loạn khuẩn
loạn khuẩn
1 Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Athens,
Greece; kreouzi.m@live.unic.ac.cy
2 NT-CardioMetabolics, Clinic for Metabolism and Athletic Performance, 47 Tirteou Str., 17564 Palaio Faliro,
Greece; n.theodorakis@flemig-hospital.gr
3 Department of Cardiology & Preventive Cardiology Outpatient Clinic, Amalia Fleming General Hospital,
14, 25th Martiou Str., 15127 Melissia, Greece; m.nikolaou@flemig-hospital.gr
4 School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
5 School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece
6 Biomedical Engineering Laboratory, National Technical University of Athens, 15780 Athens, Greece;
aanastasiou@biomed.ntua.gr
7 Department of Informatics & Telematics, Harokopio University of Athens, 17676 Kallithea, Greece;
kkalodanis@hua.gr
8 Intensive Care Unit, Sismanogleio General Hospital, 37 Sismanogleiou Str., 15126 Marousi, Greece
* Correspondence: sakagianni@sismanoglio.gr
Abstract: Metabolic disorders, including type 2 diabetes mellitus (T2DM), obesity, and
metabolic syndrome, are systemic conditions that profoundly impact the skin microbiota, a
dynamic community of bacteria, fungi, viruses, and mites essential for cutaneous health.
Dysbiosis caused by metabolic dysfunction contributes to skin barrier disruption, immune
dysregulation, and increased susceptibility to inflammatory skin diseases, including pso-
riasis, atopic dermatitis, and acne. For instance, hyperglycemia in T2DM leads to the
formation of advanced glycation end products (AGEs), which bind to the receptor for AGEs
(RAGE) on keratinocytes and immune cells, promoting oxidative stress and inflamma-
tion while facilitating Staphylococcus aureus colonization in atopic dermatitis. Similarly,
Academic Editor: Denis Roy obesity-induced dysregulation of sebaceous lipid composition increases saturated fatty
Received: 24 December 2024 acids, favoring pathogenic strains of Cutibacterium acnes, which produce inflammatory
Revised: 5 January 2025 metabolites that exacerbate acne. Advances in metabolomics and microbiome sequencing
Accepted: 14 January 2025
have unveiled critical biomarkers, such as short-chain fatty acids and microbial signatures,
Published: 14 January 2025
predictive of therapeutic outcomes. For example, elevated butyrate levels in psoriasis
Citation: Kreouzi, M.; Theodorakis,
have been associated with reduced Th17-mediated inflammation, while the presence of
N.; Nikolaou, M.; Feretzakis, G.;
specific Lactobacillus strains has shown potential to modulate immune tolerance in atopic
Anastasiou, A.; Kalodanis, K.;
Sakagianni, A. Skin Microbiota:
dermatitis. Furthermore, machine learning models are increasingly used to integrate multi-
Mediator of Interactions Between omics data, enabling personalized interventions. Emerging therapies, such as probiotics
Metabolic Disorders and Cutaneous and postbiotics, aim to restore microbial diversity, while phage therapy selectively targets
Health and Disease. Microorganisms pathogenic bacteria like Staphylococcus aureus without disrupting beneficial flora. Clinical
2025, 13, 161. https://doi.org/
trials have demonstrated significant reductions in inflammatory lesions and improved
10.3390/microorganisms13010161
quality-of-life metrics in patients receiving these microbiota-targeted treatments. This
Copyright: © 2025 by the authors. review synthesizes current evidence on the bidirectional interplay between metabolic dis-
Licensee MDPI, Basel, Switzerland.
orders and skin microbiota, highlighting therapeutic implications and future directions. By
This article is an open access article
addressing systemic metabolic dysfunction and microbiota-mediated pathways, precision
distributed under the terms and
conditions of the Creative Commons
strategies are paving the way for improved patient outcomes in dermatologic care.
Attribution (CC BY) license
(https://creativecommons.org/
licenses/by/4.0/).
1. Introduction
Metabolic disorders, including type 2 diabetes mellitus (T2DM), obesity, and metabolic
syndrome (MetS), pose significant global health challenges due to their systemic effects and
numerous comorbidities [1]. These conditions, characterized by disrupted glucose and lipid
metabolism, insulin resistance, and chronic low-grade inflammation (meta-inflammation),
have traditionally been linked to cardiovascular–renal–hepatic diseases [2]. However,
emerging evidence highlights their far-reaching impact on the skin, particularly through
interactions with the skin microbiota, a dynamic ecosystem critical for cutaneous health [3].
The skin microbiota, comprising bacteria (e.g., Staphylococcus epidermidis, Cutibac-
terium acnes), fungi (e.g., Malassezia spp.), viruses, and mites, is essential for cutaneous
health [4]. This microbial community plays a pivotal role in modulating immune re-
sponses, maintaining epidermal barrier integrity, and competing with pathogenic organ-
isms. Metabolic dysregulation alters these interactions at molecular and cellular levels. For
instance, hyperglycemia-driven advanced glycation end products (AGEs) compromise skin
barrier proteins, while imbalances in adipokines and cytokines in obesity shift immune
responses and modify epidermal lipid composition, reshaping microbial communities [5,6].
These changes exacerbate susceptibility to dermatologic conditions such as psoriasis, atopic
dermatitis (AD), and chronic wounds [7–9].
Additionally, with the rise of integrative ‘omics’ and large-scale clinical data, machine
learning (ML) approaches are increasingly being employed to identify complex patterns,
predict disease risk, and evaluate therapeutic responses. Such computational tools can
integrate data streams from metabolic profiling, microbiome sequencing, and clinical
outcomes, helping to unravel the intricate interplay between metabolic disorders, the skin
microbiome, and dermatologic health [10,11].
This review explores the bidirectional interactions between metabolic dysfunction and
the skin microbiota, focusing on their roles in barrier disruption, immune dysregulation,
and dermatologic conditions such as psoriasis, AD, and acne. We highlight insights from
omics technologies, including metabolomics and microbiomics, which have identified novel
biomarkers predictive of disease risk and therapeutic outcomes. Emerging therapies, such
as probiotics, postbiotics, and phage therapy, are examined for their potential to correct
microbial dysbiosis while addressing underlying metabolic abnormalities. Advances in
ML further enhance our ability to integrate complex multi-omics data, driving innovations
in precision dermatology.
Regarding the topic of this manuscript, there is a significant gap in the literature, with
only a few studies briefly referencing interactions between microbiomics and metabolic
dysregulation within the broader pathophysiology of certain skin diseases. This review
uniquely focuses on the interplay between metabolic disorders, the skin microbiota, and
cutaneous health and disease. By addressing this critical gap, this manuscript provides a
fresh perspective on integrated strategies to improve dermatologic outcomes in patients
with metabolic disorders.
2. Methods
This manuscript is a narrative literature review synthesizing current knowledge on
the role of the skin microbiota as a mediator of interactions between metabolic disorders
and skin health. A comprehensive literature search was conducted in PubMed and Scopus
Microorganisms 2025, 13, 161 3 of 32
for studies published up to 22 December 2024. We utilized a wide range of keywords and
Medical Subject Headings (MeSH) terms to ensure a thorough search, including but not
limited to skin microbiota, metabolic disorders, obesity, diabetes mellitus, dysbiosis, skin
diseases, psoriasis, atopic dermatitis, microbial interactions, and ML. Boolean operators
were applied to refine and optimize the search strategy.
The inclusion criteria were as follows:
■ Study Type: Reviews or original research articles published in English.
■ Focus: Articles that addressed metabolic and immunological factors influencing the
skin microbiota and its role in skin diseases.
■ Therapeutic Studies: Relevant therapeutic studies were defined as those investigating
microbiota-targeted interventions (e.g., probiotics, postbiotics, or phage therapy)
and/or systemic treatments addressing underlying metabolic dysfunction. These
studies were assessed for clinical outcomes, mechanistic insights, and validation
through preclinical or clinical data.
■ ML Applications: Key records describing the application of ML tools or algorithms in
integrating omics datasets (e.g., metabolomics, microbiomics) and predicting disease
outcomes or therapeutic responses were included. Specific ML approaches, such
as supervised learning algorithms (e.g., random forests, support vector machines)
or unsupervised clustering methods, were highlighted for their contributions to
advancing precision dermatology.
The literature search was performed independently by two authors to ensure objec-
tivity and thoroughness. Any inconsistencies in study selection or data extraction were
resolved by consensus with a third author. Articles were initially screened based on their
titles and abstracts to assess relevance, followed by full-text reviews for those meeting the
inclusion criteria. Additional relevant studies were identified through manual reviews of
reference lists from selected articles. The findings were synthesized to present an integrated
and up-to-date perspective on the bidirectional interactions between metabolic disorders,
skin microbiota, and dermatologic health, along with therapeutic and computational ad-
vancements in the field.
study observed that only high-density lipoprotein cholesterol (HDL-C) levels were
significantly lower in the patient group compared to the control group [17].
■ According to a recent meta-analysis, MetS has been strongly associated with specific
skin diseases, including HS (OR: 4.46), LP (OR: 3.79), and SD (OR: 2.45). Psoriasis
also showed a significant correlation with MetS, although with high heterogeneity
(OR: 2.89). In contrast, rosacea exhibited a weaker association with MetS, with an
odds ratio (OR) of 1.56 (95% CI: 0.96–2.52). Nonetheless, a significant relationship was
observed between rosacea and high blood pressure (OR: 1.204, 95% CI: 1.09–1.33) as
well as insulin resistance (OR: 2.33, 95% CI: 1.18–4.60) [18].
