International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
The skin, as the largest organ of the human body, provides a protective barrier against mechanical, microbial, chemical and allergenic insults (1). The gastrointestinal tract, a crucial mucosal immune organ, maintains immune homeostasis through dynamic interactions between the microbiome and the intestinal immune system (2). The intestinal mucosal immune function is mediated by the mucus layer, epithelial barrier and resident immune cells, all of which engage with the gut microbiota (3). The collective genome of gut microbes is termed the 'gut microbiome,' which is intimately linked to long-term health (4). As interfaces with the external environment, both the gut and skin host diverse microbial communities and are richly innervated and vascularized. As early as 1930, John H. Stokes and Donald M. Pillsbury proposed an intrinsic relationship between gut microbiota and skin inflammation, conceptualizing the 'gut-skin axis' (5). Recent advances in microbiology and immunology have begun to clarify the mechanisms through which gut microbiota influences skin health. Lee and Sung (6) identified immune pathways by which alterations in gut and skin microbiota contribute to dermatopathology, while Szanto et al (7) suggested that gut microbiota modulation may hold therapeutic potential for specific skin disorders. These findings not only validate the gut-skin axis theory but also open new avenues for clinical dermatology. In the context of increasing antibiotic resistance, therapeutic strategies are shifting toward alternatives such as probiotics, prebiotics and dietary interventions, which can restore microbial balance and modulate immune responses to improve skin health (8,9). This review examines novel strategies for treating skin diseases from the perspective of the gut-skin axis and explores future translational research directions.
The gut microbiota comprises diverse and dynamic microbial communities residing in the human gastrointestinal tract (10) (Fig. 1A). Anatomically and functionally, the gut is divided into the small and large intestines, each with distinct physiological conditions and microbial populations (11). Numerous factors influence the gut microbiota (Fig. 1B), with diet being a primary determinant of its structure and function (12). The gut microbiota evolves from infancy and changes in composition and diversity over time within an individual (13). There is considerable interindividual variation in gut microbial composition. Through host-microbe coevolution, the gut microbiome plays a critical role in regulating host physiology, including metabolism, immune development and behavioral responses (14,15).
As the body's outermost barrier, the skin is continuously exposed to environmental factors and serves as the first line of immune defense (16). The skin microbiota consists of microbial communities adapted to the cutaneous environment through long-term colonization, persisting in the chemical milieu of the stratum corneum, sweat and sebaceous secretions (17). Environmental factors such as ultraviolet radiation, temperature, humidity, sebum levels, oxygen availability and pH create distinct ecological niches across skin regions, leading to spatial variation in microbial composition (18). Based on physiological characteristics, Mahmud et al (19) classified skin into sebaceous (e.g., between eyebrows), moist (e.g., forearm flexure) and dry (e.g., palmar forearm) types. Lipophilic bacteria dominate sebum-rich areas, whereas dry regions may support a more diverse microbiota (Fig. 1C).
According to Petersen and Round (20), dysbiosis refers to alterations in complex biological communities. In the context of the gut microbiome, dysbiosis manifests in three primary forms: Loss of beneficial microbes, overgrowth of pathogenic microorganisms and reduced overall microbial diversity. These forms may occur independently or concurrently (20). In microbiome research, α-diversity measures species richness within a sample, while β-diversity assesses structural differences between samples, revealing characteristic dysbiosis patterns in skin diseases (21). Key factors contributing to gut microbiota imbalance include perinatal factors (e.g., mode of delivery), lifestyle (diet, stress, smoking) and medical interventions (e.g., antibiotics, chemotherapy) (Fig. 1B). Gut dysbiosis has been implicated in numerous diseases, including common skin disorders such as atopic dermatitis (AD), psoriasis, acne and alopecia areata (21). Research on rare skin diseases like lichen planus (LP) and autoimmune bullous dermatoses (AIBD) may also find breakthroughs through the gut microbiome (22).
The 'gut-skin axis' theory is part of the broader 'gut-organ axis' framework, emphasizing bidirectional communication between the gut and other organs via neurological, endocrine and immune pathways (23). This theory integrates multiple organs, the gut and the immune system with the gut microbiota (24). The gut and skin share structural and functional similarities, including embryonic origin, symbiotic microbial communities, innervation patterns and immune functions (19). As internal and external surfaces in contact with the environment, they utilize similar signaling and innervation pathways. T-cell-mediated immune responses often manifest in both intestinal and cutaneous tissues (25).
The gut microbial community is central to maintaining gut-skin homeostasis and underpins the gut-skin axis theory (26). The gut microbiota includes bacteria, fungi, parasites, protozoa and viruses, with bacteria predominating (27). Over 90% of gut bacteria belong to the Bacteroidetes and Firmicutes phyla (28). The Bacteroidota-to-Firmicutes ratio is commonly used to assess gut microbiota characteristics and diversity (29). Gut bacteria can be categorized as beneficial (e.g., Bifidobacteria, Lactobacillus) or opportunistic pathogens (e.g., Staphylococci, Clostridia), which may cause infection under certain conditions (30).
Alterations in gut microbiome composition, metabolism and immunity can impact skin health. Manos (31) identified interspecies communication within microbial communities as a key factor in maintaining cutaneous homeostasis and responding to environmental stressors, with dysregulation contributing to skin disease. External factors such as genetics, diet, antimicrobials, and lifestyle influence microbial diversity (32) (Fig. 2A).
Gut dysbiosis is associated with multiple disease states (33) (Fig. 1D). Olejniczak-Staruch et al (34) reported significant gut dysbiosis in patients with psoriasis, characterized by increased abundance of Campylobacter, Helicobacter, Escherichia coli, Alcaligenes and Salmonella. Similarly, infants with AD show reduced Bifidobacteria and increased Clostridium difficile and E. coli (35).
Gut microbiota-derived metabolites mediate skin interactions through short-chain fatty acids (SCFAs), tryptophan metabolites and amine derivatives (e.g., trimethylamine N-oxide), which exert systemic effects via specific receptors (36). SCFAs play important roles in immune regulation; Trompette et al (37) demonstrated that gut-derived butyrate enhances skin barrier function by modifying mitochondrial metabolism in keratinocytes. Fang et al (38) reported that Bifidobacterium longum produces indole derivatives that alleviate AD via the tryptophan pathway. Tryptophan metabolites (e.g., indoleacetic acid) are critical for maintaining intestinal and systemic immune homeostasis (39) (Fig. 2B).
The gut microbiota modulates both systemic and skin-specific immunity (16). Through arginine-induced barrier changes, it influences skin differentiation, immune activation and homeostatic balance (40) (Fig. 2B). Disrupted gut-immune crosstalk compromises skin barrier integrity, increasing susceptibility to inflammatory skin conditions. Marrs et al (41) associated a greater Clostridium abundance in infant guts with AD, suggesting that gut microbiota evolution during infancy affects immune tolerance development. Intestinal commensal bacteria prime innate immunity; dysbiosis may alter local immune responses via Toll-like receptor pathways or inflammasomes, promoting systemic inflammation (42).
Bidirectional interaction between the gut microbiome and skin health is central to the gut-skin axis theory. While most evidence highlights gut microbes influencing skin health, Dokoshi et al (43) demonstrated that skin injury directly remodels the gut microbiome (Fig. 2B), providing experimental support for bidirectional signaling and indicating that skin damage may impair gut immune homeostasis. Long et al (44), in a two-sample Mendelian randomization study, established a causal relationship between gut microbiota and four common inflammatory skin diseases: Eczema, acne, psoriasis and rosacea.
Understanding gut microbiota-skin interactions offers novel mechanistic insights for managing dermatological conditions. Clinical evidence reveals frequent comorbidity of skin and intestinal disorders, such as enteropathic acrodermatitis (zinc malabsorption causing dermatitis, alopecia and diarrhea) (45), and celiac disease (associated with eczema, psoriasis and urticaria) (46).
Restoring gut microbiota balance enhances intestinal barrier integrity and regulates immune responses (47). This involves upregulating immunomodulatory cytokines (e.g., IL-10) (48) and suppressing pro-inflammatory mediators (e.g., TNF-α) (49). Clinical interventions include probiotics, prebiotics, synbiotics and fecal microbiota transplantation (FMT) (50).
