability of the stratum corneum cell membrane affects the transdermal water loss, pH value, serum IgE, and eosinophil of AD, which finally leads to the defect of skin bacterial clearance and accelerates the colonization of pathogenic bacteria.

Microorganisms such as fungi and bacteria cover the skin surface and reside in skin appendages, including hair follicles, sebaceous glands, and sweat glands. Skin resident microorganisms could promote the production of AMPS by keratinocytes and the production of complement by antigen-presenting cells and/or other innate immune mediators, which interact with the host to establish a functional immune response and prevent the overgrowth of pathogenic microorganisms. Skin microbial disorders and bacteria damage the skin barrier, activate downstream molecular signals, and induce chronic inflammatory responses.

(1) Stimulates AMPS to degranulation of mast cells, stimulates dendritic cells to bind to T cells, thereby inducing proliferation and polarization of T cells (Th1/Th2 and Th17/Treg imbalance). (2) Release of toxin factors, including staphylococcal enterotoxin A (SEA), staphylococcal enterotoxin B (SEB), SEC, and α-toxin-induced cell membrane lysis. (3) Hps70 activation through TLR channels leads to NF-κB overexpression, which in turn promotes the release of inflammatory cytokines, triggering cutaneous inflammatory responses.

4.2 Immune pathways

Despite the joint involvement of skin and gut microbiota in the human immune system, there are still differences between the two. Gut microbiota modulates gene regulation by regulating intestinal lymphoid structures, controlling stability, and activating the innate immune system [5]. Skin microbiota affects the immune system by producing AMPs, increasing the activity of the complement system, and affecting the expression of functional variants of TLRs and nucleotide-binding oligomerization domain/contain caspase-recruitment domain receptors [41].

4.2.1 Skin microbial immune pathways

Microbiota can trigger pro-inflammatory mechanisms by releasing toxin factors and inflammatory drivers, thereby affecting the expression of TLRs and the function of host immune cells. It mainly involves the following aspects: (1) staphylococcal toxin factor-elicited immune responses: staphylococcal toxin factors are mainly released by the mediation of staphylococcal enterotoxins (SEs), SEA, SEB, and toxic shock syndrome toxin (TSST)-1. These toxins function as superantigens (SAgs) [42] to activate T cell clones, resulting in substances capable of mounting an extremely strong immune response, which subsequently triggers IgE-mediated mast cell degranulation and elevates Th2 cytokines [43]; moreover, they bind to human leukocyte antigen-DR on Langerhans cells and macrophages to stimulate the production of IL-1, TNF-α, and IL-12, thus contributing to the exacerbation and recurrence of AD [43]. (2) Secretion of thymic stromal lymphopoietin (TSLP): S. aureus is also able to trigger keratinocyte apoptosis, releasing TSLP. TSLP release can mediate the itch response by inducing skin dendritic cell activation, promoting IL-4 and IL-13 secretion, and influencing Th2 cell recruitment [36]. (3) Malassezia-stimulated inflammatory factors: Malassezia colonized on AD skin mediates dendritic cell maturation and stimulates keratinocytes to produce a variety of inflammatory cytokines (e.g., IL-4, IL-6, IL-8, and TNF-α); Malassezia also induces IgE-mediated mast cell degranulation to release leukotrienes, thus perpetuating inflammation [44]. However, Malassezia does not produce IL-12. IL-12 is a determinant of Th1-type immune responses that can promote the production of Th1 class factors and inhibit the production of Th2 class factors, thereby inhibiting Th2-type immune responses. Hence, Malassezia favors the induction of Th2-type immune responses in AD pathogenesis [45].

4.2.2 Gut microbial immune pathways

In addition to intestinal lymphoid structure, gut microbiota may also regulate intestinal immune homeostasis and cause peripheral inflammation through the intestinal vasculature. Several lines of evidence have suggested that gut microbiota activates TLRs 2, 6, and 4 and causes inflammatory responses by activating the canonical NF-κB pathway while upregulating vascular endothelial growth factor receptor 2 to promote intestinal angiogenesis [46]. Interestingly, intestinal vascular endothelial growth factor C (VEGF-C) can recognize microorganisms through TLR complexes. Therefore, VEGF-C may be a key downstream effector of gut microbiota, regulating the recruitment of macrophages and Tregs and the balance of IL-9/IL-17 [47]. An assessment of gut microbiota and innate immune responses in IgE-associated eczema has revealed that the number of Ruminococci in the stools of children with AD is lower than that of healthy controls, and Ruminococci has a negative correlation with TLR2-induced IL-6 and TNF-α, while Enterobacteriaceae is positively correlated with TLR4-induced TNF-α [48]. Bifidobacteria can directly or indirectly enhance the immunosuppressive function of Tregs by stimulating the IL-10/IL10Ra signaling loop, indicating that the number of Bifidobacteria in the gut affects the occurrence of AD [49]. Lactobacillus plantarum LM1004 reduces Th2 and Th17 cell transcription factors, serum IgE, and TSLP, but increases the transcription factors of Tregs, Th1 cells, and filaggrin, while butyrate, which modulates gut microbiota, significantly improves AD symptoms [50]. The decreased IL-17A expression has been found in plasma samples of AD children in association with a decrease in Romboutsia and Dorea and an increase in anaerobe and Faecalibacterium prausnitzii in the gut [51].

4.3 Metabolite pathways

Microbiota can secrete a variety of metabolites such as SCFAs, cortisol, GABA, 5-HT, dopamine, and tryptophan. These metabolites can accumulate under the skin or be released into the blood, affecting keratinocyte differentiation, skin hydration ability, and epidermal barrier function by downregulating the expression of keratin and acting on the skin immune system. The microbial metabolites in the skin–gut axis are closely related to the pathogenesis of AD, and different metabolites can be independent factors of the clinical phenotype and recurrence of AD.

4.3.1 Polyunsaturated free fatty acids (PUFAs)

PUFAs are divided into ω-6PUFAs and ω-3PUFAs. Arachidonic acid (ARA) in ω-6PUFAs is generally considered to induce and promote inflammation. Malassezia colonized on AD skin can produce a variety of enzymes (including lipase and phospholipase), which release PUFAs from sebum lipids to trigger inflammation [52]. This process may be closely related to the release of ARA. ARA can produce pro-inflammatory eicosanoid acids (prostaglandins) and leukotrienes under the action of phospholipase A2 (PLA2), resulting in inflammation and stratum corneum damage. It has been found that mice overexpressing PLA2III exhibit histologically similar skin inflammation to AD, such as hyperkeratosis, acanthosis, incomplete keratosis, erosion, ulcers, and sebaceous hyperplasia [53]. Interestingly, octadecenoic acid and ARA isomers can be detected in the T lymphocyte membrane of AD patients [54]. Therefore, Malassezia colonized on AD skin may affect the formation of trans-fatty acid isomers in the T cell membrane, the release of pro-inflammatory mediators, and the activity of T cell signaling transduction, eventually leading to the pathogenesis of AD.

4.3.2 SCFAs

SCFAs, including butyrate, acetate, and propionate, are produced during bacterial fermentation of undigested polysaccharides, which can reduce the permeability of the intestinal barrier, enter the blood, and act on the immune system. SCFAs mainly act on the immune system through two mechanisms to elicit systemic effects, namely, activating the immune signaling through G protein-coupled receptors (GPR41/GPR43/GPR109A) and inhibiting histone deacetylases [55]. For example, propionate can promote the generation of macrophage and dendritic cell precursors, hinder the differentiation of Naïve T cells into Th2 cells, and thereby prevent the development of allergic inflammation [56]. SCFAs may inhibit skin inflammation by increasing the number of skin resident Tregs [57]. Topical administration of SCFAs has been demonstrated to restore immune function and control inflammatory skin diseases potentially through the modulation of T cells, which represents a novel strategy for protocols using bacterial metabolites to control skin inflammation.

4.3.3 Tryptophan

The ligands of tryptophan metabolites can signal through the aryl hydrocarbon receptor (AhR) to inhibit the production of inflammatory cytokines in the skin and gut, thereby reducing the occurrence of inflammatory reactions [58]. Indoles and their derivatives, such as 3-indoleacrylicacid (IA), 3-indoleacetic acid (AA), indole-3-carboxaldehyde (I3C), and indole acetaldehyde (IAId), belong to tryptophan metabolites. (1) IAId, a product of tryptophan metabolism by skin microbes, can inhibit the production of TSLP in keratinocytes in an AhR-dependent manner, thus reducing skin inflammation in mice with AD-like dermatitis. (2) I3C, a tryptophan metabolite of Bifidobacterium longum CCFM1029, can activate the AhR-mediated immune signaling pathway to improve AD symptoms [59]. (3) Kynurenic acid (KYNA), a product of tryptophan metabolism of Bifidobacterium LKM512, has mucosal protection and immunomodulatory effects, which can relieve AD pruritus and improve the quality of life of AD patients [60].

4.3.4 Polyamines

Polyamines are metabolites produced by a variety of bacteria that exist widely in nearly all living organisms. The function of the intestinal mucosal barrier in healthy adults to prevent antigen invasion is mainly maintained by the concentration of polyamines secreted by gut microbiota. Polyamines alleviate systemic inflammation by inhibiting the synthesis of inflammatory cytokines. Intake of the probiotic strain LKM512 facilitates the production of polyamines and butyrates in the gut of AD patients, induces Th1 cytokine production, and promotes intestinal mucosal barrier function recovery, thus alleviating AD symptoms [61]. In contrast, reduced concentrations of intestinal polyamines have been detected in adults with refractory AD [62]. Therefore, the decrease of polyamines may be associated with the recurrence of AD, but more studies are still needed to verify the relationship between polyamines and AD.