A closer look at their molecular mechanisms reveals how these disorders drive chronic
inflammation and metabolic dysfunction.
3.1. T2DM
■ Core Mechanisms of T2DM [19,20]:
o Progressive insulin resistance and declining insulin secretion are hallmarks.
o Insulin resistance originates from defects in insulin receptor signaling, particu-
larly at the level of insulin receptor substrate-1 (IRS-1).
o Impaired IRS-1 phosphorylation disrupts interaction with phosphoinositide
3-kinase (PI3K), a critical enzyme for glucose uptake.
o Dysfunctional PI3K prevents activation of Akt (protein kinase B), inhibiting
GLUT4 translocation to the cell membrane and reducing glucose uptake in
skeletal muscle and adipocytes.
■ Role of Hyperglycemia and Glucotoxicity [19,20]:
o Chronic hyperglycemia exacerbates insulin resistance through glucotoxicity.
o High glucose levels increase the mitochondrial production of reactive oxygen
species (ROS), which damage IRS proteins and β-cells, further impairing
insulin sensitivity and secretion.
■ Systemic Metabolic Inflammation in T2DM (meta-inflammation) [19,20]:
o Mediated by pro-inflammatory cytokines largely produced by dysfunctional
adipose tissue, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6
(IL-6).
o TNF-α induces serine phosphorylation of IRS-1, impairing insulin receptor
signaling.
o IL-6 disrupts insulin sensitivity via the janus kinase/signal transducers and
activators of transcription (JAK/STAT) pathways.
■ Advanced Glycation End Products (AGEs) and RAGE Pathway [19–22]:
o Prolonged hyperglycemia in metabolic disorders leads to the non-enzymatic
glycation of proteins and lipids, forming AGEs.
o These molecules engage RAGE, a receptor expressed on immune and endothe-
lial cells, triggering several downstream effects.
o AGE-RAGE interactions stimulate the production of ROS through nicotinamide
adenine dinucleotide phosphate (NADPH) oxidase, amplifying oxidative stress.
o The resulting activation of NF-κB promotes the transcription of pro-inflammatory
cytokines, such as TNF-α and IL-1β, and vascular adhesion molecules, facili-
tating leukocyte recruitment and chronic inflammation.
o In the skin, AGEs have profound effects on structure and function. They
cross-link collagen and elastin fibers in the extracellular matrix, reducing skin
elasticity and barrier integrity.
Microorganisms 2025, 13, 161 5 of 32
3.2. Obesity
■ Core Characteristics [23–25]:
o Obesity is characterized by excessive and dysfunctional adipose tissue accu-
mulation, which acts as an active endocrine organ.
o Adipose tissue secretes hormones, cytokines, and adipokines that regulate
metabolism and immune responses.
■ Inflammatory Response [23–25]:
o Adipose tissue expansion leads to hypoxia, triggering macrophage infiltration.
o Infiltrating macrophages adopt a pro-inflammatory M1 phenotype, releasing
cytokines such as TNF-α, IL-1β, and IL-6.
■ Adipokine Dysregulation [23–25]:
o Dysregulated secretion of adipokines, including elevated leptin and resistin,
contributes to inflammation and metabolic dysfunction.
o Leptin: Promotes inflammation via activation of the JAK/STAT signaling
pathway.
o Resistin: Exacerbates insulin resistance by interacting with Toll-like receptor-4
(TLR-4).
■ Lipid Metabolism Dysregulation [23–25]:
o Excess circulating free fatty acids activate protein kinase C (PKC), impairing
insulin receptor signaling and exacerbating systemic insulin resistance.
o Lipotoxicity damages pancreatic β-cells and hepatocytes, further compound-
ing metabolic dysfunction.
■ Chronic Low-Grade Inflammation (Meta-Inflammation) [23–25]:
o A systemic inflammatory state driven by dysregulated adipokines and cytokines.
o This inflammatory milieu affects multiple organs and promotes insulin resis-
tance and obesity-related complications.
immune tolerance to commensals and activation against invading pathogens. For exam-
ple, S. epidermidis can activate TLR2 on keratinocytes to induce AMP production while
also promoting regulatory T-cell (Treg) activity, which prevents excessive inflammation.
These immune-regulating functions of the microbiota are crucial for maintaining a bal-
anced immune tone in the skin, reducing the likelihood of inflammatory and autoimmune
conditions [29].
Disruptions in the skin microbiota, known as dysbiosis, are implicated in a range
of skin diseases. Dysbiosis can arise from factors such as antibiotic use, harsh skincare
products, genetic predispositions, or underlying immune dysfunction. In inflammatory
conditions like AD, dysbiosis is mostly characterized by an overgrowth of S. aureus, which
exacerbates inflammation and disrupts the epidermal barrier. Psoriasis is another condition
linked to dysbiosis, characterized by additional shifts in the microbial composition that
may amplify T-helper (Th)17-mediated inflammatory responses. Specifically, in psoriasis,
there is an increase in S. aureus and S. pyogenes in plaques. Furthermore, psoriasis shows
a reduced presence of commensal bacteria such as Cutibacterium and Corynebacterium.
Similarly, acne vulgaris is associated with changes in the abundance and activity of C. acnes,
where certain strains may overproduce pro-inflammatory lipids and contribute to follicular
inflammation [30].
Fungal and viral components of the microbiota also play significant roles in skin health
and disease. Malassezia species, while commensal in healthy skin, are associated with
conditions such as SD and pityriasis versicolor when their growth becomes uncontrolled.
Viruses, such as human papillomaviruses and herpesviruses, can remain dormant in the
skin but may become pathogenic under conditions of immunosuppression or barrier
disruption. The interplay between microbial communities, the host immune system, and
external factors defines the balance between health and disease [30].
The composition of the skin microbiota undergoes significant changes with aging,
influenced by shifts in host factors such as sebaceous gland activity, skin lipids, natural
moisturizing factors (NMFs), and antimicrobial peptides (AMPs). A study examining
158 Caucasian females aged 20 to 74 found that bacterial diversity increases with age
across the forearm, buttock, and facial skin. Notably, the abundance of Cutibacterium and
Lactobacillus decreased with age at all sites, correlating with a reduction in sebaceous gland
size and lipid production, which are critical for maintaining these bacteria. Conversely,
genera such as Streptococcus and Anaerococcus showed site-specific increases, likely
driven by shifts in the availability of skin lipids and other host factors. Increased NMFs and
AMPs with age were positively correlated with bacterial genera like Corynebacterium and
Finegoldia, highlighting how host changes influence microbial dynamics. These findings
suggest that age-related alterations in skin biology drive the restructuring of microbial
communities [31].
Race and ethnicity also significantly influence microbiota composition, with differences
emerging as early as three months of age and persisting through childhood and adulthood.
Variability in microbial taxa is linked to inequitable environmental and social factors, rather
than biological differences. For example, specific taxa like Bifidobacterium and Lactobacil-
lus are enriched in racial groups with higher breastfeeding rates, while other differences
correlate with factors such as diet and hygiene. ML models using gut microbiome data from
children have demonstrated an 87% accuracy in predicting caregiver-identified race and
ethnicity, underscoring the role of structural disparities in shaping microbial communities.
These findings emphasize the need for personalized approaches in microbiome research to
address demographic variability and health disparities [32].
An overview of the commensal and pathogenic interactions of bacteria colonizing the
skin and/or being part of the human skin flora is presented in Table 1.
Microorganisms 2025, 13, 161 8 of 32
Furthermore, IL-6 signaling through its receptor complex (IL-6R and gp130) activates
the janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway in
keratinocytes, leading to the expression of pro-inflammatory genes and a shift in AMP pro-
duction. Cathelicidins (e.g., LL-37) and β-defensins, crucial AMPs that maintain microbial
balance, are downregulated, weakening the skin’s chemical defense against pathogens. The
cumulative effect of these changes is a weakened epidermal barrier that is highly permeable
to environmental insults and opportunistic microbes [35,36].
Emerging evidence highlights a critical link between glucose metabolism reprogram-
ming in keratinocytes and the pathogenesis of psoriasis, particularly in the context of
MetS. A recent study by Yan et al. (2024) demonstrated that psoriatic keratinocytes exhibit
enhanced glycolysis, evidenced by elevated levels of glycolysis-related metabolites such
as glucose-6-phosphate, fructose-1,6-bisphosphate, and lactate. In patients with psoriasis
and MetS, glycolysis-related protein expression (e.g., GLUT1, HK2, and PFKFB3) was
significantly upregulated, correlating with disease severity. Importantly, high-glucose
and high-fat culture intensified this metabolic reprogramming through the AKT/mTOR
pathway, exacerbating keratinocyte proliferation and inflammatory cytokine production.
Glycolysis inhibition via agents like 2-deoxyglucose effectively attenuated these pathologi-
cal features, offering potential therapeutic avenues [37].
Cathelicidin, an AMP, plays a dual role in innate immunity and systemic metabolic
regulation. Studies have linked LL-37 to inflammation and insulin resistance in MetS.