FMT involves transferring processed stool from healthy donors to patients to rebuild gut microbiota. Initially used for digestive diseases (51-53), FMT's efficacy depends on donor and recipient factors (immune status, genetic diversity, gut microbial composition) and treatment protocols (stool amount, number of infusions, delivery route and adjuvant therapy) (54) (Fig. 2C).
While most FMT research focuses on gastrointestinal disorders, recent trials suggest it may modulate systemic immune responses, including in skin cancers like melanoma. Kim et al (55) showed that FMT improved AD symptoms in mice by restoring gut microbiota. FMT enhances immunotherapy efficacy in cancer patients by modulating gut microbiota (56). Melanoma, the most common and prognostically worst skin cancer, is increasing globally (57). Baruch et al (58) observed that FMT induced favorable changes in immune cell infiltration and gene expression in intestinal and tumor microenvironments. Liu et al (59) demonstrated that FMT is effective for moderate to severe AD in adults, altering gut microbiota composition and function. However, FMT carries risks; Eshel et al (60) found that while FMT capsules rarely transmit bloodstream pathogens directly, they may indirectly promote bacterial translocation via gut inflammation, potentially compromising intestinal barrier integrity and increasing infection risk. Further in-depth studies are required to ascertain the efficacy and safety of FMT in treating other skin diseases.
Prebiotics are indigestible food components that selectively stimulate beneficial bacteria (e.g., Bifidobacterium, Lactobacillus) while inhibiting pathogenic overgrowth, thereby improving intestinal microecology (61). Probiotics are live microorganisms that, when administered in adequate amounts, confer health benefits through intestinal colonization (62). These interventions show promise beyond gastrointestinal and cardiovascular diseases (63,64) with emerging applications in dermatology.
Rahmayani et al (65) showed that oral probiotics increased serum IL-10 levels in patients with acne vulgaris (AV). IL-10 cytokines curtail excessive inflammation, enhance innate immunity and promote tissue repair, offering therapeutic potential for inflammatory diseases (66). Probiotic supplementation improves gut microbiota balance, inflammatory markers and quality of life in patients with psoriasis (67).
In oncology, gut microbiome modulation may enhance immunotherapy efficacy. Specific probiotics influence T-cell-mediated therapies (e.g., anti-programmed cell death 1, chimeric antigen receptor-T) (68), though strain-specific effects require further validation. Bender et al (69) demonstrated that Lactobacillus reuteri translocates to melanoma sites in mice, secreting indole-3-aldehyde to activate CD8+ T-cell receptors and potentiate immunotherapy. Such mechanisms hold potential for future immunomodulatory strategies pending resolution of delivery challenges and dose optimization.
Psoriasis is an immune-mediated skin disease characterized by abnormal keratinocyte proliferation and differentiation, posing significant physical and psychological burdens (70). Previous studies suggested gut microbiota dysbiosis in patients with psoriasis, but the relationship remained vague (71). Zang et al (72) used bidirectional Mendelian randomization to identify Pasteurellaceae, Brucella and Methanobrevibacter smithii as potential pathogenic contributors, suggesting microbial targets for therapy (Table I). Zhao et al (73) demonstrated through metabolomics that gut microbiota transplantation from severely affected mice exacerbated skin inflammation in mild cases, while a phosphodiesterase-4 inhibitor alleviated symptoms and restored the gut microbiota, providing a theoretical basis for gut microbiota-skin axis-based psoriasis treatment (Table I).
AD is a common chronic inflammatory skin disease characterized by persistent itching and eczematous rash (74). It increases the risk of comorbidities like food allergies, asthma, allergic rhinitis and mental health disorders (75). The pathogenesis involves hereditary immune dysregulation, skin barrier dysfunction and environmental factors (76). The gut microbiota modulates immune system development, implying a key role in AD (77).
First-line AD treatment includes topical corticosteroids and calcineurin inhibitors (e.g., pimecrolimus, tacrolimus). For moderate to severe cases, ultraviolet phototherapy is used adjunctively (78). Probiotic supplementation is increasingly used, particularly in children, to modulate gut microbiota and regulate immune responses (79,80). Meta-analyses indicate certain probiotics reduce Scoring Atopic Dermatitis indices in adults (81). Fang et al (82) showed that Bifidobacterium CCFM16 and Lactobacillus plantar CCFM8610 specifically improve AD by altering gut microbiota composition and function (Table I).
AV is a chronic inflammatory skin disease affecting sebaceous units, characterized by comedones, papules, pustules, nodules and scars, typically on the face, upper trunk and extremities (83). The global prevalence is estimated at 8.96% in men and 9.81% in women (84). The pathogenesis involves multifactorial mechanisms, with pubertal sebum hypersecretion being a key factor. AV often persists into adulthood with distinct features (85). Androgens and testosterone regulate sebum production, explaining why males often experience more severe symptoms (86).
Gut dysbiosis may exacerbate AV through upregulated insulin-like growth factor 1 signaling, insulin resistance and systemic pro-inflammatory cytokines (87). First-line treatments include topical retinoids, azelaic acid and benzoyl peroxide (88). The role of gut microbiota modulation via probiotics in improving AV is underexplored. Jung et al (89) found that probiotic-antibiotic combination therapy synergistically reduced inflammation and antibiotic side effects. Fabbrocini et al (90) showed that Lactobacillus rhamnosus SP1 (LSP1) probiotic normalized skin insulin signaling gene expression. Eguren et al (91) conducted a 12-week trial with Lactobacillus rhamnosus and Arthrospira in patients with AV aged 12-30 years, finding the probiotic adjunct safe and effective (Table I).
Rosacea is a chronic recurrent inflammatory skin condition affecting the central face (forehead, nose, cheeks, chin) (92). It is classified into erythematotelangiectatic, papulopustular, phymatous and ocular subtypes (93).
Gastrointestinal disorders are common comorbidities (94) and rosacea is linked to inflammatory bowel disease (95,96). While mechanistic studies are limited, microbiota modulation shows therapeutic potential. Manzhalii et al (97) showed that oral Escherichia coli Nissle 1917 increased beneficial bacteria (Bifidobacterium, Lactobacillus) and improved mucin barrier function, resolving rosacea symptoms. Fortuna et al (98) reported a case of scalp rosacea treated with doxycycline and probiotics (Bifidobacterium breve BR03, Lactobacillus salivarius LS01), with significant improvement. These findings support further investigation into gut microbiota-targeted rosacea management.
According to 2021 guidelines, urticaria is classified as acute (lasting <6 weeks) or chronic urticaria (CU). CU is a common inflammatory skin disorder involving mast cell-mediated allergy and autoimmunity, characterized by recurrent wheals, angioedema and itching lasting ≥6 weeks (99). Pathophysiology involves immune dysregulation, inflammatory cascade imbalance and coagulation-fibrinolytic activation (100). CU subtypes include chronic spontaneous urticaria (CSU) and chronic inducible urticarial (101).
Research links the gut microbiota to urticaria. Wang et al (102) found reduced Bacteroidetes and Proteobacteria in patients with CSU, with butyric acid metabolism alterations (Table I). Luo et al (103) observed decreased Bacteroidetes, Ruminococcus, Megasphaera and anaerobic bacilli in CSU, with altered serum metabolites (e.g., unsaturated fatty acids, purines), suggesting immune dysregulation (Table I). Few studies exist on gut microbiome-modulating therapies for urticaria. Fu et al (104) reported that probiotics reduced symptom scores (Table I). Bi et al (105) showed that probiotic mixtures were safe and effective adjuncts for childhood CU (Table I). These preliminary findings require larger validation studies.
Vitiligo is a depigmentation disorder affecting 0.5-2% globally, with a genetic component (106). It presents as white, scaleless patches (107). Pathophysiologically, vitiligo is an autoimmune condition where melanocytes are sensitive to oxidative stress, triggering inflammatory cytokine release and innate immune activation. CD8+ T cells destroy melanocytes, driven by IFN-γ. Oxidative stress may induce gut dysbiosis, promoting autoimmunity. IFN-γ blockade therapies temporarily reverse depigmentation but relapse occurs upon cessation (108). The chronic course of vitiligo affects patients' appearance and psychological well-being; Kussainova et al (109) reported a 35.8% anxiety prevalence, higher in women.