The microbial metabolites in the skin–gut axis are closely associated with the pathogenesis of AD, and different metabolites can be independent factors for the phenotype and recurrence of AD. The current AhR-dependent anti-inflammatory active agent (Tapinarof) has been shown to reduce inflammatory cytokine production and improve skin inflammatory symptoms in animal models, and its efficacy and safety in treating mild to severe AD have been confirmed in randomized controlled trials [63]. It is suggested that topical administration of microbial metabolites may alleviate inflammatory skin diseases, but the underlying mechanisms remain to be further confirmed.

4.4 Neuroendocrine pathways

Emerging evidence has revealed that gut microbiota mediates the effects of emotional and neurological states on the skin, indicating that microbiota is an important factor in the communication network of the skin–gut axis [64]. Microbiota can directly or indirectly regulate the skin–gut axis via neuroimmune and neuroendocrine pathways. This effect is mainly dependent on the interaction between microbiota and the central nervous system and the conduction of microbial signaling molecules. Among them, signaling molecules mainly include metabolites produced by microorganisms, neurotransmitters, and cytokines released during microbial immune responses, etc.

4.4.1 Interactions between the microbiota and the central nervous system

The central nervous system regulates the gastrointestinal system and enteric nervous system via the autonomic nervous system (ANS) (sympathetic and parasympathetic/vagal) and the hypothalamic–pituitary–adrenal (HPA) axis. The central nervous system can directly alter the environment of gut microbiota or indirectly affect gut microbiota through a large number of signaling molecules [65]. ANS-mediated mucus secretion may have a pivotal impact on the intestinal mucus layer, an important habitat for gut microbiota. The ANS can also respond to bacteria containing AMPs by modulating intestinal immune cells such as macrophages and mast cells [66]. Gut microbiota can also produce neuroactive molecules and neurotransmitters, which in turn influence the function of the vagus nerve. Vagotomy can abolish anxiety-like behavior caused by B. longum [67]. Pressure alopecia and neurodermatitis are significantly alleviated in mice fed with Lactobacilli.

4.4.2 Conduction of microbial signaling molecules

The nervous system is involved in regulating the immune response in the skin of AD patients, forming a neuro-immune-endocrine regulatory network, which is bi-directionally regulated by various biochemical mediators [68]. Studies have shown that AD patients can stimulate skin immune responses by releasing neuropeptides and vasoactive intestinal peptides that regulate the function of keratinocytes and other immune cells [68]. For example, vasoactive intestinal peptides are associated with itch intensity in AD, affect IFN-γ and IL-4 cytokine release, and increase the risk of IgE-mediated mycobacterial sensitization in AD, all of which suggest that neuropeptides may be used as better alternative biomarkers for AD [69]. The conduction elements of molecules are as follows.

4.4.2.1 Metabolites

SCFAs, cortisol, and tryptophan are common metabolites produced by gut microbiota, which can signal to the host through neuroendocrine pathways and receptors on local cells in the gut, implying the potential role of metabolites in the communication between the microbiota and the central nervous system. HPA axis dysfunction is commonly seen in AD patients. The acute reduction in the excitability of the HPA axis increases cortisol levels, resulting in the imbalance of Th1/Th2 and the secretion of cytokines that influence the inflammatory immune phenotype of AD patients. Tryptophan, as a serotonin synthetic precursor in the brain, can affect central serotonin concentrations and downstream neuroactive metabolites, thus directly mediating AD-independent itch responses. SCFAs (butyrate, acetate, and propionate) can stimulate the sympathetic nervous system to interact with neuronal cells and upregulate the expression of neurokines, thereby affecting the skin lesion phenotype of AD [70].

4.4.2.2 Neurotransmitters

Gut microbiota can produce neurotransmitters such as 5-HT and GABA to influence gut–brain communication [71]. Neurotransmitters may be key regulators of the skin–gut axis. Among them, 5-HT is involved in the communication between the immune system and the central nervous system. Peripheral 5-HT has been identified as a potent itch inducer that directly mediates AD-independent itch, which may be related to the expression of specific 5-HT receptors (HTR7). The coupling of HTR7 to the stimulatory receptor TRPA1 triggers neuronal excitation and mediates itch responses [72]. GABA, an inhibitory neurotransmitter mainly produced by lactic acid bacteria and Bifidobacterium, can inhibit the production of iNOS, IL-1, and TNF-α to exert anti-inflammatory activity [73]. Recent studies have shown that increased GABA ligands and GABAA receptors may mediate itch responses in inflammatory skin diseases [74]. All these findings suggest that neurotransmitters serve as novel immunomodulators for the treatment of AD.

4.4.2.3 Cytokines released during microbial immune responses

Cytokines are released during microbial immune responses and mediate host responses through neuroimmune pathways [75]. For example, S. aureus mediates mast cell degranulation by releasing SAgs, causes an increase in Th2 immunoregulatory factors, and releases TSLP by promoting keratinocyte apoptosis. These immune cytokines may mediate AD itch responses by generating itch signaling via the vagal or spinal neural pathways. Among them, mast cells communicate with the HPA axis through mediators such as tryptophanase, histamine, and neuropeptides. Concomitantly, Mas-related G-protein-coupled receptor is activated to regulate transient receptor channel potential cation channel ankyrin subtype 1 (TRPA1) expression and mediate histamine-independent itch responses [76]. TSLP can be induced to activate neurons and directly stimulate itch sensory nerve fibers, thus producing itch. IL-31 can directly act on a subpopulation of IL-31RA/transient receptor potential cation channel vanilloid subtype 1 (TRPV1)/TRPA1 + dorsal root ganglia neurons [77]. Moreover, IL-31 binds to IL-31RA, activates the JAK-STAT pathway, and thereby mediates itch responses [78]. In a phase II trial, nemolizumab, an anti-IL-31RA monoclonal antibody, significantly alleviates pruritus in patients with moderate to severe AD [79]. Therefore, interventions targeting immune cytokines released by specific microbes to block the communication with neural pathways may provide novel targets for the treatment of skin diseases.

In summary, the “microbial–skin–gut” axis affects and exacerbates AD through specific pathways such as the skin barrier pathway, immune pathway, metabolic pathway, and neuroendocrine pathway (Figure 3). The main manifestations are microorganisms adhere to human skin and interfere with the skin barrier function through physical, chemical, and inflammatory mechanisms; release toxins such as SAg and secrete metabolites such as PUAF and dopamine, which cause T-cell proliferation and differentiation, and mediate the Th1/Th2/Th17 immune response, thus participating in AD pathogenesis and influencing AD disease phenotype; and release neurotransmitters such as GABA, 5-HT, or neuroactive metabolites such as SCFAs, cortisol, and tryptophan, which generate pruritic signals and mediate the AD itching response.

Figure 3 
                        Communication on the “microbe–skin–gut” axis. IL: interleukin; 5-HT: 5-hydroxytryptamine; TNF-α: tumor necrosis factor-α; IFN-γ: interferon-γ; SCFAs: short chain fatty acids; SEA/B: staphylococcal enterotoxin A/B; TSST: toxic shock syndrome toxin; GABA: gamma-amino butyric acid; BAS: basophils; Trp: tryptophan; SCFAs: short chain fatty acids.
Figure 3

Communication on the “microbe–skin–gut” axis. IL: interleukin; 5-HT: 5-hydroxytryptamine; TNF-α: tumor necrosis factor-α; IFN-γ: interferon-γ; SCFAs: short chain fatty acids; SEA/B: staphylococcal enterotoxin A/B; TSST: toxic shock syndrome toxin; GABA: gamma-amino butyric acid; BAS: basophils; Trp: tryptophan; SCFAs: short chain fatty acids.

The “microbial–skin–gut” axis affects and exacerbates AD mainly through specific pathways such as skin. The “microbial–skin–gut” axis affects and exacerbates AD through specific pathways such as the skin barrier pathway, immune pathway, metabolic pathway, and neuroendocrine pathway.

5 AD therapy based on skin and intestinal microorganisms

Currently, the prevalent treatments for AD mainly include topical application of moisturizers, corticosteroids, and anti-inflammatory agents (such as calcineurin inhibitors and phosphodiesterase inhibitors) to control the disease symptom’s severity and duration. Phototherapy and systemic immunosuppressants (Omazumab IGE inhibitors, Fezakinumab unzanumab IL-22 inhibitors, Suchizumab IL-17A inhibitors, and Nemolizumab IL-31 inhibitors) are used for refractory and severe AD by improving skin hydration and repairing skin barrier function, and the above agents target different pathways in the immune system (JAK-STAT, phosphodiesterase 4, AhR receptor, and Th2 cytokines). Exploring the pathogenesis of AD from the microbiota–skin–gut axis and determining the possible communication pathways of microbiota in the axis and the possible mechanism of their effects on AD will provide a new targeted approach for the treatment of AD.

5.1 Ingestion of probiotics

To repair the skin barrier by ingesting probiotics and prebiotics has been gradually recognized. Probiotics possess a variety of therapeutic effects: (1) modulating gut microbiota, stimulating mucus secretion, maintaining epithelial tight junction function, and thereby repairing intestinal mucosal barrier integrity; (2) secreting a variety of metabolites and neurotransmitters (SCFAs, phenolics, 5-HT, tryptophan, etc.) that alter intestinal mucosal permeability, followed by reducing the entry of noxious substances into the circulatory system to affect skin barrier function; and (3) inducing the production of the anti-inflammatory cytokine IL-10, stimulating hypothalamic hormone secretion, and thereby modulating skin immune status [80]. In a randomized controlled clinical study, oral Lactobacillus parasarum strains can reduce skin sensitivity and increase barrier function. Intake of probiotic strain LKM512 alleviates the symptoms of AD by inhibiting the production of Th1 cytokines and increasing the concentration of polyamines, which may be due to the fact that KYNA in polyamine metabolism can be transported to the brain through the blood to relieve itching [52].