Activation of Toll-like receptors (e.g., TLR4) by LL-37 promotes pro-inflammatory cytokine
secretion, disrupting adipocyte insulin signaling and lipid metabolism. Notably, LL-37 lev-
els inversely correlate with HDL cholesterol while positively associating with triglycerides,
highlighting its potential role in atherogenic dyslipidemia. Beyond metabolic regulation,
LL-37 is implicated in psoriasis-related inflammation by modulating keratinocyte immune
responses and promoting cytokine production. These findings underscore LL-37’s role as
both a mediator of systemic inflammation and a biomarker for metabolic and inflammatory
conditions [38].
ability, impairing vasodilation and capillary perfusion. The resultant hypoxic conditions
activate hypoxia-inducible factor 1-alpha (HIF-1α) in keratinocytes and sebocytes, alter-
ing their metabolic activity. HIF-1α upregulates glycolytic enzymes, shifting cellular
metabolism toward anaerobic glycolysis. This metabolic shift reduces ATP availability and
affects keratinocyte turnover, creating niches that favor anaerobic or facultative anaerobic
microbes such as C. acnes and Pseudomonas aeruginosa. Reduced perfusion also delays the
clearance of metabolic byproducts and toxins, further modifying the microbial habitat [43].
Sweat gland dysfunction, commonly observed in diabetic neuropathy, exacerbates
these issues. Reduced sweat secretion alters ionic gradients and skin hydration, favor-
ing the colonization of halophilic and xerophilic species such as S. aureus and certain
fungi. Neural inputs that regulate microvascular tone and glandular activity are also
disrupted, creating patchy areas of skin with distinct microbial communities. Over time,
these microenvironmental disparities increase the risk of localized infections and biofilm
formation [43].
6. Psoriasis
Psoriasis is a chronic immune-mediated skin condition characterized by erythematous,
scaly plaques resulting from excessive keratinocyte proliferation and incomplete differen-
tiation. At the core of its pathogenesis lies the IL-23/IL-17 axis, driven by Th17 cells and
their cytokines, which create a pro-inflammatory environment. Recent evidence highlights
how metabolic disorders, such as obesity and MetS, and disruptions in the skin microbiome
interact to amplify the inflammatory pathways underlying psoriasis, forming a vicious
cycle of immune activation and microbial dysbiosis [44].
Dendritic cells play a critical role in psoriasis by responding to environmental trig-
gers, microbial antigens, and damage-associated molecular patterns like DNA and LL-37
complexes. These cells release IL-23, which is essential for maintaining Th17 cells. Th17
cells secrete IL-17A, IL-17F, and IL-22, which promote keratinocyte hyperproliferation and
inhibit proper differentiation. These cytokines synergistically upregulate AMPs such as
β-defensins and S100 proteins in keratinocytes, attracting neutrophils and intensifying
inflammation. This process becomes exacerbated in the presence of MetS, as hypertrophic
adipocytes and infiltrating macrophages secrete high levels of pro-inflammatory cytokines,
including TNF-α, IL-6, and IL-1β. These cytokines activate NF-κB signaling in keratinocytes
and immune cells, heightening inflammatory responses. Elevated leptin in obesity pro-
motes Th17 cell polarization, while decreased adiponectin removes an anti-inflammatory
control, further fueling psoriatic inflammation [45].
The microbiome in psoriasis is characterized by dysbiosis, marked by reduced mi-
crobial diversity and a shift toward pathogenic species. In psoriatic lesions, S. aureus and
Streptococcus pyogenes are overrepresented and can act as superantigens, stimulating T-cell
activation. Conversely, commensal bacteria like Cutibacterium and Corynebacterium are
diminished, leading to weakened immune regulation. Dysbiosis amplifies inflammation
through PRRs on keratinocytes, including TLRs (TLR2, TLR4) and NOD-like receptors, cre-
ating a feedback loop that sustains inflammation and further disrupts the microbiome [46].
Psoriatic plaques also exhibit altered lipid composition, including reduced ceramides,
elevated free fatty acids, and increased cholesterol sulfate. These changes compromise the
stratum corneum’s barrier integrity, increasing transepidermal water loss and exposing
underlying layers to microbial invasion. S. aureus exacerbates this disruption by producing
enterotoxins that activate IL-23 and IL-17 pathways, intensifying keratinocyte activation
and neutrophilic infiltration.
Recent studies emphasize the role of lipid metabolism dysregulation in psoriasis and
its potential therapeutic implications. For instance, a meta-analysis demonstrated signifi-
Microorganisms 2025, 13, 161 12 of 32
cantly higher levels of total cholesterol, LDL, and triglycerides, with a marked reduction in
HDL in psoriasis patients compared to controls (p < 0.05). These changes compromise the
stratum corneum’s barrier integrity, increasing transepidermal water loss and exposing
underlying layers to microbial invasion. Aberrant lipid profiles were further associated
with elevated serum IL-6, which positively correlated with the LDL/HDL ratio (r = 0.48,
p < 0.01). Dysregulated lipid metabolism contributes to microbial dysbiosis, where lipid-
rich psoriatic plaques foster pathogenic taxa such as Staphylococcus aureus while depleting
beneficial commensals of Cutibacterium. S. aureus exacerbates the epidermal barrier dis-
ruption by producing enterotoxins that activate IL-23 and IL-17 pathways, intensifying
keratinocyte activation and neutrophilic infiltration. Fungal dysbiosis, particularly with
Malassezia species, further contributes to psoriasis by degrading sebaceous lipids into
free fatty acids, which irritate keratinocytes and drive dendritic cell and Th17 activation,
especially in sebaceous-rich areas like the scalp [47,48].
Metabolic disorders amplify these processes through systemic effects. In obesity and
T2DM, AGEs accumulate in the skin, binding to RAGE receptors on keratinocytes and
endothelial cells. This activates MAPK and NF-κB pathways, triggering the production of
cytokines, chemokines, and adhesion molecules that perpetuate inflammation. AGEs also
impair dermal elasticity and microvascular function by cross-linking collagen and elastin,
reducing oxygen and nutrient delivery to psoriatic plaques. Hypoxia in plaques stabilizes
HIF-1α, which further drives keratinocyte hyperproliferation and AMP expression [49].
Dysregulated lipid metabolism in MetS compounds the problem. Elevated circulating
LDL and triglycerides infiltrate the dermis, where they oxidize and activate macrophages
via scavenger receptors such as CD36. This enhances the production of TNF-α and IL-
1β, creating a pro-inflammatory environment. Simultaneously, reduced ceramides in
psoriatic lesions impair epidermal lipid organization, weakening the skin’s ability to
prevent microbial colonization and promoting dysbiosis [50].
Therapeutic strategies targeting metabolic dysfunction have shown promise. Weight
loss through dietary interventions or bariatric surgery reduces systemic inflammation,
normalizes adipokine levels, and improves insulin sensitivity, thereby mitigating psori-
atic inflammation. Pharmacologic agents like glucagon-like peptide-1 receptor agonists
(GLP-1RAs) might improve psoriasis severity by addressing metabolic abnormalities and
meta-inflammation [51]. Experimental interventions targeting lipid metabolism, such as
fibrates and PPARγ agonists, demonstrated modest improvements in skin inflammation by
restoring microbial equilibrium. These findings suggest that managing lipid dysregulation
through microbiota-friendly dietary modifications or lipid-lowering agents could offer a
dual benefit in controlling systemic and localized inflammation [47,48].
Microbiome-targeted therapies are emerging as adjunctive treatments for psoriasis.
Topical or systemic probiotics that enhance commensal bacteria like Corynebacterium and
Cutibacterium are under investigation. For instance, lactobacilli have shown immunomodu-
latory effects by promoting regulatory Treg activity and suppressing Th17-driven inflam-
mation. Postbiotics are bioactive compounds produced during the fermentation process
by probiotic microorganisms. They include metabolic byproducts such as short-chain
fatty acids (SCFAs), bacteriocins, enzymes, vitamins, and other molecules that exert health
benefits. Unlike live probiotics, postbiotics are non-living and are considered safer for
use in certain populations. They play a role in modulating the microbiome, inhibiting
pathogenic microbes, and enhancing host physiological functions, including skin barrier
restoration [52].
An emerging area of interest in psoriasis management involves harnessing bacterio-
phages (phages) to selectively reduce pathogenic bacteria and rebalance the skin micro-
biome. Recent studies suggest that supplementing lesional skin with phages that target
Microorganisms 2025, 13, 161 13 of 32
7. Atopic Dermatitis
AD is a chronic inflammatory skin condition defined by intense itching, eczematous
lesions, and impaired epidermal barrier function. Its pathogenesis hinges on mutations
in structural proteins like filaggrin and dysregulated Th2 immune responses. Metabolic
disorders such as obesity and T2DM exacerbate AD by amplifying systemic inflammation,
altering lipid metabolism, and fostering microbial dysbiosis. Together, these factors perpet-
uate the cycle of barrier dysfunction, immune activation, and microbial imbalance central
to AD pathology [54].
The epidermal barrier serves as a critical defense against transepidermal water loss,
environmental allergens, and microbial invasion. Filaggrin, a key barrier protein, is crucial
for forming the cornified envelope and generating natural moisturizing factors such as
urocanic acid and pyrrolidone carboxylic acid, which maintain an acidic skin pH. Mutations
in the filaggrin gene, commonly seen in AD, reduce natural moisturizing factor produc-
tion, impair barrier integrity, and facilitate allergen and microbial penetration, initiating
inflammation. Metabolic disorders exacerbate this barrier dysfunction by promoting the
accumulation of AGEs. These molecules cross-link keratinocyte and extracellular matrix
proteins, stiffening the epidermis and activating RAGE on keratinocytes. RAGE signaling
induces NF-κB activation, increasing the production of pro-inflammatory cytokines such as
IL-1β, TNF-α, and IL-6, which further impair keratinocyte differentiation and reduce the
expression of filaggrin, involucrin, and loricrin [35,36,54].