Current evidence on the gut-vitiligo link is limited. Emerging studies suggest gut microbiome involvement. Hadi et al (110) reported vitiligo-inflammatory bowel disease comorbidity. Ni et al (111) documented a Firmicutes/Bacteroidetes ratio reduction in patients with vitiligo (Table I), while Luan et al (112) observed decreased microbial alpha diversity with altered cysteine/galactose metabolism (Table I). These findings support the gut-microbiota-skin hypothesis but require further investigation for therapeutic applications.
Skin cancer is the fifth most common cancer globally, with its incidence rising (113). It arises from genetic defects or DNA mutations in skin cells, primarily due to ultraviolet exposure (114). Skin cancers are classified as malignant melanoma (MM) or non-melanoma skin cancer (NMSC). MM originates from melanocytes, while NMSC (e.g., squamous cell carcinoma, basal cell carcinoma) arises from epidermal cells (115). Treatments include cryotherapy, radiotherapy and photodynamic therapy, but novel approaches are needed (116).
MM is highly metastatic, has a poor prognosis and is the leading cause of death among patients with skin cancer (117). Mrazek et al (118) found compositional differences between melanoma and healthy skin microbiomes, with elevated Fusobacterium and Eubacterium in tumors (Table I). Mekadim et al (119) demonstrated distinct gut and skin microbiome profiles in melanoma models, addressing knowledge gaps (Table I).
Several studies confirmed that the gut microbiota can modulate the response to cancer immune checkpoint blockade therapy (120-122). Spencer et al (123) reported improved progression-free survival in ICB-treated patients with higher fiber intake, particularly without probiotics; fiber/prebiotics enhanced antitumor T-cell responses via microbiome modulation (Table I). Routy et al (124)'s phase I trial confirmed FMT safety combined with first-line melanoma therapy, though efficacy requires further validation (Fig. 2D) (Table I). Future investigations of the gut-skin axis may yield innovative therapeutic strategies for skin cancer.
Rare dermatoses like AIBD and LP receive limited research due to funding and recruitment challenges (125). AIBD involve chronic immune-mediated blistering, with fragile cutaneous/mucosal vesicles rupturing into erosions. These include pemphigus vulgaris (PV), bullous pemphigoid (BP) and mucous membrane pemphigoid (126). PV subtypes include vulgaris, foliaceus, IgA pemphigus and paraneoplastic pemphigus. BP, the most common pemphigoid, features tense, rupture-resistant subepidermal blisters, often involving the oropharyngeal and ocular mucosa. It primarily affects adults aged >50 years but can also occur in younger individuals (127).
Pemphigus has a long course and poor prognosis, severely affecting patients' quality of life, with most deaths due to uncontrollable secondary infections (128). The pathogenesis involves Th1/Th2 and Th17/Treg imbalances, leading to IgG autoantibodies that target epidermal/mucosal antigens, causing loss of cell adhesion and blister formation (129).
Huang et al (130) reported reduced Lachnospira/Coprococcus and elevated Aspergillus in PV fecal samples, with negative correlations between Lachnospira/Coprococcus and IL-17A suggesting T-cell modulation. Han et al (131) found a divergent gut microbiota in patients with PV vs. BP, implicating autoantibody and gut-barrier pathways.
LP is a chronic inflammatory disease presenting as violaceous pruritic papules on skin, mucosa or nails. Subtypes include cutaneous LP (CLP) and oral LP (OLP), with possible esophageal, genital or nail involvement (132). The prevalence of LP ranges from 0.22 to 1%, with OLP about five times more common than CLP (133). Genital subtypes have been reported (134), and concurrent subtypes complicate diagnosis (135). The pathogenesis of CLP involves cell-mediated immune responses against basal keratinocytes (136). Georgescu et al (137) reported an oxidant-antioxidant imbalance in patients with LP, suggesting oxidative stress plays a role.
Few studies have focused on the role of the 'gut microbe-skin axis' in rare skin diseases such as LP and pemphigus (22). Li et al (138) observed a distinct gut microbiota in patients with active pemphigus, with Prevotella spp. and Coriobacteriaceae abundance correlating with autoantibodies. Roy et al (139) reviewed intestinal microecological dysregulation in several rare diseases, including LP, and highlighted the complexity of host-microbiota interactions, emphasizing knowledge gaps, the need for improved study designs, and the promise of microbiome-based therapeutics. Kamal et al (140)'s trial on 60 patients with OLP showed a greater reduction in pain and Thongprasom scores with combined clobetasol/probiotic therapy vs. clobetasol alone. Despite research challenges, such as scarce pathogenic data and small cohorts, elucidating microbiota-skin interactions could yield novel diagnostics and therapies for rare dermatoses.
Despite progress, translating gut microbiome research into clinical applications remains challenging. Methodological limitations in data collection, representativeness and analysis are key barriers. Resources like GMrepo (https://gmrepo.humangut.info/home) and gutMGene (https://bio-computing.hrbmu.edu.cn/gutmgene/#/home) provide valuable data, but heterogeneity due to geographic, demographic and lifestyle factors complicates sampling and standardization (Fig. 2A). The lack of a consensus definition for 'healthy gut microbiota' impedes benchmark establishment (141,142).
Analytical techniques involve sequencing and data analysis (Fig. 2A). Sequencing and analysis methods face several limitations. High-throughput 16S rRNA sequencing offers taxonomic profiling but lacks strain-level resolution and poorly detects archaea/viruses. Whole-genome shotgun metagenomics enables functional insights but requires computational resources for high-dimensional data integration (143). Microbiome-wide association studies link microbial patterns to diseases like obesity and colorectal cancer (144), but cannot establish any causality due to confounders like medications and comorbidities (145). Integrative multi-omics approaches (e.g., metagenomics, metabolomics) and genome-wide association studies provide deeper insights (146,147) but require advanced normalization to address batch effects. Machine learning shows promise for modeling microbial interactions and predicting functional outputs from fragmented data (148). Ma et al (149) applied machine learning to identify diagnostic gut microbiome biomarkers for AD, enabling precision management.
FMT, probiotics and prebiotics present ethical and safety challenges. Al-Bakri et al (150) highlighted skepticism among Jordanian medical workers regarding FMT efficacy, influenced by cultural and religious factors. Patients expressed safety concerns about infection risks. Regulatory complexity arises from divergent global standards; the Food and Drug Administration (FDA) classifies FMT as an investigational drug requiring rigorous oversight. Two microbiota-derived therapeutics are FDA-approved: Rebyota® (RBX2660; Ferring Pharmaceuticals) and VOWST™ (SER-109; Seres Therapeutics) (151).
Patient and provider acceptance barriers persist, especially in culturally conservative settings. Probiotics, though generally safer than FMT, carry risks such as bacteremia in immunocompromised hosts and horizontal gene transfer (152,153). Cases like Lactobacillus rhamnosus infections in immunosuppressed individuals underscore the need for further safety research (154-156).
Preclinical gut microbiome research will continue diversifying. International data-sharing collaborations may overcome ethnographic limitations (157). Integrating medicine, biology and informatics could expand the research scope, as seen in machine learning applications for microbial community analysis (158). Current algorithms predict health status and identify disease-microbiome associations through differential abundance analysis (159).
In clinical translation, precision medicine requires refinement. Longitudinal studies with larger cohorts may better evaluate probiotic efficacy across populations, informing targeted microbiome-based interventions using adaptive trial designs (e.g., cluster randomized controlled trials). These frameworks would strengthen disease prevention and treatment strategies.
Emerging technologies like high-intensity ultrasound show potential for enhancing functional food development by improving probiotic stability and bioactivity (160), Future validation studies should assess gut health optimization and personalized approaches (161,162). Beyond dairy probiotics, fermented foods and beverages are demonstrating health benefits (163,164).