Recent studies have found a strong association between gluten diet and skin diseases. Gluten, a protein found in wheat, rye, and barley, causes immune reactions in patients with celiac disease (CD) and induces cutaneous manifestations such as dermatitis herpetiformis [81]. The incidence of AD in patients complicated with CD is three times higher than that in patients complicated with other allergic diseases [82]. Gluten sensitivity is related to gut microbiota imbalance. Some probiotics (Bifidobacterium breve, B. longum, Bifidobacterium infantas, L. plantarum, Lactobacillus acidophilus, Lactobacillus casei, etc.) can hydrolyze gluten polypeptides, decrease TNF-α, and increase IL-10, thereby exerting an anti-inflammatory effect in the intestinal mucosa [83]. Therefore, probiotics may be a promising treatment approach for gluten sensitivity-related AD, especially in AD patients with CD symptoms, but the specific mechanism remains to be further discovered.

5.2 Addition of AMPs

AMPs are short peptides with a broad spectrum of antibiotic activities, which modulate the innate immune system of the host to promote pathogen clearance. The production of AMPs such as beta-defensin-2, beta-defensin-3, and LL-37 in AD patients is reduced, which may be attributed to S. aureus colonization [46]. The relative reduction of AMPs then exacerbates the colonization of pathogenic microorganisms, leading to a higher risk of developing severe skin, multi-organ infection, and systemic infection in AD patients. Omiganan (OMN) is an AMP with extensive antibacterial and anti-biofilm activities. The use of OMN can effectively restore the ecological balance of the skin in adult patients with moderate to severe AD, reduce S. aureus colonization, and increase microbial diversity indexes [84]. OMN strongly drives the interferon regulatory factor signaling induced by TLR and the NF-κB inflammatory pathway, increases the levels of II-IFNɣ secreted by T cells, and thereby achieves anti-inflammatory and anti-viral effects [85]. Thus, AMPs may reduce skin inflammation and repair epidermal barrier function by modulating the innate immune system. This also further confirms that the role of AMPs in both pro-inflammatory and anti-inflammatory pathways relies on their immunomodulatory properties rather than antimicrobial properties. The pathways to reduce the colonization of pathogenic microorganisms not only depend on the use of traditional antibiotics but may also rely on antibacterial effects by reducing the production of pro-inflammatory factors.

5.3 Microflora transplantation

5.3.1 Coagulase-negative staphylococci (CoNS) transplantation

CoNS are skin commensal bacteria found in Staphylococcus species, including S. epidermidis, Staphylococcus hominis, S. haemolyticus, Staphylococcus capitis, and other common isolates. CoNS residing in the skin can compete against invading pathogens by producing AMPs or increasing the activity of the complement system (IL-1 levels) to limit the colonization of pathogens, achieving an anti-colonization effect. The antimicrobial capacity of CoNS is strongly associated with S. aureus colonization, mainly with S. epidermidis and S. hominis [86]. For example, Staphylococcus lugdunensis, a species of CoNS, has been shown to produce a polypeptide antibiotic to inhibit S. aureus in a rat nasal colonization model [87]. The phenol-soluble modulin peptide expressed by S. epidermidis is also able to kill S. aureus [88]. Staphylococcus muciniphila transplanted into healthy humans can inhibit S. aureus via lysophosphatidylcholine and activate the innate immune system to effectively alleviate AD [89]. CoNS transplantation may produce antibiotic-like compounds that antagonize competing pathogens to effectively alleviate AD. CoNS transplantation has a stronger ability to restore the homeostasis of skin microbiota than antibiotic therapy because the nonspecific antimicrobial effects of drug-derived antibiotics are likely to kill protective strains such as cons and instead increase the possibility of S. aureus colonization.

5.3.2 Fecal microbiota transplantation (FMT)

FMT, where fecal microbiota from a healthy donor is transplanted into a patient’s gastrointestinal tract, is currently being explored as a potential therapy to reconstruct gut microbiota balance. Of note, FMT has a more long-term effect on gut microbiota than the ingestion of probiotics, probably because FMT can provide patients with core bacterial species of a healthy population and replace the bacterial components lost in patients, while orally administered probiotics fail to colonize the gastrointestinal tract [90]. For the underlying mechanisms of FMT in the treatment of AD, FMT is reported to modulate the abundance of gut microbiota, the levels of SCFAs, and the immune balance of Th1/Th2 cells in AD mice, mainly manifested as downregulation of Th2 cytokines (IL-4, IL-5, and IL-13) and Treg cytokines (IL-10 and IL-1β), upregulation of Th1 cytokines (IL-12, IFN-γ, and TNF-α), as well as elevation of butyrate, propionate, and acetate [91]. A latest clinical trial evaluating the efficacy of FMT in adult patients with AD has shown that the SCORAD score of AD patients is decreased significantly after FMT [92]. All these suggest that FMT may be a safe and effective treatment intervention for AD patients. Of course, further research is warranted to unveil its specific mechanism.

5.4 Addition of essential fatty acids (EFAs)

EFAs include PUFAs (ω-3 and ω-6), linoleic acid (LA), and alpha-linolenic acid (ALA). LA is a member of the ω-6 family, while ALA is classified into the ω-3 family. Studies have shown that LA levels in AD patients typically increase, while their metabolite linolenic acid (DGLA) decreases [93]. As mentioned earlier, the colonization of Malassezia on the skin can trigger inflammatory reactions by producing various enzymes (including lipase and phospholipase) and releasing PUFA. Unsaturated fatty acids have antibacterial effects, thus preventing excessive growth of pathogenic bacteria in the gastrointestinal tract [94]. This suggests that fatty acids may regulate microbial metabolism and mediate the occurrence of AD. Currently, the main focus is on supplementing ω-3PUFAs and ω-6PUFAs safe and biologically active fatty acids to improve clinical symptoms of moderate to severe AD [95]. ω-3PUFAs have powerful anti-inflammatory functions, they can inhibit lymphocyte proliferation, cytokine production, adhesion molecule expression, and trigger cell apoptosis, which can be used to treat the most common chronic inflammatory skin diseases, such as AD, psoriasis, and acne [96]. ω-3PUFAs may also affect the bacterial diversity of the gut microbiota, creating a less inflammatory environment for the colon mucosa during human health and disease treatment [97].

In a word, PUFAs and their derivatives play an important role in the prevention or treatment of AD. The possible mechanisms of PUFAs in treating AD include promoting the differentiation and maturation of the stratum corneum, repairing skin barrier function, reducing the production of cytokines, inhibiting mast cell degranulation, and regulating immunity.

6 Summary

This review focuses on the possible mechanisms of the microbiota–skin–gut axis in the pathogenesis of AD and explores new treatment approaches for AD. The therapeutic advantages and potential mechanisms of probiotics, AMPs, microbiota transplantation, and fatty acids are discussed. Among them, probiotics and FMT can not only modulate gut microbiota, but also secrete a variety of metabolites and neurotransmitters to induce the production of anti-inflammatory factors, thereby modulating the immune status of the skin. However, whether supplementation with intestinal probiotics affects AD skin microbiota still warrants further investigation. The addition of AMPs and cons transplantation can reduce S. aureus colonization through antagonistic competition against microbiota and affect the innate immune system to restore the immune balance in AD skin.

The microbiota has the ability to function as a signaling molecule, facilitating communication between the skin and gut and thereby impacting the overall balance of the skin–gut axis. This influence extends to several components of the skin–gut axis, including the epidermal barrier, immune system, metabolism, and neuroendocrine system. Based on the role of serum 25(OH)D, UVB, and EFAs in AD, it can be inferred that alterations in skin microbiota may affect the intestinal immune system and thus remodel gut microbiota, which confers novel insights into the possible communication of the skin–gut axis. So far, the underlying mechanisms of the microbiota–skin–gut axis in the treatment of AD have not been thoroughly elucidated. Therefore, we still need to integrate multi-omics, such as meta-transcriptomics, metagenomics, and metabolomics, to provide a new perspective for targeting microbiota in the treatment of allergic skin inflammation.


# These are co-first authors of the article.


Acknowledgements

The authors thank Professor Yu-hua He for her helpful comments and suggestions on the review.

  1. Funding information: Authors state no funding involved.

  2. Author contributions: Bo Qu and Xue-er Zhang performed the selection of literature, drafted the manuscript, and prepared the figures. Bo Qu and Xue-er Zhang contributed equally. Hao-yue Feng, Bo-nan Yan, and Ying-chun Bai helped to collect literature, participated in the discussion, and revised the manuscript. Shan-lin Liu and Yu-hua He carried out the design of this review and revised the manuscript. All approved the final version of the manuscript.

  3. Conflict of interest: Authors state no conflict interest.

  4. Data availability statement: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1] Nutten S. Atopic dermatitis: global epidemiology and risk factors. Ann Nutr Metab. 2015;66(Suppl 1):8–16.10.1159/000370220Search in Google Scholar PubMed

[2] Auriemma M, Vianale G, Amerio P, Reale M. Cytokines and T cells in atopic dermatitis. Eur Cytokine Netw. 2013;24(1):37–44.10.1684/ecn.2013.0333Search in Google Scholar PubMed

[3] Chen YE, Tsao H. The skin microbiome: current perspectives and future challenges. J Am Acad Dermatol. 2013;69(1):143–55.10.1016/j.jaad.2013.01.016Search in Google Scholar PubMed PubMed Central

[4] Godlewska U, Brzoza P, Kwiecień K, Kwitniewski M, Cichy J. Metagenomic studies in inflammatory skin diseases. Curr Microbiol. 2020;77(11):3201–12.10.1007/s00284-020-02163-4Search in Google Scholar PubMed PubMed Central

[5] Belkaid Y, Segre JA. Dialogue between skin microbiota and immunity. Science (New York, NY). 2014;346(6212):954–9.10.1126/science.1260144Search in Google Scholar PubMed

[6] Findley K, Oh J, Yang J, Conlan S, Deming C, Meyer JA, et al. Topographic diversity of fungal and bacterial communities in human skin. Nature. 2013;498(7454):367–70.10.1038/nature12171Search in Google Scholar PubMed PubMed Central

[7] Grice EA, Segre JA. The skin microbiome. Nat Rev Microbiol. 2011;9(4):244–53.10.1038/nrmicro2537Search in Google Scholar PubMed PubMed Central