Th2-driven inflammation dominates AD’s immune landscape, with cytokines like IL-4,
IL-5, and IL-13 impairing keratinocyte differentiation and suppressing AMP production.
IL-4 and IL-13 further reduce filaggrin expression and promote IgE-mediated allergic
sensitization. Chronic inflammation can shift to mixed Th2/Th22 or Th1/Th17 responses,
especially in severe or chronic lesions. Metabolic inflammation amplifies these immune
dysregulations, with adipokines like leptin promoting Th2 polarization and IL-4/IL-13
production. Simultaneously, adiponectin, an anti-inflammatory adipokine, is reduced in
obesity and T2DM, removing a regulatory brake on Th2 activity. This dual effect intensifies
AD’s chronic inflammatory state [55].
AD is strongly associated with microbial dysbiosis, characterized by overgrowth of
S. aureus and diminished diversity of commensal microbes. S. aureus plays a direct role in
inflammation by producing virulence factors, including superantigens like staphylococcal
enterotoxins that activate T cells and amplify Th2 and Th17 responses. Additionally, S. au-
Microorganisms 2025, 13, 161 14 of 32
reus secretes proteases, such as V8 protease, which degrade filaggrin and further weaken the
epidermal barrier. Its ability to form biofilms enhances resistance to antimicrobial defenses
and contributes to persistent inflammation. Hyperglycemia and altered lipid metabolism
in metabolic disorders exacerbate S. aureus colonization by providing an environment rich
in nutrients and substrates, including glucose and free fatty acids. Dysregulated AMP
production in AD further weakens microbial control, allowing S. aureus to dominate and
outcompete commensals [56].
Lipid composition in the stratum corneum is critical for maintaining barrier integrity
and microbial balance. Ceramides, cholesterol, and free fatty acids are organized into lamel-
lar structures that prevent TEWL and microbial penetration. Systemic lipid abnormalities
in AD, such as increased LDL cholesterol and altered HDL composition, correlate with
disease severity and systemic inflammation, as measured by SCORAD scores (r = 0.64,
p < 0.001). Notably, ceramide abnormalities in the stratum corneum impair the lipid barrier,
predisposing the skin to colonization by Staphylococcus aureus and contributing to chronic
inflammation. Furthermore, metabolic disorders exacerbate cutaneous lipid abnormalities
by influencing sebaceous gland activity and epidermal lipid synthesis. For instance, insulin
resistance increases the ratio of saturated to unsaturated fatty acids, favoring colonization
by pro-inflammatory microbes like S. aureus over commensal species such as C. acnes. S.
aureus lipases further degrade lipids into irritant free fatty acids, aggravating keratinocyte
dysfunction and inflammation [57].
Clinically, patients with AD and coexisting metabolic disorders often present with
more severe and widespread lesions. Systemic inflammation, oxidative stress, and dysbiosis
associated with metabolic dysfunction worsen disease activity and reduce responsiveness
to standard treatments like topical corticosteroids and calcineurin inhibitors. However, im-
proved metabolic control can significantly mitigate these effects. Weight loss and glycemic
management reduce systemic inflammation, restore AMP production, and improve barrier
integrity, thereby decreasing S. aureus colonization and inflammation [14].
Emerging microbiome-targeted therapies are gaining attention in AD management.
Probiotics, including lactobacilli and Bifidobacterium species, enhance microbial diversity
and modulate immune responses by increasing Treg activity and reducing Th2 polariza-
tion. Postbiotics, such as SCFAs, strengthen the epidermal barrier and suppress S. aureus
virulence [52].
Recent research demonstrates that bacteriophages can specifically target Staphylococ-
cus aureus—a bacterial species closely linked to AD flares—while preserving beneficial
skin microbes such as S. epidermidis. In a 2020 study investigating a phage called SaGU1,
scientists observed a sharp reduction in S. aureus populations both in vitro and on the skin
of atopic mouse models. Notably, SaGU1 did not harm S. epidermidis, suggesting a targeted
antimicrobial effect. In vitro, S. aureus began re-emerging after 14 h, which may reflect the
development of phage resistance; however, adding S. epidermidis alongside SaGU1 pre-
vented this bacterial rebound, highlighting a potential synergy between beneficial skin flora
and phage therapy. Interestingly, in vivo experiments demonstrated that phage therapy
alone was effective enough to suppress S. aureus overgrowth, with no statistically significant
additional benefit observed when S. epidermidis was combined with SaGU1. These findings
underscore the promise of phage-based treatments in AD, particularly those designed to
selectively eliminate pathogenic staphylococci while leaving commensals intact. Future
research will clarify the best formulations, dosing strategies, and adjunctive options—such
as pairing phages with topical probiotics or supportive skincare regimens—to enhance
barrier function and achieve longer-lasting disease control in AD [53].
When combined with metabolic interventions, these approaches offer a holistic strat-
egy for managing AD, addressing both its systemic and localized drivers. This dual
Microorganisms 2025, 13, 161 15 of 32
focus could lead to more effective, long-term control of AD in patients with metabolic
dysfunctions [58].
8. Acne
Acne vulgaris is a multifactorial inflammatory condition that primarily affects the
pilosebaceous unit. It is clinically characterized by comedones, inflammatory papules, pus-
tules, and nodules. Its pathogenesis is driven by hyperseborrhea, follicular hyperkeratiniza-
tion, microbial dysbiosis—particularly involving C. acnes—and inflammation. Metabolic
disorders, such as obesity and insulin resistance, exacerbate acne through systemic and lo-
calized mechanisms, including altered sebaceous gland activity, lipid composition changes,
immune dysregulation, and microbial shifts, fostering an environment conducive to acne
development [59].
Emerging evidence highlights a significant relationship between insulin resistance and acne
vulgaris. In a study by Gruszczyńska et al., involving 41 acne vulgaris patients and 47 healthy
BMI-matched controls, insulin resistance was assessed using the homeostasis model assessment
of insulin resistance (HOMA-IR). The mean HOMA-IR value was significantly higher in the
acne group (3.40 ± 1.49) compared to controls (2.34 ± 0.91, p < 0.001). Furthermore, 78% of
acne patients met the criteria for insulin resistance (HOMA-IR > 2.1), versus 55% in the control
group (p = 0.026). When the cut-off value was adjusted to 2.69 (as per the Polish population
standard), 63% of acne patients and 30% of controls were diagnosed with insulin resistance,
further emphasizing the association (p = 0.002). Elevated fasting glucose levels were also observed
in the acne group (94.88 ± 7.73 mg/dL) compared to controls (79.51 ± 7.18 mg/dL, p < 0.001).
These findings suggest that insulin resistance may be an independent factor in acne pathogenesis,
warranting consideration during diagnosis and treatment [60].
Elevated insulin levels increase the secretion of insulin-like growth factor-1 (IGF-1),
which directly enhances sebaceous gland activity. IGF-1 activates the phosphoinositide
3-kinase (PI3K)/Akt and mammalian target of rapamycin complex 1 (mTORC1) pathways,
driving sebocyte proliferation and increasing sebum lipid synthesis. The overproduc-
tion of sebum provides a nutrient-rich substrate for C. acnes, promoting its colonization
and metabolic activity. Moreover, IGF-1 suppresses the nuclear receptor peroxisome
proliferator-activated receptor gamma (PPAR-γ), disrupting sebocyte differentiation and
lipid homeostasis, which further contributes to sebaceous gland dysfunction [60,61].
Additionally, obesity and insulin resistance compound acne-related inflammation
through the secretion of pro-inflammatory adipokines like leptin, which promotes Th1 and
Th17 polarization and increases cytokines such as interferon-gamma (IFN-γ), IL-17, and
IL-22. These cytokines drive keratinocyte hyperproliferation and sebaceous gland dysfunc-
tion. Simultaneously, reduced adiponectin removes a critical anti-inflammatory mediator,
exacerbating immune activation. Hyperglycemia further promotes the accumulation of
AGEs, which bind to RAGE on keratinocytes and immune cells, activating NF-κB and
MAPK pathways and amplifying the inflammatory milieu [60,61].
The composition of sebaceous lipids undergoes significant changes in metabolic disor-
ders. Insulin resistance shifts the balance toward a higher ratio of saturated to unsaturated
fatty acids, which have pro-inflammatory properties. Saturated fatty acids stimulate ker-
atinocytes via TLR2, promoting cytokine production, including IL-1β and IL-8. These
cytokines recruit neutrophils and drive inflammation. Concurrently, oxidative stress associ-
ated with hyperglycemia generates lipid peroxidation products, such as malondialdehyde,
which disrupt sebaceous lipid profiles, exacerbating inflammation [60,61].
Polycystic ovary syndrome (PCOS), a common metabolic–endocrine disorder, is in-
tricately linked to acne through hyperandrogenism and insulin resistance. Elevated an-
drogens, such as testosterone and dihydrotestosterone, stimulate sebaceous gland hyper-
Microorganisms 2025, 13, 161 16 of 32
by promoting the persistence of pathogenic C. acnes strains, making their disruption a key
target for acne therapy. In addition, SCFAs modulate keratinocyte differentiation and lipid
metabolism, contributing to a healthier skin barrier and reducing the sebum production
that fuels C. acnes growth [52,67].