The following conclusions can be drawn from the present review: i) The gut-skin axis framework highlights the influence of gut microbiota on skin homeostasis, with dysbiosis implicated in psoriasis, AD, acne and AIBD; ii) the gut microbiota modulates skin health through immune regulation, microbial metabolite production (e.g., SCFAs) and systemic inflammation control. FMT, probiotics and prebiotics show promise in restoring microbial balance and reducing inflammation, though larger trials are needed; iii) current evidence primarily establishes correlations, but causal relationships require validation via multi-omics approaches integrating genomics, metabolomics and immune profiling; iv) clinical translation faces challenges including methodological limitations, ethical concerns with FMT and insufficient long-term safety data for probiotics; v) mechanistic insights into rare skin diseases via the gut-skin axis are scarce, warranting targeted investigations; vi) future research should prioritize patient-specific microbiome interventions, machine-learning diagnostics and cross-disciplinary collaboration (e.g., dermatology-gastroenterology-bioinformatics) to advance precision dermatology; and vii) the gut-skin axis redefines skincare paradigms, emphasizing gut health as integral to dermatological wellness, with breakthroughs in microbiome engineering and Artificial Intelligence-driven interventions offering transformative potential.
Not applicable.
YZ performed the analyses and wrote the first draft of the manuscript. CY, JZ, QY, XZ and XZ performed the literature search and discussed and edited the manuscript. XZ and XZ supervised the preparation of the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
This study was partially supported by the National Natural Science Foundation of China (grant nos. 32170915 and 82172931).
|
Karimzadeh F, Soltani Fard E, Nadi A, Malekzadeh R, Elahian F and Mirzaei SA: Advances in skin gene therapy: Utilizing innovative dressing scaffolds for wound healing, a comprehensive review. J Mater Chem B. 12:6033–6062. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Shi N, Li N, Duan X and Niu H: Interaction between the gut microbiome and mucosal immune system. Mil Med Res. 4:142017.PubMed/NCBI | |
|
Wang J, He M, Yang M and Ai X: Gut microbiota as a key regulator of intestinal mucosal immunity. Life Sci. 345:1226122024. View Article : Google Scholar : PubMed/NCBI | |
|
Ezenabor EH, Adeyemi AA and Adeyemi OS: Gut microbiota and metabolic syndrome: Relationships and opportunities for new therapeutic strategies. Scientifica (Cairo). 2024:42220832024. View Article : Google Scholar : PubMed/NCBI | |
|
Saarialho-Kere U: The gut-skin axis. J Pediatr Gastroenterol Nutr. 39(Suppl 3): S734–S735. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HR and Sung JH: Multiorgan-on-a-chip for the realization of gut-skin axis. Biotechnol Bioeng. 119:2590–2601. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Szanto M, Dozsa A, Antal D, Szabo K, Kemeny L and Bai P: Targeting the gut-skin axis-Probiotics as new tools for skin disorder management? Exp Dermatol. 28:1210–1218. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Suaini NHA, Siah KTH and Tham EH: Role of the gut-skin axis in IgE-mediated food allergy and atopic diseases. Curr Opin Gastroenterol. 37:557–564. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Alesa DI, Alshamrani HM, Alzahrani YA, Alamssi DN, Alzahrani NS and Almohammadi ME: The role of gut microbiome in the pathogenesis of psoriasis and the therapeutic effects of probiotics. J Family Med Prim Care. 8:3496–3503. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gomaa EZ: Human gut microbiota/microbiome in health and diseases: A review. Antonie Van Leeuwenhoek. 113:2019–2040. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Adak A and Khan MR: An insight into gut microbiota and its functionalities. Cell Mol Life Sci. 76:473–493. 2019. View Article : Google Scholar | |
|
Zmora N, Suez J and Elinav E: You are what you eat: Diet, health, and the gut microbiota. Nat Rev Gastroenterol Hepatol. 16:35–56. 2019. View Article : Google Scholar | |
|
Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, et al: The first microbial colonizers of the human gut: Composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev. 81:e00036–17. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Aggarwal N, Kitano S, Puah GRY, Kittelmann S, Hwang IY and Chang MW: Microbiome and human health: Current understanding, engineering, and enabling technologies. Chem Rev. 123:31–72. 2023. View Article : Google Scholar : | |
|
Banaszak M, Gorna I, Wozniak D, Przyslawski J and Drzymala-Czyz S: Association between gut dysbiosis and the occurrence of SIBO, LIBO, SIFO and IMO. Microorganisms. 11:5732023. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HJ and Kim M: Skin barrier function and the microbiome. Int J Mol Sci. 23:130712022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen YE, Fischbach MA and Belkaid Y: Skin microbiota-host interactions. Nature. 553:427–436. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Smythe P and Wilkinson HN: The skin microbiome: Current landscape and future opportunities. Int J Mol Sci. 24:39502023. View Article : Google Scholar : PubMed/NCBI | |
|
Mahmud MR, Akter S, Tamanna SK, Mazumder L, Esti IZ, Banerjee S, Akter S, Hasan MR, Acharjee M, Hossain MS and Pirttilä AM: Impact of gut microbiome on skin health: Gut-skin axis observed through the lenses of therapeutics and skin diseases. Gut Microbes. 14:20969952022. View Article : Google Scholar : PubMed/NCBI | |
|
Petersen C and Round JL: Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol. 16:1024–1033. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Rygula I, Pikiewicz W, Grabarek BO, Wojcik M and Kaminiow K: The role of the gut microbiome and microbial dysbiosis in common skin diseases. Int J Mol Sci. 25:19842024. View Article : Google Scholar : PubMed/NCBI | |
|
Karimova M, Moyes D, Ide M and Setterfield JF: The human microbiome in immunobullous disorders and lichen planus. Clin Exp Dermatol. 47:522–528. 2022. View Article : Google Scholar | |
|
Ahlawat S, Asha and Sharma KK: Gut-organ axis: A microbial outreach and networking. Lett Appl Microbiol. 72:636–668. 2021. View Article : Google Scholar | |
|
Guo Y, Chen X, Gong P, Li G, Yao W and Yang W: The gut-organ-axis concept: Advances the application of gut-on-chip technology. Int J Mol Sci. 24:40892023. View Article : Google Scholar : PubMed/NCBI | |
|
Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK and Hsiao EY: Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 161:264–276. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Salem I, Ramser A, Isham N and Ghannoum MA: The gut microbiome as a major regulator of the gut-skin axis. Front Microbiol. 9:14592018. View Article : Google Scholar : PubMed/NCBI | |
|
Komine M: Recent advances in psoriasis research; The clue to mysterious relation to gut microbiome. Int J Mol Sci. 21:25822020. View Article : Google Scholar : PubMed/NCBI | |
|
Buhas MC, Gavrilas LI, Candrea R, Catinean A, Mocan A, Miere D and Tătaru A: Gut microbiota in psoriasis. Nutrients. 14:29702022. View Article : Google Scholar : PubMed/NCBI | |
|
Frioux C, Ansorge R, Ozkurt E, Ghassemi Nedjad C, Fritscher J, Quince C, Waszak SM and Hildebrand F: Enterosignatures define common bacterial guilds in the human gut microbiome. Cell Host Microbe. 31:1111–1125 e6. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Quaglio AEV, Grillo TG, De Oliveira ECS, Di Stasi LC and Sassaki LY: Gut microbiota, inflammatory bowel disease, and colorectal cancer. World J Gastroenterol. 28:4053–4060. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Manos J: The human microbiome in disease and pathology. APMIS. 130:690–705. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Wang K, Wang X, Pang Y and Jiang C: The role of the gut microbiome and its metabolites in metabolic diseases. Protein Cell. 12:360–373. 2021. View Article : Google Scholar : | |