[8] Fitz-Gibbon S, Tomida S, Chiu BH, Nguyen L, Du C, Liu M, et al. Propionibacterium acnes strain populations in the human skin microbiome associated with acne. J Investig Dermatol. 2013;133(9):2152–60.10.1038/jid.2013.21Search in Google Scholar PubMed PubMed Central

[9] Geoghegan JA, Irvine AD, Foster TJ. Staphylococcus aureus and atopic dermatitis: a complex and evolving relationship. Trends Microbiol. 2018;26(6):484–97.10.1016/j.tim.2017.11.008Search in Google Scholar PubMed

[10] Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A. 2010;107(26):11971–5.10.1073/pnas.1002601107Search in Google Scholar PubMed PubMed Central

[11] Ahmad-Nejad P, Mrabet-Dahbi S, Breuer K, Klotz M, Werfel T, Herz U, et al. The toll-like receptor 2 R753Q polymorphism defines a subgroup of patients with atopic dermatitis having severe phenotype. J Allergy Clin Immunol. 2004;113(3):565–7.10.1016/j.jaci.2003.12.583Search in Google Scholar PubMed

[12] Hugon P, Dufour JC, Colson P, Fournier PE, Sallah K, Raoult D. A comprehensive repertoire of prokaryotic species identified in human beings. Lancet Infect Dis. 2015;15(10):1211–9.10.1016/S1473-3099(15)00293-5Search in Google Scholar PubMed

[13] Dinan TG, Cryan JF. The microbiome–gut–brain axis in health and disease. Gastroenterol Clin North Am. 2017;46(1):77–89.10.1016/j.gtc.2016.09.007Search in Google Scholar PubMed

[14] Gonzalez ME, Schaffer JV, Orlow SJ, Gao Z, Li H, Alekseyenko AV, et al. Cutaneous microbiome effects of fluticasone propionate cream and adjunctive bleach baths in childhood atopic dermatitis. J Am Acad Dermatol. 2016;75(3):481–93.e8.10.1016/j.jaad.2016.04.066Search in Google Scholar PubMed PubMed Central

[15] Ahluwalia B, Magnusson MK, Öhman L. Mucosal immune system of the gastrointestinal tract: maintaining balance between the good and the bad. Scand J Gastroenterol. 2017;52(11):1185–93.10.1080/00365521.2017.1349173Search in Google Scholar PubMed

[16] Iwasaki A, Kelsall BL. Localization of distinct Peyer’s patch dendritic cell subsets and their recruitment by chemokines macrophage inflammatory protein (MIP)-3alpha, MIP-3beta, and secondary lymphoid organ chemokine. J Exp Med. 2000;191(8):1381–94.10.1084/jem.191.8.1381Search in Google Scholar PubMed PubMed Central

[17] Usuda H, Okamoto T, Wada K. Leaky gut: effect of dietary fiber and fats on microbiome and intestinal barrier. Int J Mol Sci. 2021;22(14):7613.10.3390/ijms22147613Search in Google Scholar PubMed PubMed Central

[18] Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535(7610):65–74.10.1038/nature18847Search in Google Scholar PubMed

[19] Song H, Yoo Y, Hwang J, Na YC, Kim HS. Faecalibacterium prausnitzii subspecies-level dysbiosis in the human gut microbiome underlying atopic dermatitis. J Allergy Clin Immunol. 2016;137(3):852–60.10.1016/j.jaci.2015.08.021Search in Google Scholar PubMed

[20] Miyazaki K, Masuoka N, Kano M, Iizuka R. Bifidobacterium fermented milk and galacto-oligosaccharides lead to improved skin health by decreasing phenols production by gut microbiota. Benefic Microbes. 2014;5(2):121–8.10.3920/BM2012.0066Search in Google Scholar PubMed

[21] Bowe WP, Logan AC. Acne vulgaris, probiotics and the gut–brain–skin axis – back to the future. Gut Pathog. 2011;3(1):1.10.1186/1757-4749-3-1Search in Google Scholar PubMed PubMed Central

[22] O’Neill CA, Monteleone G, McLaughlin JT, Paus R. The gut–skin axis in health and disease: a paradigm with therapeutic implications. BioEssays: N Rev Mol Cell Dev Biol. 2016;38(11):1167–76.10.1002/bies.201600008Search in Google Scholar PubMed

[23] Benyacoub J, Bosco N, Blanchard C, Demont A, Philippe D, Castiel-Higounenc I, et al. Immune modulation property of Lactobacillus paracasei NCC2461 (ST11) strain and impact on skin defences. Benefic Microbes. 2014;5(2):129–36.10.3920/BM2013.0014Search in Google Scholar PubMed

[24] Kinashi Y, Hase K. Partners in leaky gut syndrome: intestinal dysbiosis and autoimmunity. Front Immunol. 2021;12:673708.10.3389/fimmu.2021.673708Search in Google Scholar PubMed PubMed Central

[25] Andrews C, McLean MH, Durum SK. Cytokine tuning of intestinal epithelial function. Front Immunol. 2018;9:1270.10.3389/fimmu.2018.01270Search in Google Scholar PubMed PubMed Central

[26] Bosman ES, Albert AY, Lui H, Dutz JP, Vallance BA. Skin exposure to narrow band ultraviolet (UVB) light modulates the human intestinal microbiome. Front Microbiol. 2019;10:2410.10.3389/fmicb.2019.02410Search in Google Scholar PubMed PubMed Central

[27] Clark A, Mach N. Role of vitamin D in the hygiene hypothesis: the interplay between vitamin D, vitamin D receptors, gut microbiota, and immune response. Front Immunol. 2016;7:627.10.3389/fimmu.2016.00627Search in Google Scholar PubMed PubMed Central

[28] Weiland SK, Hüsing A, Strachan DP, Rzehak P, Pearce N. Climate and the prevalence of symptoms of asthma, allergic rhinitis, and atopic eczema in children. Occup Environ Med. 2004;61(7):609–15.10.1136/oem.2002.006809Search in Google Scholar PubMed PubMed Central

[29] Kang JW, Kim JH, Kim HJ, Lee JG, Yoon JH, Kim CH. Association of serum 25-hydroxyvitamin D with serum IgE levels in Korean adults. Auris, Nasus, larynx. 2016;43(1):84–8.10.1016/j.anl.2015.06.010Search in Google Scholar PubMed

[30] Luthold RV, Fernandes GR, Franco-de-Moraes AC, Folchetti LG, Ferreira SR. Gut microbiota interactions with the immunomodulatory role of vitamin D in normal individuals. Metabolism: Clin Exp. 2017;69:76–86.10.1016/j.metabol.2017.01.007Search in Google Scholar PubMed

[31] Joachim RA, Handjiski B, Blois SM, Hagen E, Paus R, Arck PC. Stress-induced neurogenic inflammation in murine skin skews dendritic cells towards maturation and migration: key role of intercellular adhesion molecule-1/leukocyte function-associated antigen interactions. Am J Pathol. 2008;173(5):1379–88.10.2353/ajpath.2008.080105Search in Google Scholar PubMed PubMed Central

[32] Rosa AC, Fantozzi R. The role of histamine in neurogenic inflammation. Br J Pharmacol. 2013;170(1):38–45.10.1111/bph.12266Search in Google Scholar PubMed PubMed Central

[33] Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF, Dinan TG. Minireview: Gut microbiota: the neglected endocrine organ. Mol Endocrinol (Baltimore, MD). 2014;28(8):1221–38.10.1210/me.2014-1108Search in Google Scholar PubMed PubMed Central

[34] Nakamizo S, Egawa G, Honda T, Nakajima S, Belkaid Y, Kabashima K. Commensal bacteria and cutaneous immunity. Semimmunopathology. 2015;37(1):73–80.10.1007/s00281-014-0452-6Search in Google Scholar PubMed

[35] Berube BJ, Bubeck Wardenburg J. Staphylococcus aureus α-toxin: nearly a century of intrigue. Toxins. 2013;5(6):1140–66.10.3390/toxins5061140Search in Google Scholar PubMed PubMed Central

[36] Williams MR, Nakatsuji T, Gallo RL. Staphylococcus aureus: master manipulator of the skin. Cell Host Microbe. 2017;22(5):579–81.10.1016/j.chom.2017.10.015Search in Google Scholar PubMed PubMed Central

[37] Brauweiler AM, Goleva E, Leung DYM. Staphylococcus aureus lipoteichoic acid damages the skin barrier through an IL-1-mediated pathway. J Investig Dermatol. 2019;139(8):1753–61.e4.10.1016/j.jid.2019.02.006Search in Google Scholar PubMed PubMed Central

[38] Cho SH, Strickland I, Tomkinson A, Fehringer AP, Gelfand EW, Leung DY. Preferential binding of Staphylococcus aureus to skin sites of Th2-mediated inflammation in a murine model. J Investig Dermatol. 2001;116(5):658–63.10.1046/j.0022-202x.2001.01331.xSearch in Google Scholar PubMed

[39] Rippke F, Schreiner V, Doering T, Maibach HI. Stratum corneum pH in atopic dermatitis: impact on skin barrier function and colonization with Staphylococcus aureus. Am J Clin Dermatol. 2004;5(4):217–23.10.2165/00128071-200405040-00002Search in Google Scholar PubMed

[40] Christoffersen C, Obinata H, Kumaraswamy SB, Galvani S, Ahnström J, Sevvana M, et al. Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M. Proc Natl Acad Sci USA. 2011;108(23):9613–8.10.1073/pnas.1103187108Search in Google Scholar PubMed PubMed Central

[41] Baker BS. The role of microorganisms in atopic dermatitis. Clin Exp Immunol. 2006;144(1):1–9.10.1111/j.1365-2249.2005.02980.xSearch in Google Scholar PubMed PubMed Central

[42] Edslev SM, Agner T, Andersen PS. Skin microbiome in atopic dermatitis. Acta Dermato-venereologica. 2020;100(12):adv00164.10.2340/00015555-3514Search in Google Scholar PubMed PubMed Central