Bacteriocins, another class of postbiotics, are antimicrobial peptides produced by
probiotics that specifically target pathogenic bacteria without affecting commensal popu-
lations. For instance, bacteriocins like nisin and pediocin have demonstrated the ability
to selectively inhibit C. acnes proliferation. They achieve this by permeabilizing bacterial
membranes or interfering with cell wall synthesis, offering a highly targeted approach to
microbial regulation [67].
Furthermore, emerging research suggests that probiotics and postbiotics can alter the
skin’s pH and lipid composition, creating an environment less conducive to pathogenic bac-
terial colonization. By promoting the growth of beneficial commensals such as Staphylococ-
cus epidermidis, these interventions not only counteract the dominance of acne-associated
C. acnes strains but also restore microbial diversity—a hallmark of healthy skin. Preclinical
studies have shown that S. epidermidis can antagonize C. acnes through the production of
antimicrobial peptides, further emphasizing the synergistic role of commensal bacteria in
acne management [67].
In addition to their direct effects on the microbiome, probiotics and postbiotics have
systemic benefits. Oral administration of probiotics has been linked to reduced systemic
inflammation and oxidative stress, factors that exacerbate acne severity. These benefits
extend to the regulation of metabolic pathways, including insulin sensitivity and lipid
metabolism, which are often dysregulated in individuals with acne [67].
Bacteriophage-based treatments for acne aim to selectively eliminate C. acnes without
disturbing the broader skin microbiome. Recent murine experiments have shown that
injecting bacteriophages targeting C. acnes can reduce inflammatory nodules and epidermal
thickening in mice, supporting the therapeutic potential of phages as an alternative or
complement to antibiotics—especially given growing antibiotic resistance. Topical formu-
lations containing C. acnes-lytic phages have also been investigated: semi-solid creams
and hydrogels maintained significant phage viability for weeks to months under proper
storage conditions (e.g., refrigeration and limited light exposure). Preliminary human trials
with topical phage cocktails have demonstrated a dose-dependent reduction in C. acnes
colonization, suggesting that optimized phage concentrations could be key to improving
clinical outcomes. Future research will need to explore long-term efficacy, ideal phage com-
binations, and potential synergy with established acne therapies such as topical retinoids
or benzoyl peroxide. If validated, phage therapy could offer patients a novel, targeted
approach to controlling C. acnes overgrowth and associated inflammation while preserving
beneficial cutaneous microbes [53].
These approaches, combined with metabolic interventions, offer a comprehensive
strategy for managing acne, addressing both the systemic and localized drivers of this
multifactorial disease [67].
9. Rosacea
Rosacea is a chronic inflammatory skin disorder affecting approximately 5% of the
global population. It presents as facial erythema, telangiectasia, and inflammatory lesions
such as papules and pustules, with severe cases leading to phymatous changes character-
ized by thickened and irregular skin. Beyond its visible manifestations, rosacea involves
complex interactions between immune dysregulation, neurovascular mechanisms, and
environmental triggers. Increasingly, the role of the skin microbiome and its interaction
Microorganisms 2025, 13, 161 18 of 32
with metabolic dysfunctions, such as obesity and dyslipidemia, has come under scrutiny,
highlighting their collective contribution to rosacea’s pathogenesis [68].
Rosacea is strongly associated with alterations in the skin microbiome, particularly an
overgrowth of Demodex mites, including Demodex folliculorum and Demodex brevis. These
mites are found in significantly higher densities on the skin of rosacea patients—ranging
from 0.7/cm² in healthy controls to 10.8/cm² in affected individuals. Their exoskeleton and
endosymbiotic bacterium Bacillus oleronius contribute to inflammation by releasing proteins
and heat-shock proteins that activate Toll-like receptor 2 (TLR2) on keratinocytes, trigger-
ing innate immune responses. This activation results in the production of cathelicidins,
AMPs that are excessively expressed in rosacea and contribute to both the condition’s
pro-inflammatory and vasoactive properties. These inflammatory responses are further
amplified by metabolic dysfunction. For instance, elevated levels of TNF-α and IL-6 in
obesity and MetS enhance microbial overgrowth and contribute to persistent inflamma-
tion, creating a feedback loop between dysbiosis and immune activation. Additionally,
Staphylococcus epidermidis overgrowth and reduced C. acnes abundance are noted, with
other species like Geobacillus and Gordonia correlating with disease severity. These microbial
disruptions, alongside environmental and neurological factors, exacerbate skin barrier
dysfunction, promote inflammation, and intensify rosacea symptoms, including erythema
and telangiectasia [69–71].
The neurovascular component of rosacea intricately interfaces with the microbiome
through activation of transient receptor potential (TRP) channels, particularly TRPV1 and
TRPA1. These channels, expressed on sensory neurons, keratinocytes, and immune cells,
are highly sensitive to microbial metabolites such as SCFAs, lipopolysaccharides, and other
bacterial byproducts. Environmental stimuli, including ultraviolet (UV) radiation, heat,
and dietary triggers (e.g., capsaicin and alcohol), further potentiate their activation. Upon
activation, TRPV1 and TRPA1 stimulate the release of pro-inflammatory neuropeptides,
such as substance P and calcitonin gene-related peptide (CGRP), which contribute to
neurogenic inflammation. This cascade disrupts neurocutaneous signaling, impairing
the production of AMPs such as cathelicidins and β-defensins. AMPs are critical for
maintaining microbial homeostasis by directly inhibiting pathogenic bacterial growth and
preserving skin barrier integrity. Dysregulated signaling creates a permissive environment
for microbial overgrowth, particularly of Demodex folliculorum, which harbors symbiotic
bacteria such as Bacillus oleronius. These bacteria release antigens that further amplify
inflammation by activating Toll-like receptor 2 (TLR2) on keratinocytes and immune cells.
Additionally, the altered microenvironment fosters the growth of opportunistic pathogens,
including S. epidermidis and C. acnes. The interaction between these microbial populations
and the impaired neurovascular signaling exacerbates inflammatory pathways, disrupts
skin barrier function, and reinforces the chronicity of rosacea. Importantly, the feedback
loop of inflammation, microbial imbalance, and neurovascular dysfunction underscores
the need for targeted therapies addressing both microbial and neurovascular components
in rosacea management [69,70].
Cutaneous barrier dysfunction in rosacea exacerbates microbial dysbiosis. Downregu-
lation of the ABCA12 gene, a key regulator of lipid lamellae formation, has been observed
in rosacea patients. This defect compromises the skin barrier, increasing transepidermal
water loss and facilitating microbial infiltration. Dyslipidemia further alters the skin’s
lipid environment. Elevated LDL and triglycerides, along with reduced HDL, shift the
sebaceous lipid composition, favoring conditions conducive to pathogenic growth. For
example, S. epidermidis, typically a commensal, may adopt pathogenic traits in response
to an altered lipid environment, while oxidative modifications of lipids, such as oxidized
Microorganisms 2025, 13, 161 19 of 32
substrates for Malassezia. Through lipase activity, Malassezia species metabolize these lipids
into pro-inflammatory free fatty acids that irritate keratinocytes, inducing the release of
IL-6 and IL-1β. These cytokines drive inflammation, disrupt lipid organization in the
stratum corneum, and weaken the epidermal barrier, perpetuating clinical symptoms such
as scaling, redness, and skin peeling. The inflammatory microenvironment also stimulates
abnormal keratinocyte proliferation, further disrupting the epidermal barrier. Although
increased sebum production was historically considered essential in SD, newer evidence
suggests that seborrhea is not strictly required, underscoring the importance of microbiota
and immune factors in disease pathogenesis [51,57].
Altered lipid metabolism contributes significantly to SD’s pathology. Skin lipidomics
studies have revealed unique profiles in SD lesions compared to healthy skin, with older
research identifying decreased squalene, wax esters, and free fatty acids alongside increased
cholesterol and triglycerides in lesional skin. These findings suggest disrupted lipid
homeostasis due to abnormal keratinization and sebaceous gland dysfunction. Recent
work has shown higher mean skin surface lipid levels in SD patients, correlating with
disease severity. Systemic lipid abnormalities, including elevated total cholesterol, LDL,
and reduced HDL, are also common in SD patients, with studies linking these dyslipidemias
to increased SD severity. Reduced HDL, in particular, diminishes its antimicrobial functions,
allowing Malassezia overgrowth, sustaining inflammation, and exacerbating dysbiosis [57].
Malassezia species are central to SD, with their lipase-driven metabolism of sebaceous
lipids fueling inflammation. By hydrolyzing triglycerides into free fatty acids, Malassezia
disrupts keratinocyte membranes and activates PRRs, such as TLR2, triggering cytokine
production and neutrophil recruitment. Different species, including M. globosa and M. re-
stricta, exhibit variations in lipase activity and immune interactions, leading to site-specific
manifestations. In SD-affected skin, microbial dysbiosis is evident, with reduced bacterial
diversity and a dominance of Malassezia. This imbalance disrupts the competitive effects of
commensal bacteria, such as S. epidermidis, further enabling Malassezia colonization. Addi-
tionally, altered ceramide and squalene levels in SD create a lipid environment favorable
for Malassezia proliferation, exacerbating inflammation [74].
The exaggerated inflammatory response in SD involves the upregulation of cytokines
like TNF-α, IL-6, and IL-1β, alongside activation of NF-κB and MAPK pathways. These
inflammatory signals promote keratinocyte hyperproliferation, barrier dysfunction, and
scaling. MetS and dyslipidemia amplify these immune dysregulations, with hypertriglyc-
eridemia and elevated LDL levels activating TLRs on keratinocytes and macrophages. This
activation promotes further cytokine release and oxidative stress, creating a feedback loop
between systemic lipid imbalances and local skin inflammation. Reduced HDL removes
a critical anti-inflammatory factor, worsening the inflammatory cascade and microbial
dysbiosis in SD [75].