|
Chen Y, Zhou J and Wang L: Role and mechanism of gut microbiota in human disease. Front Cell Infect Microbiol. 11:6259132021. View Article : Google Scholar : PubMed/NCBI | |
|
Olejniczak-Staruch I, Ciazynska M, Sobolewska-Sztychny D, Narbutt J, Skibinska M and Lesiak A: Alterations of the skin and gut microbiome in psoriasis and psoriatic arthritis. Int J Mol Sci. 22:39982021. View Article : Google Scholar : PubMed/NCBI | |
|
Park DH, Kim JW, Park HJ and Hahm DH: Comparative analysis of the microbiome across the gut-skin axis in atopic dermatitis. Int J Mol Sci. 22:42282021. View Article : Google Scholar : PubMed/NCBI | |
|
Stec A, Sikora M, Maciejewska M, Paralusz-Stec K, Michalska M, Sikorska E and Rudnicka L: Bacterial metabolites: A link between gut microbiota and dermatological diseases. Int J Mol Sci. 24:34942023. View Article : Google Scholar : PubMed/NCBI | |
|
Trompette A, Pernot J, Perdijk O, Alqahtani RAA, Domingo JS, Camacho-Munoz D, Wong NC, Kendall AC, Wiederkehr A, Nicod LP, et al: Gut-derived short-chain fatty acids modulate skin barrier integrity by promoting keratinocyte metabolism and differentiation. Mucosal Immunol. 15:908–926. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Fang Z, Pan T, Li L, Wang H, Zhu J, Zhang H, Zhao J, Chen W and Lu W: Bifidobacterium longum mediated tryptophan metabolism to improve atopic dermatitis via the gut-skin axis. Gut Microbes. 14:20447232022. View Article : Google Scholar : PubMed/NCBI | |
|
Su X, Gao Y and Yang R: Gut microbiota-derived tryptophan metabolites maintain gut and systemic homeostasis. Cells. 11:22962022. View Article : Google Scholar : PubMed/NCBI | |
|
Kinashi Y and Hase K: Partners in leaky gut syndrome: intestinal dysbiosis and autoimmunity. Front Immunol. 12:6737082021. View Article : Google Scholar : PubMed/NCBI | |
|
Marrs T, Jo JH, Perkin MR, Rivett DW, Witney AA, Bruce KD, Logan K, Craven J, Radulovic S, Versteeg SA, et al: Gut microbiota development during infancy: Impact of introducing allergenic foods. J Allergy Clin Immunol. 147:613–621 e9. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiao Y, Wu L, Huntington ND and Zhang X: Crosstalk between gut microbiota and innate immunity and its implication in autoimmune diseases. Front Immunol. 11:2822020. View Article : Google Scholar : PubMed/NCBI | |
|
Dokoshi T, Chen Y, Cavagnero KJ, Rahman G, Hakim D, Brinton S, Schwarz H, Brown EA, O'Neill A, Nakamura Y, et al: Dermal injury drives a skin-to-gut axis that disrupts the intestinal microbiome and intestinal immune homeostasis in mice. Nat Commun. 15:30092024. View Article : Google Scholar | |
|
Long J, Gu J, Yang J, Chen P, Dai Y, Lin Y, Wu M and Wu Y: Exploring the association between gut microbiota and inflammatory skin diseases: A two-sample mendelian randomization analysis. Microorganisms. 11:25862023. View Article : Google Scholar : PubMed/NCBI | |
|
Glutsch V, Hamm H and Goebeler M: Zinc and skin: An update. J Dtsch Dermatol Ges. 17:589–596. 2019.PubMed/NCBI | |
|
Therrien A, Kelly CP and Silvester JA: Celiac disease: Extraintestinal manifestations and associated conditions. J Clin Gastroenterol. 54:8–21. 2020. View Article : Google Scholar | |
|
Ni Q, Zhang P, Li Q and Han Z: Oxidative stress and gut microbiome in inflammatory skin diseases. Front Cell Dev Biol. 10:8499852022. View Article : Google Scholar : PubMed/NCBI | |
|
Lee SY, Jhun J, Woo JS, Lee KH, Hwang SH, Moon J, Park G, Choi SS, Kim SJ, Jung YJ, et al: Gut microbiome-derived butyrate inhibits the immunosuppressive factors PD-L1 and IL-10 in tumor-associated macrophages in gastric cancer. Gut Microbes. 16:23008462024. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao P, Hu Z, Lang J, Pan T, Mertens RT, Zhang H, Guo K, Shen M, Cheng H, Zhang X, et al: Mannose metabolism normalizes gut homeostasis by blocking the TNF-α-mediated proinflammatory circuit. Cell Mol Immunol. 20:119–130. 2023. View Article : Google Scholar | |
|
Yadegar A, Bar-Yoseph H, Monaghan TM, Pakpour S, Severino A, Kuijper EJ, Smits WK, Terveer EM, Neupane S, Nabavi-Rad A, et al: Fecal microbiota transplantation: current challenges and future landscapes. Clin Microbiol Rev. 37:e00060222024. View Article : Google Scholar : PubMed/NCBI | |
|
Joachim A, Schwerd T, Holz H, Sokollik C, Konrad LA, Jordan A, Lanzersdorfer R, Schmidt-Choudhury A, Hünseler C and Adam R: Fecal Microbiota Transfer (FMT) in children and adolescents-review and statement by the GPGE microbiome working group. Z Gastroenterol. 60:963–969. 2022.PubMed/NCBI | |
|
Porcari S, Severino A, Rondinella D, Bibbo S, Quaranta G, Masucci L, Maida M, Scaldaferri F, Sanguinetti M, Gasbarrini A, et al: Fecal microbiota transplantation for recurrent Clostridioides difficile infection in patients with concurrent ulcerative colitis. J Autoimmun. 141:1030332023. View Article : Google Scholar : PubMed/NCBI | |
|
Porcari S, Baunwall SMD, Occhionero AS, Ingrosso MR, Ford AC, Hvas CL, Gasbarrini A, Cammarota G and Ianiro G: Fecal microbiota transplantation for recurrent C. difficile infection in patients with inflammatory bowel disease: A systematic review and meta-analysis. J Autoimmun. 141:1030362023. View Article : Google Scholar : PubMed/NCBI | |
|
Porcari S, Benech N, Valles-Colomer M, Segata N, Gasbarrini A, Cammarota G, Sokol H and Ianiro G: Key determinants of success in fecal microbiota transplantation: From microbiome to clinic. Cell Host Microbe. 31:712–733. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JH, Kim K and Kim W: Gut microbiota restoration through fecal microbiota transplantation: A new atopic dermatitis therapy. Exp Mol Med. 53:907–916. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu YH, Chen J, Chen X and Liu H: Factors of faecal microbiota transplantation applied to cancer management. J Drug Target. 32:101–114. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Arnold M, Singh D, Laversanne M, Vignat J, Vaccarella S, Meheus F, Cust AE, de Vries E, Whiteman DC and Bray F: Global burden of cutaneous melanoma in 2020 and projections to 2040. JAMA Dermatol. 158:495–503. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al: Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 371:602–609. 2021. View Article : Google Scholar | |
|
Liu X, Luo Y, Chen X, Wu M, Xu X, Tian J, Gao Y, Zhu J, Wang Z, Zhou Y, et al: Fecal microbiota transplantation against moderate-to-severe atopic dermatitis: A randomized, double-blind controlled exploratory trial. Allergy. 80:1377–1388. 2025. View Article : Google Scholar | |
|
Eshel A, Sharon I, Nagler A, Bomze D, Danylesko I, Fein JA, Geva M, Henig I, Shimoni A, Zuckerman T, et al: Origins of bloodstream infections following fecal microbiota transplantation: A strain-level analysis. Blood Adv. 6:568–573. 2022. View Article : Google Scholar : | |
|
Gibson GR and Roberfroid MB: Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics. J Nutr. 125:1401–1412. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani RB, Flint HJ, Salminen S, et al: Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 11:506–514. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Manzoor S, Wani SM, Ahmad Mir S and Rizwan D: Role of probiotics and prebiotics in mitigation of different diseases. Nutrition. 96:1116022022. View Article : Google Scholar : PubMed/NCBI | |
|