[43] Wedi B, Wieczorek D, Stünkel T, Breuer K, Kapp A. Staphylococcal exotoxins exert proinflammatory effects through inhibition of eosinophil apoptosis, increased surface antigen expression (CD11b, CD45, CD54, and CD69), and enhanced cytokine-activated oxidative burst, thereby triggering allergic inflammatory reactions. J Allergy Clin Immunol. 2002;109(3):477–84.10.1067/mai.2002.121702Search in Google Scholar PubMed

[44] Ferreri C, Angelini F, Chatgilialoglu C, Dellonte S, Moschese V, Rossi P, et al. Trans fatty acids and atopic eczema/dermatitis syndrome: the relationship with a free radical cis-trans isomerization of membrane lipids. Lipids. 2005;40(7):661–7.10.1007/s11745-005-1428-7Search in Google Scholar PubMed

[45] Watanabe S, Kano R, Sato H, Nakamura Y, Hasegawa A. The effects of Malassezia yeasts on cytokine production by human keratinocytes. J Investig Dermatol. 2001;116(5):769–73.10.1046/j.1523-1747.2001.01321.xSearch in Google Scholar PubMed

[46] Schirbel A, Kessler S, Rieder F, West G, Rebert N, Asosingh K, et al. Pro-angiogenic activity of TLRs and NLRs: a novel link between gut microbiota and intestinal angiogenesis. Gastroenterology. 2013;144(3):613–23.e9.10.1053/j.gastro.2012.11.005Search in Google Scholar PubMed PubMed Central

[47] Wang X, Zhao J, Qin L. VEGF-C mediated enhancement of lymphatic drainage reduces intestinal inflammation by regulating IL-9/IL-17 balance and improving gut microbiota in experimental chronic colitis. Am J Transl Res. 2017;9(11):4772–84.Search in Google Scholar

[48] West CE, Rydén P, Lundin D, Engstrand L, Tulic MK, Prescott SL. Gut microbiome and innate immune response patterns in IgE-associated eczema. Clin Exp Allergy: J Br Soc Allergy Clin Immunol. 2015;45(9):1419–29.10.1111/cea.12566Search in Google Scholar PubMed

[49] Sun S, Luo L, Liang W, Yin Q, Guo J, Rush AM, et al. Bifidobacterium alters the gut microbiota and modulates the functional metabolism of T regulatory cells in the context of immune checkpoint blockade. Proc Natl Acad Sci USA. 2020;117(44):27509–15.10.1073/pnas.1921223117Search in Google Scholar PubMed PubMed Central

[50] Kim IS, Lee SH, Kwon YM, Adhikari B, Kim JA, Yu DY, et al. Oral administration of β-glucan and Lactobacillus plantarum alleviates atopic dermatitis-like symptoms. J Microbiol Biotechnol. 2019;29(11):1693–706.10.4014/jmb.1907.07011Search in Google Scholar PubMed

[51] Fang Z, Li L, Zhang H, Zhao J, Lu W, Chen W. Gut microbiota, probiotics, and their interactions in prevention and treatment of atopic dermatitis: a review. Front Immunol. 2021;12:720393.10.3389/fimmu.2021.720393Search in Google Scholar PubMed PubMed Central

[52] Selander C, Engblom C, Nilsson G, Scheynius A, Andersson CL. TLR2/MyD88-dependent and -independent activation of mast cell IgE responses by the skin commensal yeast Malassezia sympodialis. J Immunol (Baltimore, MD). 2009;182(7):4208–16.10.4049/jimmunol.0800885Search in Google Scholar PubMed

[53] Saini RK, Keum YS. Omega-3 and omega-6 polyunsaturated fatty acids: dietary sources, metabolism, and significance – a review. Life Sci. 2018;203:255–67.10.1016/j.lfs.2018.04.049Search in Google Scholar PubMed

[54] Sato H, Taketomi Y, Isogai Y, Masuda S, Kobayashi T, Yamamoto K, et al. Group III secreted phospholipase A2 transgenic mice spontaneously develop inflammation. Biochem J. 2009;421(1):17–27.10.1042/BJ20082429Search in Google Scholar PubMed PubMed Central

[55] Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277.10.3389/fimmu.2019.00277Search in Google Scholar PubMed PubMed Central

[56] Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014;20(2):159–66.10.1038/nm.3444Search in Google Scholar PubMed

[57] Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science (NY). 2013;341(6145):569–73.10.1126/science.1241165Search in Google Scholar PubMed PubMed Central

[58] Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. 2018;23(6):716–24.10.1016/j.chom.2018.05.003Search in Google Scholar PubMed

[59] Fang Z, Pan T, Li L, Wang H, Zhu J, Zhang H, et al. Bifidobacterium longum mediated tryptophan metabolism to improve atopic dermatitis via the gut–skin axis. Gut Microbes. 2022;14(1):2044723.10.1080/19490976.2022.2044723Search in Google Scholar PubMed PubMed Central

[60] Matsumoto M, Ebata T, Hirooka J, Hosoya R, Inoue N, Itami S, et al. Antipruritic effects of the probiotic strain LKM512 in adults with atopic dermatitis. Ann Allergy Asthma Immunol. 2014;113(2):209–16.e7.10.1016/j.anai.2014.05.002Search in Google Scholar PubMed

[61] Matsumoto M, Aranami A, Ishige A, Watanabe K, Benno Y. LKM512 yogurt consumption improves the intestinal environment and induces the T-helper type 1 cytokine in adult patients with intractable atopic dermatitis. Clin Exp Allergy: J Br Soc Allergy Clin Immunol. 2007;37(3):358–70.10.1111/j.1365-2222.2007.02642.xSearch in Google Scholar PubMed

[62] Matsumoto M, Kakizoe K, Benno Y. Comparison of fecal microbiota and polyamine concentration in adult patients with intractable atopic dermatitis and healthy adults. Microbiol Immunol. 2007;51(1):37–46.10.1111/j.1348-0421.2007.tb03888.xSearch in Google Scholar PubMed

[63] Smith SH, Jayawickreme C, Rickard DJ, Nicodeme E, Bui T, Simmons C, et al. Tapinarof is a natural AhR agonist that resolves skin inflammation in mice and humans. J Investig Dermatol. 2017;137(10):2110–9.10.1016/j.jid.2017.05.004Search in Google Scholar PubMed

[64] Alonso C, Guilarte M, Vicario M, Ramos L, Ramadan Z, Antolín M, et al. Maladaptive intestinal epithelial responses to life stress may predispose healthy women to gut mucosal inflammation. Gastroenterology. 2008;135(1):163–72.e1.10.1053/j.gastro.2008.03.036Search in Google Scholar PubMed

[65] Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication. Neurogastroenterol Motil. 2011;23(12):1132–9.10.1111/j.1365-2982.2011.01796.xSearch in Google Scholar PubMed PubMed Central

[66] Lonne-Rahm SB, Rickberg H, El-Nour H, Mårin P, Azmitia EC, Nordlind K. Neuroimmune mechanisms in patients with atopic dermatitis during chronic stress. J Eur Acad Dermatol Venereol: JEADV. 2008;22(1):11–8.10.1111/j.1468-3083.2007.02202.xSearch in Google Scholar PubMed PubMed Central

[67] Hendrix S. Neuroimmune communication in skin: far from peripheral. J Investig Dermatol. 2008;128(2):260–1.10.1038/sj.jid.5701171Search in Google Scholar PubMed

[68] Roosterman D, Goerge T, Schneider SW, Bunnett NW, Steinhoff M. Neuronal control of skin function: the skin as a neuroimmunoendocrine organ. Physiol Rev. 2006;86(4):1309–79.10.1152/physrev.00026.2005Search in Google Scholar PubMed

[69] Teresiak-Mikołajczak E, Czarnecka-Operacz M, Jenerowicz D, Silny W. Neurogenic markers of the inflammatory process in atopic dermatitis: relation to the severity and pruritus. Postepy Dermatologii Alergologii. 2013;30(5):286–92.10.5114/pdia.2013.38357Search in Google Scholar PubMed PubMed Central

[70] Lee HJ, Son Y, Lee M, Moon C, Kim SH, Shin IS, et al. Sodium butyrate prevents radiation-induced cognitive impairment by restoring pCREB/BDNF expression. Neural Regener Res. 2019;14(9):1530–5.10.4103/1673-5374.255974Search in Google Scholar PubMed PubMed Central

[71] Valles-Colomer M, Falony G, Darzi Y, Tigchelaar EF, Wang J, Tito RY, et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol. 2019;4(4):623–32.10.1038/s41564-018-0337-xSearch in Google Scholar PubMed

[72] Morita T, McClain SP, Batia LM, Pellegrino M, Wilson SR, Kienzler MA, et al. HTR7 mediates serotonergic acute and chronic itch. Neuron. 2015;87(1):124–38.10.1016/j.neuron.2015.05.044Search in Google Scholar PubMed PubMed Central

[73] Han D, Kim HY, Lee HJ, Shim I, Hahm DH. Wound healing activity of gamma-aminobutyric acid (GABA) in rats. J Microbiol Biotechnol. 2007;17(10):1661–9.Search in Google Scholar

[74] Nigam R, El-Nour H, Amatya B. Nordlind KGABA. and GABA(A) receptor expression on immune cells in psoriasis: a pathophysiological role. Arch Dermatol Res. 2010;302(7):507–15.10.1007/s00403-010-1052-5Search in Google Scholar PubMed

[75] Bailey MT, Dowd SE, Galley JD, Hufnagle AR, Allen RG, Lyte M. Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain, Behavior, Immun. 2011;25(3):397–407.10.1016/j.bbi.2010.10.023Search in Google Scholar PubMed PubMed Central

[76] Wilson SR, Gerhold KA, Bifolck-Fisher A, Liu Q, Patel KN, Dong X, et al. TRPA1 is required for histamine-independent, Mas-related G protein-coupled receptor-mediated itch. Nat Neurosci. 2011;14(5):595–602.10.1038/nn.2789Search in Google Scholar PubMed PubMed Central