Treatment of SD traditionally focuses on managing inflammation and Malassezia
overgrowth. Topical antifungal agents, such as ketoconazole and ciclopirox, and anti-
inflammatory therapies, such as calcineurin inhibitors and corticosteroids, form the corner-
stone of therapy. For severe cases, oral antifungals like itraconazole or systemic retinoids
may be required. However, systemic metabolic factors offer additional therapeutic targets.
Weight loss and dietary interventions improve lipid profiles and reduce systemic inflam-
mation, indirectly benefiting SD. Statins, which lower LDL and exhibit anti-inflammatory
effects, could serve as an adjunct therapy in SD patients with significant dyslipidemia or
MetS [76].
Emerging therapies targeting lipid metabolism and the skin microbiome present ex-
citing opportunities for SD management. Topical formulations restoring ceramides and
squalene levels may strengthen the epidermal barrier and reduce Malassezia colonization.
Microorganisms 2025, 13, 161 21 of 32
Probiotics and postbiotics, aimed at enhancing microbial diversity, can suppress inflamma-
tion and dysbiosis. For instance, probiotics such as lactobacilli and Bifidobacterium species
modulate immune responses and increase commensal bacterial populations, countering
Malassezia overgrowth. Postbiotic compounds, including SCFAs, may further stabilize
microbial communities and reduce cytokine-driven inflammation [77,78].
Specifically, a recently published study evaluated the efficacy of EUTOPLAC, a
probiotic-enriched oily suspension containing Lactobacillus crispatus P17631 and Lacticas-
eibacillus paracasei I1688, in modulating the skin mycobiome–bacteriome and alleviating
SD symptoms. The study enrolled 25 patients with moderate to severe SD who applied
EUTOPLAC daily for one week. Symptom severity, assessed using the Seborrheic Der-
matitis Area Severity Index (SDASI), significantly improved, with mean SDASI scores
decreasing from baseline (T0) to T8 (p < 0.0001). Improvements persisted three weeks
post-treatment (T28), though scores plateaued between T8 and T28. Fungal diversity,
measured via Shannon and Pielou indices, significantly increased at T8 (Shannon index:
2.66 ± 0.99 vs. T0: 1.37 ± 0.07; p < 0.0001), accompanied by a reduction in Malassezia
relative abundance (74.8% at T8 vs. 96.5% at T0; p < 0.0001). By T28, Malassezia abundance
partially rebounded to 93.0%. Bacteriome analysis showed a marked rise in Lactobacillus
and Lacticaseibacillus genera at T8 (23.1% and 0.6%, respectively, vs. 0.1% and 0.003%
at T0; p < 0.0001), accompanied by a significant decrease in Staphylococcus (6.9% at T8
vs. 28.7% at T0; p < 0.0001). These effects were transient, normalizing by T28. Network
analysis revealed significant disruptions in microbial correlations at T8, reflecting transient
community instability post-treatment. These findings highlight EUTOPLAC’s potential
as a targeted SD therapy, offering temporary symptom relief and microbiome modulation
without long-term dysbiosis. Further randomized controlled trials are needed to confirm
these benefits and optimize probiotic formulations [77].
These integrative approaches, targeting both systemic and local factors, hold promise
for improving outcomes in SD while addressing its complex pathophysiology [76–78].
support the role of GLP1-RAs in addressing both metabolic and inflammatory components of
HS, warranting further investigation in controlled trials [82].
Microbiome-targeted therapies are emerging as adjuncts in HS management. Pro-
biotics, such as lactobacilli and Bifidobacterium strains, enhance microbial diversity and
modulate immune responses by increasing Treg activity and suppressing Th17-driven
inflammation. Postbiotics, including SCFAs, directly inhibit pathogenic biofilm formation
and virulence while promoting commensal populations. These approaches, combined with
systemic metabolic interventions, provide a comprehensive strategy for addressing both the
localized and systemic drivers of HS, highlighting the central role of the skin microbiome
in mediating metabolic interactions in this debilitating condition [81].
Lifestyle interventions, including weight loss, smoking cessation, and dietary adjust-
ments, play a significant role in managing HS. These modifications not only have the
potential to alleviate disease symptoms but also influence microbial diversity. For example,
smoking has been associated with an increase in the gut phyla Proteobacteria and Bac-
teroidetes while reducing Actinobacteria and Firmicutes. Smoking also lowers overall gut
microbiome diversity through mechanisms such as oxidative stress, alterations in mucin
composition, and disruption of intestinal tight junctions. Consequently, smoking cessation
may restore microbial balance, providing an additional pathway for therapeutic benefit.
Probiotics have emerged as a promising adjunctive treatment in HS due to their ability to
restore microbial homeostasis, particularly by increasing beneficial skin bacteria such as
Cutibacterium spp., Corynebacterium, and Staphylococcus. Targeting prelesional skin with
topical probiotics or using oral formulations to modulate gut microbiota offers potential
therapeutic advantages. Probiotics can impact systemic inflammation, oxidative stress,
glycemic control, and lipid metabolism, all of which are implicated in HS pathogenesis.
While extensive evidence supports the use of probiotics in conditions like AD and psoria-
sis, their role in HS is still underexplored. More studies are needed to identify the most
effective strains and their mechanisms. Dietary interventions, such as reducing high-fat
and high-sugar intake, avoiding dairy, and eliminating brewer’s yeast, may also benefit
HS patients. However, the full impact of diet on HS severity remains insufficiently under-
stood, necessitating further research to establish evidence-based dietary recommendations.
Together, these lifestyle and dietary approaches could pave the way for personalized HS
management strategies [81].
A schematic illustration of the effects of Metabolic Disorders on Skin Microbiome and
Cutaneous Health is presented in Figure 1. An overview of the correlations between skin
diseases, microbiota shifts, metabolic influences, and therapeutic opportunities is presented
in Table 2. A flowchart of the complex interactions between AGE-RAGE pathway and the
skin is illustrated in Figure 2.
Table 2. Correlation between skin diseases, microbiota shifts, metabolic influences, and therapeutic
opportunities.
Table 2. Cont.
dysregulation plays a significant role, as adipokines like leptin promote Th1/Th17 polarization,
while reduced adiponectin removes anti-inflammatory control, intensifying immune activation and
microbial imbalances. Neurovascular dysregulation, a notable mechanism in rosacea, is driven by
the increased activation of pathways such as TRPV1 channels and exacerbates skin sensitivity and
dysbiosis. Microvascular dysfunction and reduced capillary perfusion create hypoxic conditions
that favor anaerobic or facultative anaerobic microbes, altering microbial ecology. Dysregulated
Microorganisms 2025, 13, x FOR PEER lipid
REVIEWmetabolism, particularly altered sebaceous gland activity in insulin resistance, 26 of 33 leads to changes
in sebum composition, such as increased saturated fatty acids, which promote the colonization of
pathogenic microbes and disrupt the balance of commensal microbes. Systemic nutritional and
dietary interventions
metabolic influences, including hyperglycemia to im- provide substrates for microbial
and dyslipidemia,
prove lipid profiles, anti-in-
growth, destabilizing skin homeostasis. Oxidative stress and lipid peroxidation further damage ker-
flammatory therapies (e.g.,
atinocytes and lipids, compromising skin integrity and promoting microbial overgrowth. Sebaceous
calcineurin inhibitors).
gland hyperactivity, induced by hyperinsulinemia and IGF-1, stimulates excessive lipid production,
GLP-1 receptor agonists (e.g.,
creating a nutrient-rich environmentliraglutide),
for opportunistic microbes.
weight loss, tar- Cytokines and oxidative stress
reduce over-
Reduced diversity, the expression of barrier proteins
Obesity, hyperinsuline- getedlike filaggrin
probiotics, and involucrin, increasing transepidermal
postbiotics,
Hidradenitis
growth of S.water
aureus,loss
S. and
mia,weakening physical defenses
elevated IL-17/TNF- biologicsagainst microbial
(e.g., TNF-α inhibi-invasion. AGEs, formed under hy-
[15,57,79–82]
Suppurativa
pyogenes, and C. acnes. α,
perglycemic dyslipidemia.
conditions, tors), lifestyle
bind to their receptor RAGE, interventions to
triggering NF-κB-mediated inflammation and
oxidative stress. This process impairs address metabolic
skin barrier dysregula-
proteins, disrupts collagen cross-linking, and
tion.
affects keratinocyte function. These mechanisms collectively illustrate how metabolic disorders create
Abbreviations. AGEs (Advanced Glycation End Products);
both systemic and localized environments conducive GLP-1to(Glucagon-Like Peptide-1);
skin dysbiosis, IL (In-
inflammation, and disease,
terleukin); IGF-1 (Insulin-Like Growth Factor-1); SCFAs (Short-Chain Fatty Acids); TNF-α (Tumor
underscoring the need for integrated therapeutic strategies targeting metabolic dysfunction and skin
Necrosis Factor-Alpha).
health. Systemic effects are marked in blue, while localized effects are marked in orange.