Oniszczuk A, Oniszczuk T, Gancarz M and Szymanska J: Role of gut microbiota, probiotics and prebiotics in the cardiovascular diseases. Molecules. 26:11722021. View Article : Google Scholar : PubMed/NCBI | |
|
Rahmayani T, Putra IB and Jusuf NK: The effect of oral probiotics on the interleukin-10 serum levels of acne vulgaris. Open Access Maced J Med Sci. 7:3249–5322. 2019. View Article : Google Scholar | |
|
Ouyang W and O'Garra A: IL-10 family cytokines IL-10 and IL-22: From basic science to clinical translation. Immunity. 50:871–891. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Buhas MC, Candrea R, Gavrilas LI, Miere D, Tataru A, Boca A and Cătinean A: Transforming psoriasis care: Probiotics and prebiotics as novel therapeutic approaches. Int J Mol Sci. 24:112252023. View Article : Google Scholar : PubMed/NCBI | |
|
Stein-Thoeringer CK, Saini NY, Zamir E, Blumenberg V, Schubert ML, Mor U, Fante MA, Schmidt S, Hayase E, Hayase T, et al: A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat Med. 29:906–916. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bender MJ, McPherson AC, Phelps CM, Pandey SP, Laughlin CR, Shapira JH, Medina Sanchez L, Rana M, Richie TG, Mims TS, et al: Dietary tryptophan metabolite released by intratumoral Lactobacillus reuteri facilitates immune checkpoint inhibitor treatment. Cell. 186:1846–1862 e26. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Raharja A, Mahil SK and Barker JN: Psoriasis: A brief overview. Clin Med (Lond). 21:170–173. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hidalgo-Cantabrana C, Gomez J, Delgado S, Requena-Lopez S, Queiro-Silva R, Margolles A, Coto E, Sánchez B and Coto-Segura P: Gut microbiota dysbiosis in a cohort of patients with psoriasis. Br J Dermatol. 181:1287–1295. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zang C, Liu J, Mao M, Zhu W, Chen W and Wei B: Causal associations between gut microbiota and psoriasis: A mendelian randomization study. Dermatol Ther (Heidelb). 13:2331–2343. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Q, Yu J, Zhou H, Wang X, Zhang C, Hu J, Hu Y, Zheng H, Zeng F, Yue C, et al: Intestinal dysbiosis exacerbates the pathogenesis of psoriasis-like phenotype through changes in fatty acid metabolism. Signal Transduct Target Ther. 8:402023. View Article : Google Scholar : PubMed/NCBI | |
|
Sroka-Tomaszewska J and Trzeciak M: Molecular mechanisms of atopic dermatitis pathogenesis. Int J Mol Sci. 22:41302021. View Article : Google Scholar : PubMed/NCBI | |
|
Langan SM, Irvine AD and Weidinger S: Atopic dermatitis. Lancet. 396:345–360. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Schuler CF IV, Tsoi LC, Billi AC, Harms PW, Weidinger S and Gudjonsson JE: Genetic and immunological pathogenesis of atopic dermatitis. J Invest Dermatol. 144:954–968. 2024. View Article : Google Scholar : | |
|
Lee SY, Lee E, Park YM and Hong SJ: Microbiome in the gut-skin axis in atopic dermatitis. Allergy Asthma Immunol Res. 10:354–362. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Frazier W and Bhardwaj N: Atopic dermatitis: Diagnosis and treatment. Am Fam Physician. 101:590–598. 2020.PubMed/NCBI | |
|
Jiang W, Ni B, Liu Z, Liu X, Xie W, Wu IXY and Li X: The role of probiotics in the prevention and treatment of atopic dermatitis in children: An updated systematic review and meta-analysis of randomized controlled trials. Paediatr Drugs. 22:535–549. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
D'Elios S, Trambusti I, Verduci E, Ferrante G, Rosati S, Marseglia GL, Drago L and Peroni DG: Probiotics in the prevention and treatment of atopic dermatitis. Pediatr Allergy Immunol. 31(Suppl 26): S43–S45. 2020. View Article : Google Scholar | |
|
Umborowati MA, Damayanti D, Anggraeni S, Endaryanto A, Surono IS, Effendy I and Prakoeswa CRS: The role of probiotics in the treatment of adult atopic dermatitis: A meta-analysis of randomized controlled trials. J Health Popul Nutr. 41:372022. View Article : Google Scholar : PubMed/NCBI | |
|
Fang Z, Lu W, Zhao J, Zhang H, Qian L, Wang Q and Chen W: Probiotics modulate the gut microbiota composition and immune responses in patients with atopic dermatitis: A pilot study. Eur J Nutr. 59:2119–2130. 2020. View Article : Google Scholar | |
|
Sanchez-Pellicer P, Navarro-Moratalla L, Nunez-Delegido E, Ruzafa-Costas B, Aguera-Santos J and Navarro-Lopez V: Acne, microbiome, and probiotics: The gut-skin axis. Microorganisms. 10:13032022. View Article : Google Scholar : PubMed/NCBI | |
|
Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, Shibuya K, Salomon JA, Abdalla S, Aboyans V, et al: Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries, 1990-2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet. 380:2163–2196. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Kutlu O, Karadag AS and Wollina U: Adult acne versus adolescent acne: A narrative review with a focus on epidemiology to treatment. An Bras Dermatol. 98:75–83. 2023. View Article : Google Scholar : | |
|
Chilicka K, Rogowska AM, Szygula R, Dziendziora-Urbinska I and Taradaj J: A comparison of the effectiveness of azelaic and pyruvic acid peels in the treatment of female adult acne: A randomized controlled trial. Sci Rep. 10:126122020. View Article : Google Scholar : PubMed/NCBI | |
|
Bowe W, Patel NB and Logan AC: Acne vulgaris, probiotics, and the gut-brain-skin axis: From anecdote to translational medicine. Beneficial Microbes. 5:185–199. 2014. View Article : Google Scholar | |
|
Mohsin N, Hernandez LE, Martin MR, Does AV and Nouri K: Acne treatment review and future perspectives. Dermatol Ther. 35:e157192022. View Article : Google Scholar : PubMed/NCBI | |
|
Jung GW, Tse JE, Guiha I and Rao J: Prospective, randomized, open-label trial comparing the safety, efficacy, and tolerability of an acne treatment regimen with and without a probiotic supplement and minocycline in subjects with mild to moderate acne. J Cutan Med Surg. 17:114–122. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Fabbrocini G, Bertona M, Picazo O, Pareja-Galeano H, Monfrecola G and Emanuele E: Supplementation with Lactobacillus rhamnosus SP1 normalises skin expression of genes implicated in insulin signalling and improves adult acne. Benef Microbes. 7:625–630. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Eguren C, Navarro-Blasco A, Corral-Forteza M, Reolid-Perez A, Seto-Torrent N, Garcia-Navarro A, Prieto-Merino D, Núñez-Delegido E, Sánchez-Pellicer P and Navarro-López V: A randomized clinical trial to evaluate the efficacy of an oral probiotic in acne vulgaris. Acta Derm Venereol. 104:adv332062024. View Article : Google Scholar : PubMed/NCBI | |
|
van Zuuren EJ, Arents BWM, van der Linden MMD, Vermeulen S, Fedorowicz Z and Tan J: Rosacea: New concepts in classification and treatment. Am J Clin Dermatol. 22:457–465. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ivanic MG, Oulee A, Norden A, Javadi SS, Gold MH and Wu JJ: Neurogenic rosacea treatment: A literature review. J Drugs Dermatol. 22:566–575. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Haber R and El Gemayel M: Comorbidities in rosacea: A systematic review and update. J Am Acad Dermatol. 78:786–792 e8. 2018. View Article : Google Scholar | |
|
Jun YK, Yu DA, Han YM, Lee SR, Koh SJ and Park H: The relationship between rosacea and inflammatory bowel disease: A systematic review and meta-analysis. Dermatol Ther (Heidelb). 13:1465–1475. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, He SX, He YX, Hu XH and Zhou Z: Detecting potential causal relationship between inflammatory bowel disease and rosacea using bi-directional Mendelian randomization. Sci Rep. 13:149102023. View Article : Google Scholar : PubMed/NCBI | |