[77] Fujii M. Current understanding of pathophysiological mechanisms of atopic dermatitis: interactions among skin barrier dysfunction, immune abnormalities and pruritus. Biol Pharm Bull. 2020;43(1):12–9.10.1248/bpb.b19-00088Search in Google Scholar PubMed

[78] Foster RH, Hardy G, Alany RG. Borage oil in the treatment of atopic dermatitis. Nutrition (Burbank, Los Angeles County, Calif). 2010;26(7–8):708–18.10.1016/j.nut.2009.10.014Search in Google Scholar PubMed

[79] Drake MG, Scott GD, Blum ED, Lebold KM, Nie Z, Lee JJ, et al. Eosinophils increase airway sensory nerve density in mice and in human asthma. Sci Transl Med. 2018;10(457):eaar8447.10.1126/scitranslmed.aar8477Search in Google Scholar PubMed PubMed Central

[80] Lyte M. Microbial endocrinology and the microbiota–gut–brain axis. Adv Exp Med Biol. 2014;817:3–24.10.1007/978-1-4939-0897-4_1Search in Google Scholar PubMed

[81] Muddasani S, Rusk AM, Baquerizo Nole KL. Gluten and skin disease beyond dermatitis herpetiformis: a review. Int J Dermatol. 2021;60(3):281–8.10.1111/ijd.15098Search in Google Scholar PubMed

[82] Ciacci C, Cavallaro R, Iovino P, Sabbatini F, Palumbo A, Amoruso D, et al. Allergy prevalence in adult celiac disease. J Allergy Clin Immunol. 2004;113(6):1199–203.10.1016/j.jaci.2004.03.012Search in Google Scholar PubMed

[83] de Sousa Moraes LF, Grzeskowiak LM, de Sales Teixeira TF, Gouveia Peluzio Mdo C. Intestinal microbiota and probiotics in celiac disease. Clin Microbiol Rev. 2014;27(3):482–9.10.1128/CMR.00106-13Search in Google Scholar PubMed PubMed Central

[84] Niemeyer-van der Kolk T, van der Wall H, Hogendoorn GK, Rijneveld R, Luijten S, van Alewijk D, et al. Pharmacodynamic effects of topical omiganan in patients with mild to moderate atopic dermatitis in a randomized, placebo-controlled, phase II trial. Clin Transl Sci. 2020;13(5):994–1003.10.1111/cts.12792Search in Google Scholar PubMed PubMed Central

[85] Niemeyer-van der Kolk T, Assil S, Buters TP, Rijsbergen M, Klaassen ES, Feiss G, et al. Omiganan enhances imiquimod-induced inflammatory responses in skin of healthy volunteers. Clin Transl Sci. 2020;13(3):573–9.10.1111/cts.12741Search in Google Scholar PubMed PubMed Central

[86] Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med. 2017;9(378):eaah4680.10.1126/scitranslmed.aah4680Search in Google Scholar PubMed PubMed Central

[87] Zipperer A, Konnerth MC, Laux C, Berscheid A, Janek D, Weidenmaier C, et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature. 2016;535(7613):511–6.10.1038/nature18634Search in Google Scholar PubMed

[88] Cogen AL, Yamasaki K, Muto J, Sanchez KM, Crotty Alexander L, Tanios J, et al. Staphylococcus epidermidis antimicrobial delta-toxin (phenol-soluble modulin-gamma) cooperates with host antimicrobial peptides to kill group A Streptococcus. PLoS One. 2010;5(1):e8557.10.1371/journal.pone.0008557Search in Google Scholar PubMed PubMed Central

[89] Myles IA, Williams KW, Reckhow JD, Jammeh ML, Pincus NB, Sastalla I, et al. Transplantation of human skin microbiota in models of atopic dermatitis. JCI Insight. 2016;1(10):e86955.10.1172/jci.insight.86955Search in Google Scholar PubMed PubMed Central

[90] Tannock GW, Munro K, Harmsen HJ, Welling GW, Smart J, Gopal PK. Analysis of the fecal microflora of human subjects consuming a probiotic product containing Lactobacillus rhamnosus DR20. Appl Environ Microbiol. 2000;66(6):2578–88.10.1128/AEM.66.6.2578-2588.2000Search in Google Scholar PubMed PubMed Central

[91] Kim JH, Kim K, Kim W. Gut microbiota restoration through fecal microbiota transplantation: a new atopic dermatitis therapy. Exp Mol Med. 2021;53(5):907–16.10.1038/s12276-021-00627-6Search in Google Scholar PubMed PubMed Central

[92] Mashiah J, Karady T, Fliss-Isakov N, Sprecher E, Slodownik D, Artzi O, et al. Clinical efficacy of fecal microbial transplantation treatment in adults with moderate-to-severe atopic dermatitis. Immun Inflamm Dis. 2022;10(3):e570.10.1002/iid3.570Search in Google Scholar PubMed PubMed Central

[93] Horrobin DF. Essential fatty acid metabolism and its modification in atopic eczema. Am J Clin Nutr. 2000;71(1 Suppl):367s–72ss.10.1093/ajcn/71.1.367SSearch in Google Scholar PubMed

[94] Lauridsen C. Effects of dietary fatty acids on gut health and function of pigs pre- and post-weaning. J Anim Sci. 2020;98(4):1–12.10.1093/jas/skaa086Search in Google Scholar PubMed PubMed Central

[95] Balić A, Vlašić D, Žužul K, Marinović B, Bukvić Mokos Z. Omega-3 versus omega-6 polyunsaturated fatty acids in the prevention and treatment of inflammatory skin diseases. Int J Mol Sci. 2020;21(3):741.10.3390/ijms21030741Search in Google Scholar PubMed PubMed Central

[96] Li X, Bi X, Wang S, Zhang Z, Li F, Zhao AZ. Therapeutic potential of ω-3 polyunsaturated fatty acids in human autoimmune diseases. Front Immunol. 2019;10:2241.10.3389/fimmu.2019.02241Search in Google Scholar PubMed PubMed Central

[97] Calder PC. Is increasing microbiota diversity a novel anti-inflammatory action of marine n-3 fatty acids? J Nutr. 2019;149(7):1102–4.10.1093/jn/nxz043Search in Google Scholar PubMed

Received: 2023-08-23
Revised: 2023-10-26
Accepted: 2023-10-27
Published Online: 2024-04-13