Flowchart
Figure2.2.Flowchart
Figure of complex
of the the complex interactions
interactions betweenbetween AGE-RAGE
AGE-RAGE pathway
pathway and the skin.and the skin. Abbrevi-
Abbre-
ations. AGEs
viations. (Advanced
AGEs (Advanced Glycation
Glycation EndEnd Products);
Products); AMP (Antimicrobial
AMP (Antimicrobial Peptides);Peptides);
NF-κB (Nu-NF-κB (Nuclear
Factor
clear Kappa-Light-Chain-Enhancer
Factor Kappa-Light-Chain-Enhancer ofof Activated
Activated B cells);
B cells); RAGERAGE (Receptor
(Receptor for Advanced
for Advanced Gly- Glycation
End Products);
cation ROS
End Products); (Reactive
ROS (ReactiveOxygen Species);TEWL
Oxygen Species); TEWL (Transepidermal
(Transepidermal Loss); Loss);
WaterWater (in- ↑ (increased);
(decreased).
↓ (decreased).
creased);
Microorganisms 2025, 13, 161 26 of 32
regulation profoundly affects microbial equilibrium, immune responses, and skin barrier
function, contributing to disease onset and progression.
Comprehensive management strategies that address metabolic dysfunction—through
lifestyle modifications, pharmacological interventions like GLP-1 receptor agonists and
SGLT-2 inhibitors, and bariatric surgery—can mitigate systemic inflammation and re-
store microbial homeostasis. Concurrently, microbiome-targeted therapies, including
probiotics, prebiotics, postbiotics, and bacteriophage treatments, have shown promise
in re-establishing microbial balance and enhancing barrier integrity. Integrating these
approaches with ML models allows for the identification of predictive biomarkers and
enables the development of highly personalized, effective therapeutic strategies.
Future research must focus on overcoming clinical translation barriers, such as patient
variability, regulatory challenges, and the lack of long-term studies on microbiota-targeted
therapies. High-resolution, longitudinal studies integrating multi-omics datasets will be
essential to elucidate host–microbiome dynamics. Interdisciplinary collaboration across
dermatology, endocrinology, microbiology, and computational biology will be crucial
in advancing precision medicine. These efforts aim to create durable, patient-specific
treatments that bridge systemic and localized therapies, ultimately improving outcomes
and reducing the burden of skin diseases influenced by metabolic dysfunction.
Author Contributions: Conceptualization, M.K. and N.T.; methodology, M.N. and G.F.; writing—original
draft preparation, M.K., N.T. and A.A.; writing—review and editing, G.F., A.S., M.N. and K.K.; visualization,
K.K.; supervision, A.S. All authors have read and agreed to the published version of the manuscript.
References
1. Garus-Pakowska, A. Metabolic Diseases—A Challenge for Public Health in the 21st Century. Int. J. Environ. Res. Public Health
2023, 20, 6789. [CrossRef] [PubMed]
2. Theodorakis, N.; Nikolaou, M. Integrated Management of Cardiovascular—Renal—Hepatic—Metabolic Syndrome: Expanding
Roles of SGLT2is, GLP-1RAs, and GIP/GLP-1RAs. Biomedicines 2025, 13, 135. [CrossRef]
3. Mehta, A.S. Metabolic Syndrome and Skin: Interactions and Implications. Indian J. Dermatol. 2022, 67, 138–145. [CrossRef]
4. Santiago-Rodriguez, T.M.; Le François, B.; Macklaim, J.M.; Doukhanine, E.; Hollister, E.B. The Skin Microbiome: Current
Techniques, Challenges, and Future Directions. Microorganisms 2023, 11, 1222. [CrossRef] [PubMed]
5. Naik, P.P.; Farrukh, S.N. Clinical Significance of Diabetic Dermatopathy. Diabetes Metab. Syndr. Obes. 2020, 13, 4823–4827.
[CrossRef]
6. Witte, K.; Wolk, K.; Witte-Händel, E.; Krause, T.; Kokolakis, G.; Sabat, R. Targeting Metabolic Syndrome in Hidradenitis
Suppurativa by Phytochemicals as a Potential Complementary Therapeutic Strategy. Nutrients 2023, 15, 3797. [CrossRef]
[PubMed]
7. Mustata, M.-L.; Neagoe, C.-D.; Ionescu, M.; Predoi, M.-C.; Mitran, A.-M.; Ianosi, S.-L. Clinical Implications of Metabolic Syndrome
in Psoriasis Management. Diagnostics 2024, 14, 1774. [CrossRef] [PubMed]
8. Kim, J.; Kim, B.E.; Goleva, E.; Berdyshev, E.; Bae, J.; Kim, S.; Kim, H.Y.; Lee, U.H.; Kim, M.S.; Jung, M.; et al. Alterations of
Epidermal Lipid Profiles and Skin Microbiome in Children with Atopic Dermatitis. Allergy Asthma Immunol. Res. 2023, 15,
186–200. [CrossRef]
9. Duff, M.; Demidova, O.; Blackburn, S.; Shubrook, J. Cutaneous Manifestations of Diabetes Mellitus. Clin. Diabetes 2015, 33, 40–48.
[CrossRef] [PubMed]
10. Chen, D.; He, J.; Li, J.; Zou, Q.; Si, J.; Guo, Y.; Yu, J.; Li, C.; Wang, F.; Chan, T.; et al. Microbiome and Metabolome Analyses Reveal
Novel Interplay Between the Skin Microbiota and Plasma Metabolites in Psoriasis. Front. Microbiol. 2021, 12, 643449. [CrossRef]
11. Theodorakis, N.; Feretzakis, G.; Tzelves, L.; Paxinou, E.; Hitas, C.; Vamvakou, G.; Verykios, V.S.; Nikolaou, M. Integrating
Machine Learning with Multi-Omics Technologies in Geroscience: Towards Personalized Medicine. J. Pers. Med. 2024, 14, 931.
[CrossRef]
12. Wu, J.J.; Kavanaugh, A.; Lebwohl, M.G.; Gniadecki, R.; Merola, J.F. Psoriasis and Metabolic Syndrome: Implications for the
Management and Treatment of Psoriasis. J. Eur. Acad. Dermatol. Venereol. 2022, 36, 797–806. [CrossRef] [PubMed]
Microorganisms 2025, 13, 161 29 of 32
13. Okpala, I.C.; Akinboro, A.O.; Ezejoifor, I.O.; Onunu, A.N.; Okwara, B.U. Metabolic Syndrome and Dyslipidemia among Nigerians
with Lichen Planus: A Cross-Sectional Study. Indian J. Dermatol. 2019, 64, 303–310. [CrossRef] [PubMed]
14. Shalom, G.; Dreiher, J.; Kridin, K.; Horev, A.; Khoury, R.; Battat, E.; Freud, T.; Comaneshter, D.; Cohen, A.D. Atopic Dermatitis
and the Metabolic Syndrome: A Cross-Sectional Study of 116,816 Patients. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1762–1767.
[CrossRef] [PubMed]
15. Mintoff, D.; Benhadou, F.; Pace, N.P.; Frew, J.W. Metabolic Syndrome and Hidradenitis Suppurativa: Epidemiological, Molecular,
and Therapeutic Aspects. Int. J. Dermatol. 2022, 61, 1175–1186. [CrossRef] [PubMed]
16. Akbaş, A.; Kılınç, F.; Şener, S.; Hayran, Y. Investigation of the Relationship Between Seborrheic Dermatitis and Metabolic
Syndrome Parameters. J. Cosmet. Dermatol. 2022, 21, 6079–6085. [CrossRef] [PubMed]
17. İmamoğlu, B.; Hayta, S.B.; Guner, R.; Akyol, M.; Özçelik, S. Metabolic Syndrome May Be an Important Comorbidity in Patients
with Seborrheic Dermatitis. Arch. Med. Sci. Atheroscler. Dis. 2016, 1, e158–e161. [CrossRef] [PubMed]
18. Sodagar, S.; Ghane, Y.; Heidari, A.; Heidari, N.; Khodadust, E.; Ahmadi, S.A.Y.; Seirafianpour, F.; Baradaran, H.; Goodarzi, A.
Association Between Metabolic Syndrome and Prevalent Skin Diseases: A Systematic Review and Meta-Analysis of Case-Control
Studies. Health Sci. Rep. 2023, 6, e1576. [CrossRef] [PubMed]
19. Galicia-Garcia, U.; Benito-Vicente, A.; Jebari, S.; Larrea-Sebal, A.; Siddiqi, H.; Uribe, K.B.; Ostolaza, H.; Martín, C. Pathophysiology
of Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2020, 21, 6275. [CrossRef] [PubMed]
20. Guo, H.; Wu, H.; Li, Z. The Pathogenesis of Diabetes. Int. J. Mol. Sci. 2023, 24, 6978. [CrossRef]
21. Twarda-Clapa, A.; Olczak, A.; Białkowska, A.M.; Koziołkiewicz, M. Advanced Glycation End-Products (AGEs): Formation,
Chemistry, Classification, Receptors, and Diseases Related to AGEs. Cells 2022, 11, 1312. [CrossRef]
22. Radziszewski, M.; Galus, R.; Łuszczyński, K.; Winiarski, S.; Wasowski,
˛ D.; Malejczyk, J.; Włodarski, P.; Ścieżyńska, A. The RAGE
Pathway in Skin Pathology Development: A Comprehensive Review of Its Role and Therapeutic Potential. Int. J. Mol. Sci. 2024,
25, 13570. [CrossRef] [PubMed]
23. Theodorakis, N.; Kreouzi, M.; Hitas, C.; Anagnostou, D.; Nikolaou, M. Adipokines and Cardiometabolic Heart Failure with
Preserved Ejection Fraction: A State-of-the-Art Review. Diagnostics 2024, 14, 2677. [CrossRef] [PubMed]
24. Theodorakis, N.; Kreouzi, M.; Pappas, A.; Nikolaou, M. Beyond Calories: Individual Metabolic and Hormonal Adaptations
Driving Variability in Weight Management—A State-of-the-Art Narrative Review. Int. J. Mol. Sci. 2024, 25, 13438. [CrossRef]
[PubMed]
25. Gjermeni, E.; Kirstein, A.S.; Kolbig, F.; Kirchhof, M.; Bundalian, L.; Katzmann, J.L.; Laufs, U.; Blüher, M.; Garten, A.; Le Duc,
D. Obesity–An Update on the Basic Pathophysiology and Review of Recent Therapeutic Advances. Biomolecules 2021, 11, 1426.