|
Manzhalii E, Hornuss D and Stremmel W: Intestinal-borne dermatoses significantly improved by oral application of Escherichia coli Nissle 1917. World J Gastroenterol. 22:5415–5421. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Fortuna MC, Garelli V, Pranteda G, Romaniello F, Cardone M, Carlesimo M and Rossi A: A case of scalp rosacea treated with low-dose doxycycline and probiotic therapy and literature review on therapeutic options. Dermatol Ther. 29:249–251. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zuberbier T, Abdul Latiff AH, Abuzakouk M, Aquilina S, Asero R, Baker D, Ballmer-Weber B, Bangert C, Ben-Shoshan M, Bernstein JA, et al: The international EAACI/GA(2)LEN/EuroGuiDerm/APAAACI guideline for the definition, classification, diagnosis, and management of urticaria. Allergy. 77:734–766. 2022. View Article : Google Scholar | |
|
Kaplan A, Lebwohl M, Gimenez-Arnau AM, Hide M, Armstrong AW and Maurer M: Chronic spontaneous urticaria: Focus on pathophysiology to unlock treatment advances. Allergy. 78:389–401. 2023. View Article : Google Scholar | |
|
Kolkhir P, Gimenez-Arnau AM, Kulthanan K, Peter J, Metz M and Maurer M: Urticaria. Nat Rev Dis Primers. 8:612022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang D, Guo S, He H, Gong L and Cui H: Gut microbiome and serum metabolome analyses identify unsaturated fatty acids and butanoate metabolism induced by gut microbiota in patients with chronic spontaneous urticaria. Front Cell Infect Microbiol. 10:242020. View Article : Google Scholar : PubMed/NCBI | |
|
Luo Z, Jin Z, Tao X, Wang T, Wei P, Zhu C and Wang Z: Combined microbiome and metabolome analysis of gut microbiota and metabolite interactions in chronic spontaneous urticaria. Front Cell Infect Microbiol. 12:10947372023. View Article : Google Scholar : PubMed/NCBI | |
|
Fu HY, Yu HD, Bai YP, Yue LF, Wang HM and Li LL: Effect and safety of probiotics for treating urticaria: A systematic review and meta-analysis. J Cosmet Dermatol. 22:2663–2670. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bi XD, Lu BZ, Pan XX, Liu S and Wang JY: Adjunct therapy with probiotics for chronic urticaria in children: Randomised placebo-controlled trial. Allergy Asthma Clin Immunol. 17:392021. View Article : Google Scholar : PubMed/NCBI | |
|
Spritz RA and Santorico SA: The genetic basis of vitiligo. J Invest Dermatol. 141:265–273. 2021. View Article : Google Scholar | |
|
Bergqvist C and Ezzedine K: Vitiligo: A review. Dermatology. 236:571–592. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Frisoli ML, Essien K and Harris JE: Vitiligo: Mechanisms of pathogenesis and treatment. Annu Rev Immunol. 38:621–648. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kussainova A, Kassym L, Akhmetova A, Glushkova N, Sabirov U, Adilgozhina S, Tuleutayeva R and Semenova Y: Vitiligo and anxiety: A systematic review and meta-analysis. PLoS One. 15:e02414452020. View Article : Google Scholar : PubMed/NCBI | |
|
Hadi A, Wang JF, Uppal P, Penn LA and Elbuluk N: Comorbid diseases of vitiligo: A 10-year cross-sectional retrospective study of an urban US population. J Am Acad Dermatol. 82:628–633. 2020. View Article : Google Scholar | |
|
Ni Q, Ye Z, Wang Y, Chen J, Zhang W, Ma C, Li K, Liu Y, Liu L, Han Z, et al: Gut microbial dysbiosis and plasma metabolic profile in individuals with vitiligo. Front Microbiol. 11:5922482020. View Article : Google Scholar : | |
|
Luan M, Niu M, Yang P, Han D, Zhang Y, Li W, He Q, Zhao Y, Mao B, Chen J, et al: Metagenomic sequencing reveals altered gut microbial compositions and gene functions in patients with non-segmental vitiligo. BMC Microbiol. 23:2652023. View Article : Google Scholar : PubMed/NCBI | |
|
Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023.PubMed/NCBI | |
|
Arivazhagan N, Mukunthan MA, Sundaranarayana D, Shankar A, Vinoth Kumar S, Kesavan R, Chandrasekaran S, Shyamala Devi M, Maithili K, Barakkath Nisha U and Abebe TG: Analysis of skin cancer and patient healthcare using data mining techniques. Comput Intell Neurosci. 2022:22502752022. View Article : Google Scholar : PubMed/NCBI | |
|
Hasan N, Nadaf A, Imran M, Jiba U, Sheikh A, Almalki WH, Almujri SS, Mohammed YH, Kesharwani P and Ahmad FJ: Skin cancer: Understanding the journey of transformation from conventional to advanced treatment approaches. Mol Cancer. 22:1682023. View Article : Google Scholar : PubMed/NCBI | |
|
Jindal M, Kaur M, Nagpal M, Singh M, Aggarwal G and Dhingra GA: Skin cancer management: Current scenario and future perspectives. Curr Drug Saf. 18:143–158. 2023. View Article : Google Scholar | |
|
Abbas O, Miller DD and Bhawan J: Cutaneous malignant melanoma: Update on diagnostic and prognostic biomarkers. Am J Dermatopathol. 36:363–379. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Mrazek J, Mekadim C, Kucerova P, Svejstil R, Salmonova H, Vlasakova J, Tarasová R, Čížková J and Červinková M: Melanoma-related changes in skin microbiome. Folia Microbiol (Praha). 64:435–442. 2019. View Article : Google Scholar | |
|
Mekadim C, Skalnikova HK, Cizkova J, Cizkova V, Palanova A, Horak V and Mrazek J: Dysbiosis of skin microbiome and gut microbiome in melanoma progression. BMC Microbiol. 22:632022. View Article : Google Scholar : PubMed/NCBI | |
|
Jia D, Wang Q, Qi Y, Jiang Y, He J, Lin Y, Sun Y, Xu J, Chen W, Fan L, et al: Microbial metabolite enhances immunotherapy efficacy by modulating T cell stemness in pan-cancer. Cell. 187:1651–1665 e21. 2024. View Article : Google Scholar | |
|
Zhou CB, Zhou YL and Fang JY: Gut microbiota in cancer immune response and immunotherapy. Trends Cancer. 7:647–660. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Andrews MC, Duong CPM, Gopalakrishnan V, Iebba V, Chen WS, Derosa L, Khan MAW, Cogdill AP, White MG, Wong MC, et al: Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat Med. 27:1432–1441. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Spencer CN, McQuade JL, Gopalakrishnan V, McCulloch JA, Vetizou M, Cogdill AP, Khan MAW, Zhang X, White MG, Peterson CB, et al: Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science. 374:1632–1640. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Routy B, Lenehan JG, Miller WH Jr, Jamal R, Messaoudene M, Daisley BA, Hes C, Al KF, Martinez-Gili L, Punčochář M, et al: Author correction: Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: A phase I trial. Nat Med. 30:6042024. View Article : Google Scholar | |
|
Ingram J: Editor's choice: Rare skin diseases themed issue. Br J Dermatol. 182:ix2020. View Article : Google Scholar : PubMed/NCBI | |
|
Buonavoglia A, Leone P, Dammacco R, Di Lernia G, Petruzzi M, Bonamonte D, Vacca A, Racanelli V and Dammacco F: Pemphigus and mucous membrane pemphigoid: An update from diagnosis to therapy. Autoimmun Rev. 18:349–358. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Malik AM, Tupchong S, Huang S, Are A, Hsu S and Otaparthi K: An updated review of pemphigus diseases. Medicina (Kaunas). 57:10802021. View Article : Google Scholar : PubMed/NCBI | |
|
Kridin K: Pemphigus group: overview, epidemiology, mortality, and comorbidities. Immunol Res. 66:255–270. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yamagami J: B-cell targeted therapy of pemphigus. J Dermatol. 50:124–131. 2023. View Article : Google Scholar | |
|
Huang S, Mao J, Zhou L, Xiong X and Deng Y: The imbalance of gut microbiota and its correlation with plasma inflammatory cytokines in pemphigus vulgaris patients. Scand J Immunol. 90:e127992019. View Article : Google Scholar : PubMed/NCBI | |
|
Han Z, Fan Y, Wu Q, Guo F, Li S, Hu X and Zuo YG: Comparison of gut microbiota dysbiosis between pemphigus vulgaris and bullous pemphigoid. Int Immunopharmacol. 128:1114702024. View Article : Google Scholar : PubMed/NCBI | |