© 2024 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Biomedical Sciences
  2. Constitutive and evoked release of ATP in adult mouse olfactory epithelium
  3. LARP1 knockdown inhibits cultured gastric carcinoma cell cycle progression and metastatic behavior
  4. PEGylated porcine–human recombinant uricase: A novel fusion protein with improved efficacy and safety for the treatment of hyperuricemia and renal complications
  5. Research progress on ocular complications caused by type 2 diabetes mellitus and the function of tears and blepharons
  6. The role and mechanism of esketamine in preventing and treating remifentanil-induced hyperalgesia based on the NMDA receptor–CaMKII pathway
  7. Brucella infection combined with Nocardia infection: A case report and literature review
  8. Detection of serum interleukin-18 level and neutrophil/lymphocyte ratio in patients with antineutrophil cytoplasmic antibody-associated vasculitis and its clinical significance
  9. Ang-1, Ang-2, and Tie2 are diagnostic biomarkers for Henoch-Schönlein purpura and pediatric-onset systemic lupus erythematous
  10. PTTG1 induces pancreatic cancer cell proliferation and promotes aerobic glycolysis by regulating c-myc
  11. Role of serum B-cell-activating factor and interleukin-17 as biomarkers in the classification of interstitial pneumonia with autoimmune features
  12. Effectiveness and safety of a mumps containing vaccine in preventing laboratory-confirmed mumps cases from 2002 to 2017: A meta-analysis
  13. Low levels of sex hormone-binding globulin predict an increased breast cancer risk and its underlying molecular mechanisms
  14. A case of Trousseau syndrome: Screening, detection and complication
  15. Application of the integrated airway humidification device enhances the humidification effect of the rabbit tracheotomy model
  16. Preparation of Cu2+/TA/HAP composite coating with anti-bacterial and osteogenic potential on 3D-printed porous Ti alloy scaffolds for orthopedic applications
  17. Aquaporin-8 promotes human dermal fibroblasts to counteract hydrogen peroxide-induced oxidative damage: A novel target for management of skin aging
  18. Current research and evidence gaps on placental development in iron deficiency anemia
  19. Single-nucleotide polymorphism rs2910829 in PDE4D is related to stroke susceptibility in Chinese populations: The results of a meta-analysis
  20. Pheochromocytoma-induced myocardial infarction: A case report
  21. Kaempferol regulates apoptosis and migration of neural stem cells to attenuate cerebral infarction by O‐GlcNAcylation of β-catenin
  22. Sirtuin 5 regulates acute myeloid leukemia cell viability and apoptosis by succinylation modification of glycine decarboxylase
  23. Apigenin 7-glucoside impedes hypoxia-induced malignant phenotypes of cervical cancer cells in a p16-dependent manner
  24. KAT2A changes the function of endometrial stromal cells via regulating the succinylation of ENO1
  25. Current state of research on copper complexes in the treatment of breast cancer
  26. Exploring antioxidant strategies in the pathogenesis of ALS
  27. Helicobacter pylori causes gastric dysbacteriosis in chronic gastritis patients
  28. IL-33/soluble ST2 axis is associated with radiation-induced cardiac injury
  29. The predictive value of serum NLR, SII, and OPNI for lymph node metastasis in breast cancer patients with internal mammary lymph nodes after thoracoscopic surgery
  30. Carrying SNP rs17506395 (T > G) in TP63 gene and CCR5Δ32 mutation associated with the occurrence of breast cancer in Burkina Faso
  31. P2X7 receptor: A receptor closely linked with sepsis-associated encephalopathy
  32. Probiotics for inflammatory bowel disease: Is there sufficient evidence?
  33. Identification of KDM4C as a gene conferring drug resistance in multiple myeloma
  34. Microbial perspective on the skin–gut axis and atopic dermatitis
  35. Thymosin α1 combined with XELOX improves immune function and reduces serum tumor markers in colorectal cancer patients after radical surgery
  36. Highly specific vaginal microbiome signature for gynecological cancers
  37. Sample size estimation for AQP4-IgG seropositive optic neuritis: Retinal damage detection by optical coherence tomography
  38. The effects of SDF-1 combined application with VEGF on femoral distraction osteogenesis in rats
  39. Fabrication and characterization of gold nanoparticles using alginate: In vitro and in vivo assessment of its administration effects with swimming exercise on diabetic rats
  40. Mitigating digestive disorders: Action mechanisms of Mediterranean herbal active compounds
  41. Distribution of CYP2D6 and CYP2C19 gene polymorphisms in Han and Uygur populations with breast cancer in Xinjiang, China
  42. VSP-2 attenuates secretion of inflammatory cytokines induced by LPS in BV2 cells by mediating the PPARγ/NF-κB signaling pathway
  43. Factors influencing spontaneous hypothermia after emergency trauma and the construction of a predictive model
  44. Long-term administration of morphine specifically alters the level of protein expression in different brain regions and affects the redox state
  45. Application of metagenomic next-generation sequencing technology in the etiological diagnosis of peritoneal dialysis-associated peritonitis
  46. Clinical diagnosis, prevention, and treatment of neurodyspepsia syndrome using intelligent medicine
  47. Case report: Successful bronchoscopic interventional treatment of endobronchial leiomyomas
  48. Preliminary investigation into the genetic etiology of short stature in children through whole exon sequencing of the core family
  49. Cystic adenomyoma of the uterus: Case report and literature review
  50. Mesoporous silica nanoparticles as a drug delivery mechanism
  51. Dynamic changes in autophagy activity in different degrees of pulmonary fibrosis in mice
  52. Vitamin D deficiency and inflammatory markers in type 2 diabetes: Big data insights
  53. Lactate-induced IGF1R protein lactylation promotes proliferation and metabolic reprogramming of lung cancer cells
  54. Meta-analysis on the efficacy of allogeneic hematopoietic stem cell transplantation to treat malignant lymphoma
  55. Mitochondrial DNA drives neuroinflammation through the cGAS-IFN signaling pathway in the spinal cord of neuropathic pain mice
  56. Application value of artificial intelligence algorithm-based magnetic resonance multi-sequence imaging in staging diagnosis of cervical cancer
  57. Embedded monitoring system and teaching of artificial intelligence online drug component recognition
  58. Investigation into the association of FNDC1 and ADAMTS12 gene expression with plumage coloration in Muscovy ducks
  59. Yak meat content in feed and its impact on the growth of rats
  60. A rare case of Richter transformation with breast involvement: A case report and literature review
  61. First report of Nocardia wallacei infection in an immunocompetent patient in Zhejiang province
  62. Rhodococcus equi and Brucella pulmonary mass in immunocompetent: A case report and literature review
  63. Downregulation of RIP3 ameliorates the left ventricular mechanics and function after myocardial infarction via modulating NF-κB/NLRP3 pathway
  64. Evaluation of the role of some non-enzymatic antioxidants among Iraqi patients with non-alcoholic fatty liver disease
  65. The role of Phafin proteins in cell signaling pathways and diseases
  66. Ten-year anemia as initial manifestation of Castleman disease in the abdominal cavity: A case report
  67. Coexistence of hereditary spherocytosis with SPTB P.Trp1150 gene variant and Gilbert syndrome: A case report and literature review
  68. Utilization of convolutional neural networks to analyze microscopic images for high-throughput screening of mesenchymal stem cells
  69. Exploratory evaluation supported by experimental and modeling approaches of Inula viscosa root extract as a potent corrosion inhibitor for mild steel in a 1 M HCl solution
  70. Imaging manifestations of ductal adenoma of the breast: A case report
  71. Gut microbiota and sleep: Interaction mechanisms and therapeutic prospects
  72. Isomangiferin promotes the migration and osteogenic differentiation of rat bone marrow mesenchymal stem cells
  73. Prognostic value and microenvironmental crosstalk of exosome-related signatures in human epidermal growth factor receptor 2 positive breast cancer
  74. Circular RNAs as potential biomarkers for male severe sepsis
  75. Knockdown of Stanniocalcin-1 inhibits growth and glycolysis in oral squamous cell carcinoma cells
  76. The expression and biological role of complement C1s in esophageal squamous cell carcinoma
  77. A novel GNAS mutation in pseudohypoparathyroidism type 1a with articular flexion deformity: A case report
  78. Predictive value of serum magnesium levels for prognosis in patients with non-small cell lung cancer undergoing EGFR-TKI therapy
  79. HSPB1 alleviates acute-on-chronic liver failure via the P53/Bax pathway
  80. IgG4-related disease complicated by PLA2R-associated membranous nephropathy: A case report
  81. Baculovirus-mediated endostatin and angiostatin activation of autophagy through the AMPK/AKT/mTOR pathway inhibits angiogenesis in hepatocellular carcinoma
  82. Metformin mitigates osteoarthritis progression by modulating the PI3K/AKT/mTOR signaling pathway and enhancing chondrocyte autophagy
  83. Evaluation of the activity of antimicrobial peptides against bacterial vaginosis
  84. Atypical presentation of γ/δ mycosis fungoides with an unusual phenotype and SOCS1 mutation
  85. Analysis of the microecological mechanism of diabetic kidney disease based on the theory of “gut–kidney axis”: A systematic review
  86. Omega-3 fatty acids prevent gestational diabetes mellitus via modulation of lipid metabolism
  87. Refractory hypertension complicated with Turner syndrome: A case report
  88. Interaction of ncRNAs and the PI3K/AKT/mTOR pathway: Implications for osteosarcoma
  89. Association of low attenuation area scores with pulmonary function and clinical prognosis in patients with chronic obstructive pulmonary disease
  90. Long non-coding RNAs in bone formation: Key regulators and therapeutic prospects
  91. The deubiquitinating enzyme USP35 regulates the stability of NRF2 protein
  92. Neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio as potential diagnostic markers for rebleeding in patients with esophagogastric variceal bleeding
  93. G protein-coupled receptor 1 participating in the mechanism of mediating gestational diabetes mellitus by phosphorylating the AKT pathway
  94. LL37-mtDNA regulates viability, apoptosis, inflammation, and autophagy in lipopolysaccharide-treated RLE-6TN cells by targeting Hsp90aa1
  95. The analgesic effect of paeoniflorin: A focused review
  96. Chemical composition’s effect on Solanum nigrum Linn.’s antioxidant capacity and erythrocyte protection: Bioactive components and molecular docking analysis
  97. Knockdown of HCK promotes HREC cell viability and inner blood–retinal barrier integrity by regulating the AMPK signaling pathway
  98. The role of rapamycin in the PINK1/Parkin signaling pathway in mitophagy in podocytes
  99. Laryngeal non-Hodgkin lymphoma: Report of four cases and review of the literature
  100. Clinical value of macrogenome next-generation sequencing on infections
  101. Overview of dendritic cells and related pathways in autoimmune uveitis
  102. TAK-242 alleviates diabetic cardiomyopathy via inhibiting pyroptosis and TLR4/CaMKII/NLRP3 pathway
  103. Hypomethylation in promoters of PGC-1α involved in exercise-driven skeletal muscular alterations in old age
  104. Profile and antimicrobial susceptibility patterns of bacteria isolated from effluents of Kolladiba and Debark hospitals
  105. The expression and clinical significance of syncytin-1 in serum exosomes of hepatocellular carcinoma patients
  106. A histomorphometric study to evaluate the therapeutic effects of biosynthesized silver nanoparticles on the kidneys infected with Plasmodium chabaudi
  107. PGRMC1 and PAQR4 are promising molecular targets for a rare subtype of ovarian cancer
  108. Analysis of MDA, SOD, TAOC, MNCV, SNCV, and TSS scores in patients with diabetes peripheral neuropathy
  109. SLIT3 deficiency promotes non-small cell lung cancer progression by modulating UBE2C/WNT signaling