|
Ioannides D, Vakirlis E, Kemeny L, Marinovic B, Massone C, Murphy R, Nast A, Ronnevig J, Ruzicka T, Cooper SM, et al: European S1 guidelines on the management of lichen planus: A cooperation of the European Dermatology Forum with the European academy of dermatology and venereology. J Eur Acad Dermatol Venereol. 34:1403–1414. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen K, Qin Y, Yan L, Dong Y, Lv S, Xu J, Kang N, Luo Z, Liu Y, Pu J, et al: Variations in salivary microbiota and metabolic phenotype related to oral lichen planus with psychiatric symptoms. BMC Oral Health. 25:9932025. View Article : Google Scholar : PubMed/NCBI | |
|
Jacques L, Kornik R, Bennett DD and Eschenbach DA: Diagnosis and management of vulvovaginal lichen planus. Obstet Gynecol Surv. 75:624–635. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
van Hees CLM and van der Meij EH: Lichen planus. Ned Tijdschr Tandheelkd. 130:221–226. 2023.In Dutch. View Article : Google Scholar : PubMed/NCBI | |
|
Husein-ElAhmed H, Gieler U and Steinhoff M: Lichen planus: A comprehensive evidence-based analysis of medical treatment. J Eur Acad Dermatol Venereol. 33:1847–1862. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Georgescu SR, Mitran CI, Mitran MI, Nicolae I, Matei C, Ene CD, Popa GL and Tampa M: Oxidative stress in cutaneous lichen planus narrative review. J Clin Med. 10:26922021. View Article : Google Scholar | |
|
Li SZ, Wu QY, Fan Y, Guo F, Hu XM and Zuo YG: Gut microbiome dysbiosis in patients with pemphigus and correlation with pathogenic autoantibodies. Biomolecules. 14:8802024. View Article : Google Scholar : PubMed/NCBI | |
|
Roy S, Nag S, Saini A and Choudhury L: Association of human gut microbiota with rare diseases: A close peek through. Intractable Rare Dis Res. 11:52–62. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kamal Y, Abdelwhab A, Salem ST and Fakhr M: Evaluation of the efficacy of supplementary probiotic capsules with topical clobetasol propionate 0.05% versus topical clobetasol propionate 0.05% in the treatment of oral lichen planus (a randomized clinical trial). BMC Oral Health. 25:3442025. View Article : Google Scholar : PubMed/NCBI | |
|
Shanahan F, Ghosh TS and O'Toole PW: The healthy microbiome-what is the definition of a healthy gut microbiome? Gastroenterology. 160:483–494. 2021. View Article : Google Scholar | |
|
Shalon D, Culver RN, Grembi JA, Folz J, Treit PV, Shi H, Rosenberger FA, Dethlefsen L, Meng X, Yaffe E, et al: Profiling the human intestinal environment under physiological conditions. Nature. 617:581–591. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Schmidt TSB, Raes J and Bork P: The human gut microbiome: From association to modulation. Cell. 172:1198–1215. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
McCallum G and Tropini C: The gut microbiota and its biogeography. Nat Rev Microbiol. 22:105–118. 2024. View Article : Google Scholar | |
|
Wang J and Jia H: Metagenome-wide association studies: fine-mining the microbiome. Nat Rev Microbiol. 14:508–522. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Uitterlinden AG: An introduction to genome-wide association studies: GWAS for dummies. Semin Reprod Med. 34:196–204. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Li L, Yang S, Li R, Su J, Zhou X, Zhu X and Gao R: Unraveling shared and unique genetic causal relationship between gut microbiota and four types of uterine-related diseases: Bidirectional mendelian inheritance approaches to dissect the 'gut-uterus axis'. Ann Epidemiol. 100:16–26. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Asnicar F, Thomas AM, Passerini A, Waldron L and Segata N: Machine learning for microbiologists. Nat Rev Microbiol. 22:191–205. 2024. View Article : Google Scholar | |
|
Ma J, Fang Y, Li S, Zeng L, Chen S, Li Z, Ji G, Yang X and Wu W: Interpretable machine learning algorithms reveal gut microbiome features associated with atopic dermatitis. Front Immunol. 16:15280462025. View Article : Google Scholar : PubMed/NCBI | |
|
Al-Bakri AG, Akour AA and Al-Delaimy WK: Knowledge, attitudes, ethical and social perspectives towards fecal microbiota transplantation (FMT) among Jordanian healthcare providers. BMC Med Ethics. 22:192021. View Article : Google Scholar | |
|
Benech N, Barbut F, Fitzpatrick F, Krutova M, Davies K, Druart C, Cordaillat-Simmons M, Heritage J, Guery B and Kuijper E; ESGCD and ESGHAMI: Update on microbiota-derived therapies for recurrent Clostridioides difficile infections. Clin Microbiol Infect. 30:462–468. 2024. View Article : Google Scholar | |
|
Sada RM, Matsuo H, Motooka D, Kutsuna S, Hamaguchi S, Yamamoto G and Ueda A: Clostridium butyricum Bacteremia associated with probiotic use, Japan. Emerg Infect Dis. 30:665–671. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zawistowska-Rojek A and Tyski S: Are probiotics safe for humans? Pol J Microbiol. 67:251–258. 2018. View Article : Google Scholar | |
|
Gouriet F, Million M, Henri M, Fournier PE and Raoult D: Lactobacillus rhamnosus bacteremia: An emerging clinical entity. Eur J Clin Microbiol Infect Dis. 31:2469–2480. 2021. View Article : Google Scholar | |
|
Harty DW, Oakey HJ, Patrikakis M, Hume EB and Knox KW: Pathogenic potential of lactobacilli. Int J Food Microbiol. 24:179–189. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Bartalesi F, Veloci S, Baragli F, Mantengoli E, Guidi S, Bartolesi AM, Mannino R, Pecile P and Bartoloni A: Successful tigecycline lock therapy in a Lactobacillus rhamnosus catheter-related bloodstream infection. Infection. 40:331–334. 2012. View Article : Google Scholar | |
|
Jin DM, Morton JT and Bonneau R: Meta-analysis of the human gut microbiome uncovers shared and distinct microbial signatures between diseases. mSystems. 9:e00295242024. View Article : Google Scholar : PubMed/NCBI | |
|
Camacho DM, Collins KM, Powers RK, Costello JC and Collins JJ: Next-generation machine learning for biological networks. Cell. 173:1581–1592. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Su Q, Liu Q, Lau RI, Zhang J, Xu Z, Yeoh YK, Leung TWH, Tang W, Zhang L, Liang JQY, et al: Faecal microbiome-based machine learning for multi-class disease diagnosis. Nat Commun. 13:68182022. View Article : Google Scholar : PubMed/NCBI | |
|
Chuang YF, Fan KC, Su YY, Wu MF, Chiu YL, Liu YC and Lin CC: Precision probiotics supplement strategy in an aging population based on gut microbiome composition. Brief Bioinform. 25:bbae3512024. View Article : Google Scholar | |
|
Guimaraes JT, Balthazar CF, Scudino H, Pimentel TC, Esmerino EA, Ashokkumar M, Freitas MQ and Cruz AG: High-intensity ultrasound: A novel technology for the development of probiotic and prebiotic dairy products. Ultrason Sonochem. 57:12–21. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Balthazar CF, Guimaraes JF, Coutinho NM, Pimentel TC, Ranadheera CS, Santillo A, Albenzio M, Cruz AG and Sant'Ana AS: The future of functional food: Emerging technologies application on prebiotics, probiotics, and postbiotics. Compr Rev Food Sci Food Saf. 21:2560–2586. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kucukgoz K and Trzaskowska M: Nondairy probiotic products: Functional foods that require more attention. Nutrients. 14:7532022. View Article : Google Scholar : PubMed/NCBI | |
|
Shumye Gebre T, Admassu Emire S, Okomo Aloo S, Chelliah R, Vijayalakshmi S and Hwan Oh D: Unveiling the potential of African fermented cereal-based beverages: Probiotics, functional drinks, health benefits and bioactive components. Food Res Int. 191:1146562024. View Article : Google Scholar : PubMed/NCBI |