  110. The relationship between TMCO1 and CALR in the pathological characteristics of prostate cancer and its effect on the metastasis of prostate cancer cells
  111. Heterogeneous nuclear ribonucleoprotein K is a potential target for enhancing the chemosensitivity of nasopharyngeal carcinoma
  112. PHB2 alleviates retinal pigment epithelium cell fibrosis by suppressing the AGE–RAGE pathway
  113. Anti-γ-aminobutyric acid-B receptor autoimmune encephalitis with syncope as the initial symptom: Case report and literature review
  114. Comparative analysis of chloroplast genome of Lonicera japonica cv. Damaohua
  115. Human umbilical cord mesenchymal stem cells regulate glutathione metabolism depending on the ERK–Nrf2–HO-1 signal pathway to repair phosphoramide mustard-induced ovarian cancer cells
  116. Electroacupuncture on GB acupoints improves osteoporosis via the estradiol–PI3K–Akt signaling pathway
  117. Renalase protects against podocyte injury by inhibiting oxidative stress and apoptosis in diabetic nephropathy
  118. Review: Dicranostigma leptopodum: A peculiar plant of Papaveraceae
  119. Combination effect of flavonoids attenuates lung cancer cell proliferation by inhibiting the STAT3 and FAK signaling pathway
  120. Renal microangiopathy and immune complex glomerulonephritis induced by anti-tumour agents: A case report
  121. Correlation analysis of AVPR1a and AVPR2 with abnormal water and sodium and potassium metabolism in rats
  122. Gastrointestinal health anti-diarrheal mixture relieves spleen deficiency-induced diarrhea through regulating gut microbiota
  123. Myriad factors and pathways influencing tumor radiotherapy resistance
  124. Exploring the effects of culture conditions on Yapsin (YPS) gene expression in Nakaseomyces glabratus
  125. Screening of prognostic core genes based on cell–cell interaction in the peripheral blood of patients with sepsis
  126. Coagulation factor II thrombin receptor as a promising biomarker in breast cancer management
  127. Ileocecal mucinous carcinoma misdiagnosed as incarcerated hernia: A case report
  128. Methyltransferase like 13 promotes malignant behaviors of bladder cancer cells through targeting PI3K/ATK signaling pathway
  129. The debate between electricity and heat, efficacy and safety of irreversible electroporation and radiofrequency ablation in the treatment of liver cancer: A meta-analysis
  130. ZAG promotes colorectal cancer cell proliferation and epithelial–mesenchymal transition by promoting lipid synthesis
  131. Baicalein inhibits NLRP3 inflammasome activation and mitigates placental inflammation and oxidative stress in gestational diabetes mellitus
  132. Impact of SWCNT-conjugated senna leaf extract on breast cancer cells: A potential apoptotic therapeutic strategy
  133. MFAP5 inhibits the malignant progression of endometrial cancer cells in vitro
  134. Major ozonated autohemotherapy promoted functional recovery following spinal cord injury in adult rats via the inhibition of oxidative stress and inflammation
  135. Axodendritic targeting of TAU and MAP2 and microtubule polarization in iPSC-derived versus SH-SY5Y-derived human neurons
  136. Differential expression of phosphoinositide 3-kinase/protein kinase B and Toll-like receptor/nuclear factor kappa B signaling pathways in experimental obesity Wistar rat model
  137. The therapeutic potential of targeting Oncostatin M and the interleukin-6 family in retinal diseases: A comprehensive review
  138. BA inhibits LPS-stimulated inflammatory response and apoptosis in human middle ear epithelial cells by regulating the Nf-Kb/Iκbα axis
  139. Role of circRMRP and circRPL27 in chronic obstructive pulmonary disease
  140. Investigating the role of hyperexpressed HCN1 in inducing myocardial infarction through activation of the NF-κB signaling pathway
  141. Characterization of phenolic compounds and evaluation of anti-diabetic potential in Cannabis sativa L. seeds: In vivo, in vitro, and in silico studies
  142. Quantitative immunohistochemistry analysis of breast Ki67 based on artificial intelligence
  143. Ecology and Environmental Science
  144. Screening of different growth conditions of Bacillus subtilis isolated from membrane-less microbial fuel cell toward antimicrobial activity profiling
  145. Degradation of a mixture of 13 polycyclic aromatic hydrocarbons by commercial effective microorganisms
  146. Evaluation of the impact of two citrus plants on the variation of Panonychus citri (Acari: Tetranychidae) and beneficial phytoseiid mites
  147. Prediction of present and future distribution areas of Juniperus drupacea Labill and determination of ethnobotany properties in Antalya Province, Türkiye
  148. Population genetics of Todarodes pacificus (Cephalopoda: Ommastrephidae) in the northwest Pacific Ocean via GBS sequencing
  149. A comparative analysis of dendrometric, macromorphological, and micromorphological characteristics of Pistacia atlantica subsp. atlantica and Pistacia terebinthus in the middle Atlas region of Morocco
  150. Macrofungal sporocarp community in the lichen Scots pine forests
  151. Assessing the proximate compositions of indigenous forage species in Yemen’s pastoral rangelands
  152. Food Science
  153. Gut microbiota changes associated with low-carbohydrate diet intervention for obesity
  154. Reexamination of Aspergillus cristatus phylogeny in dark tea: Characteristics of the mitochondrial genome
  155. Differences in the flavonoid composition of the leaves, fruits, and branches of mulberry are distinguished based on a plant metabolomics approach
  156. Investigating the impact of wet rendering (solventless method) on PUFA-rich oil from catfish (Clarias magur) viscera
  157. Non-linear associations between cardiovascular metabolic indices and metabolic-associated fatty liver disease: A cross-sectional study in the US population (2017–2020)
  158. Knockdown of USP7 alleviates atherosclerosis in ApoE-deficient mice by regulating EZH2 expression
  159. Utility of dairy microbiome as a tool for authentication and traceability
  160. Agriculture
  161. Enhancing faba bean (Vicia faba L.) productivity through establishing the area-specific fertilizer rate recommendation in southwest Ethiopia
  162. Impact of novel herbicide based on synthetic auxins and ALS inhibitor on weed control
  163. Perspectives of pteridophytes microbiome for bioremediation in agricultural applications
  164. Fertilizer application parameters for drip-irrigated peanut based on the fertilizer effect function established from a “3414” field trial
  165. Improving the productivity and profitability of maize (Zea mays L.) using optimum blended inorganic fertilization
  166. Application of leaf multispectral analyzer in comparison to hyperspectral device to assess the diversity of spectral reflectance indices in wheat genotypes
  167. Animal Sciences
  168. Knockdown of ANP32E inhibits colorectal cancer cell growth and glycolysis by regulating the AKT/mTOR pathway
  169. Development of a detection chip for major pathogenic drug-resistant genes and drug targets in bovine respiratory system diseases
  170. Exploration of the genetic influence of MYOT and MB genes on the plumage coloration of Muscovy ducks
  171. Transcriptome analysis of adipose tissue in grazing cattle: Identifying key regulators of fat metabolism
  172. Comparison of nutritional value of the wild and cultivated spiny loaches at three growth stages
  173. Transcriptomic analysis of liver immune response in Chinese spiny frog (Quasipaa spinosa) infected with Proteus mirabilis
  174. Disruption of BCAA degradation is a critical characteristic of diabetic cardiomyopathy revealed by integrated transcriptome and metabolome analysis
  175. Plant Sciences
  176. Effect of long-term in-row branch covering on soil microorganisms in pear orchards
  177. Photosynthetic physiological characteristics, growth performance, and element concentrations reveal the calcicole–calcifuge behaviors of three Camellia species
  178. Transcriptome analysis reveals the mechanism of NaHCO3 promoting tobacco leaf maturation
  179. Bioinformatics, expression analysis, and functional verification of allene oxide synthase gene HvnAOS1 and HvnAOS2 in qingke
  180. Water, nitrogen, and phosphorus coupling improves gray jujube fruit quality and yield
  181. Improving grape fruit quality through soil conditioner: Insights from RNA-seq analysis of Cabernet Sauvignon roots
  182. Role of Embinin in the reabsorption of nucleus pulposus in lumbar disc herniation: Promotion of nucleus pulposus neovascularization and apoptosis of nucleus pulposus cells
  183. Revealing the effects of amino acid, organic acid, and phytohormones on the germination of tomato seeds under salinity stress
  184. Combined effects of nitrogen fertilizer and biochar on the growth, yield, and quality of pepper
  185. Comprehensive phytochemical and toxicological analysis of Chenopodium ambrosioides (L.) fractions
  186. Impact of “3414” fertilization on the yield and quality of greenhouse tomatoes
  187. Exploring the coupling mode of water and fertilizer for improving growth, fruit quality, and yield of the pear in the arid region
  188. Metagenomic analysis of endophytic bacteria in seed potato (Solanum tuberosum)
  189. Antibacterial, antifungal, and phytochemical properties of Salsola kali ethanolic extract
  190. Exploring the hepatoprotective properties of citronellol: In vitro and in silico studies on ethanol-induced damage in HepG2 cells
  191. Enhanced osmotic dehydration of watermelon rind using honey–sucrose solutions: A study on pre-treatment efficacy and mass transfer kinetics
  192. Effects of exogenous 2,4-epibrassinolide on photosynthetic traits of 53 cowpea varieties under NaCl stress
  193. Comparative transcriptome analysis of maize (Zea mays L.) seedlings in response to copper stress
  194. An optimization method for measuring the stomata in cassava (Manihot esculenta Crantz) under multiple abiotic stresses
  195. Fosinopril inhibits Ang II-induced VSMC proliferation, phenotype transformation, migration, and oxidative stress through the TGF-β1/Smad signaling pathway
  196. Antioxidant and antimicrobial activities of Salsola imbricata methanolic extract and its phytochemical characterization
  197. Bioengineering and Biotechnology
  198. Absorbable calcium and phosphorus bioactive membranes promote bone marrow mesenchymal stem cells osteogenic differentiation for bone regeneration
  199. New advances in protein engineering for industrial applications: Key takeaways
  200. An overview of the production and use of Bacillus thuringiensis toxin
  201. Research progress of nanoparticles in diagnosis and treatment of hepatocellular carcinoma
  202. Bioelectrochemical biosensors for water quality assessment and wastewater monitoring
  203. PEI/MMNs@LNA-542 nanoparticles alleviate ICU-acquired weakness through targeted autophagy inhibition and mitochondrial protection
  204. Unleashing of cytotoxic effects of thymoquinone-bovine serum albumin nanoparticles on A549 lung cancer cells
  205. Erratum
  206. Erratum to “Investigating the association between dietary patterns and glycemic control among children and adolescents with T1DM”
  207. Erratum to “Activation of hypermethylated P2RY1 mitigates gastric cancer by promoting apoptosis and inhibiting proliferation”
  208. Retraction
  209. Retraction to “MiR-223-3p regulates cell viability, migration, invasion, and apoptosis of non-small cell lung cancer cells by targeting RHOB”
  210. Retraction to “A data mining technique for detecting malignant mesothelioma cancer using multiple regression analysis”
  211. Special Issue on Advances in Neurodegenerative Disease Research and Treatment
  212. Transplantation of human neural stem cell prevents symptomatic motor behavior disability in a rat model of Parkinson’s disease
  213. Special Issue on Multi-omics
  214. Inflammasome complex genes with clinical relevance suggest potential as therapeutic targets for anti-tumor drugs in clear cell renal cell carcinoma
  215. Gastroesophageal varices in primary biliary cholangitis with anti-centromere antibody positivity: Early onset?
Downloaded on 26.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/biol-2022-0782/html?lang=en
Scroll to top button