[CrossRef]
26. Thomaz, F.S.; John, O.D.; Sinha, P.; Shafie, S.R.; Worrall, S. The Metabolic Syndrome: An Overview and Proposed Mechanisms.
Obesities 2024, 4, 226–255. [CrossRef]
27. Fahed, G.; Aoun, L.; Bou Zerdan, M.; Allam, S.; Bou Zerdan, M.; Bouferraa, Y.; Assi, H.I. Metabolic Syndrome: Updates on
Pathophysiology and Management in 2021. Int. J. Mol. Sci. 2022, 23, 786. [CrossRef] [PubMed]
28. Glatthardt, T.; Lima, R.D.; de Mattos, R.M.; Ferreira, R.B.R. Microbe Interactions within the Skin Microbiome. Antibiotics 2024, 13,
49. [CrossRef]
29. Belkaid, Y.; Tamoutounour, S. The Influence of Skin Microorganisms on Cutaneous Immunity. Nat. Rev. Immunol. 2016, 16,
353–366. [CrossRef]
30. Ryguła, I.; Pikiewicz, W.; Grabarek, B.O.; Wójcik, M.; Kaminiów, K. The Role of the Gut Microbiome and Microbial Dysbiosis in
Common Skin Diseases. Int. J. Mol. Sci. 2024, 25, 1984. [CrossRef]
31. Howard, B.; Bascom, C.C.; Hu, P.; Binder, R.L.; Fadayel, G.; Huggins, T.G.; Jarrold, B.B.; Osborne, R.; Rocchetta, H.L.; Swift,
D.; et al. Aging-Associated Changes in the Adult Human Skin Microbiome and the Host Factors That Affect Skin Microbiome
Composition. J. Investig. Dermatol. 2022, 142, 1934–1946.e21. [CrossRef]
32. Mallott, E.K.; Sitarik, A.R.; Leve, L.D.; Cioffi, C.; Camargo, C.A., Jr.; Hasegawa, K.; Bordenstein, S.R. Human Microbiome Variation
Associated with Race and Ethnicity Emerges as Early as 3 Months of Age. PLoS Biol. 2023, 21, e3002230. [CrossRef]
33. Qu, L.; Matz, A.J.; Karlinsey, K.; Cao, Z.; Vella, A.T.; Zhou, B. Macrophages at the Crossroad of Meta-Inflammation and
Inflammaging. Genes 2022, 13, 2074. [CrossRef] [PubMed]
34. Sindhu, S.; Ahmad, R. Metabolic Inflammation and Cellular Immunity. Cells 2023, 12, 1615. [CrossRef] [PubMed]
35. Baker, P.; Huang, C.; Radi, R.; Moll, S.B.; Jules, E.; Arbiser, J.L. Skin Barrier Function: The Interplay of Physical, Chemical, and
Immunologic Properties. Cells 2023, 12, 2745. [CrossRef] [PubMed]
36. Yang, G.; Seok, J.K.; Kang, H.C.; Cho, Y.-Y.; Lee, H.S.; Lee, J.Y. Skin Barrier Abnormalities and Immune Dysfunction in Atopic
Dermatitis. Int. J. Mol. Sci. 2020, 21, 2867. [CrossRef]
37. Yan, L.; Wang, W.; Qiu, Y.; Yu, C.; Wang, R.; Li, C. Role of Glucose Metabolism Reprogramming in Keratinocytes in the Link
Between Psoriasis and Metabolic Syndrome. Int. Immunopharmacol. 2024, 139, 112704. [CrossRef] [PubMed]
Microorganisms 2025, 13, 161 30 of 32
38. Popa, A.D.; Gherasim, A.; Caba, L.; Nit, ă, O.; Graur, M.; Mihalache, L.; Arhire, L.I. Cathelicidin: Insights into Its Impact on
Metabolic Syndrome and Chronic Inflammation. Metabolites 2024, 14, 672. [CrossRef] [PubMed]
39. Niedźwiedzka, A.; Micallef, M.P.; Biazzo, M.; Podrini, C. The Role of the Skin Microbiome in Acne: Challenges and Future
Therapeutic Opportunities. Int. J. Mol. Sci. 2024, 25, 11422. [CrossRef]
40. Galizia, G.; Belloni Fortina, A.; Semenzato, A. Seborrheic Dermatitis: From Microbiome and Skin Barrier Involvement to Emerging
Approaches in Dermocosmetic Treatment. Cosmetics 2024, 11, 208. [CrossRef]
41. Liu, Q.; Ranallo, R.; Rios, C.; Grice, E.A.; Moon, K.; Gallo, R.L. Crosstalk between Skin Microbiota and Immune System in Health
and Disease. Nat. Immunol. 2023, 24, 895–898. [CrossRef]
42. Zhang, X.E.; Zheng, P.; Ye, S.Z.; Ma, X.; Liu, E.; Pang, Y.B.; He, Q.Y.; Zhang, Y.X.; Li, W.Q.; Zeng, J.H.; et al. Microbiome: Role in
Inflammatory Skin Diseases. J. Inflamm. Res. 2024, 17, 1057–1082. [CrossRef]
43. Treml, B.; Kleinsasser, A.; Stadlbauer, K.H.; Steiner, I.; Pajk, W.; Pilch, M.; Burtscher, M.; Knotzer, H. Cutaneous Microvascular
Blood Flow and Reactivity in Hypoxia. Front. Physiol. 2018, 9, 160. [CrossRef] [PubMed]
44. Dascălu, R.C.; Bărbulescu, A.L.; Stoica, L.E.; Dinescu, S, .C.; Bit, ă, C.E.; Popoviciu, H.V.; Ionescu, R.A.; Vreju, F.A. Review: A
Contemporary, Multifaceted Insight into Psoriasis Pathogenesis. J. Pers. Med. 2024, 14, 535. [CrossRef] [PubMed]
45. Man, A.-M.; Orăsan, M.S.; Hoteiuc, O.-A.; Olănescu-Vaida-Voevod, M.-C.; Mocan, T. Inflammation and Psoriasis: A Comprehen-
sive Review. Int. J. Mol. Sci. 2023, 24, 16095. [CrossRef] [PubMed]
46. Radaschin, D.S.; Tatu, A.; Iancu, A.V.; Beiu, C.; Popa, L.G. The Contribution of the Skin Microbiome to Psoriasis Pathogenesis and
Its Implications for Therapeutic Strategies. Medicina 2024, 60, 1619. [CrossRef] [PubMed]
47. Nowowiejska, J.; Baran, A.; Flisiak, I. Aberrations in Lipid Expression and Metabolism in Psoriasis. Int. J. Mol. Sci. 2021, 22, 6561.
[CrossRef] [PubMed]
48. Rudramurthy, S.M.; Honnavar, P.; Chakrabarti, A.; Dogra, S.; Singh, P.; Handa, S. Association of Malassezia Species with Psoriatic
Lesions. Mycoses 2014, 57, 483–488. [CrossRef] [PubMed]
49. Maurelli, M.; Gisondi, P.; Girolomoni, G. Advanced Glycation End Products and Psoriasis. Vaccines 2023, 11, 617. [CrossRef]
50. Karunakaran, U.; Elumalai, S.; Moon, J.-S.; Won, K.-C. CD36 Signal Transduction in Metabolic Diseases: Novel Insights and
Therapeutic Targeting. Cells 2021, 10, 1833. [CrossRef]
51. Haran, K.; Johnson, C.E.; Smith, P.; Venable, Z.; Kranyak, A.; Bhutani, T.; Jeon, C.; Liao, W. Impact of GLP-1 Receptor Agonists on
Psoriasis and Cardiovascular Comorbidities: A Narrative Review. Psoriasis 2024, 14, 143–152. [CrossRef] [PubMed]
52. Kumar, A.; Green, K.M.; Rawat, M. A Comprehensive Overview of Postbiotics with a Special Focus on Discovery Techniques and
Clinical Applications. Foods 2024, 13, 2937. [CrossRef] [PubMed]
53. Natarelli, N.; Gahoonia, N.; Sivamani, R.K. Bacteriophages and the Microbiome in Dermatology: The Role of the Phageome and a
Potential Therapeutic Strategy. Int. J. Mol. Sci. 2023, 24, 2695. [CrossRef] [PubMed]
54. Pareek, A.; Kumari, L.; Pareek, A.; Chaudhary, S.; Ratan, Y.; Janmeda, P.; Chuturgoon, S.; Chuturgoon, A. Unraveling Atopic
Dermatitis: Insights into Pathophysiology, Therapeutic Advances, and Future Perspectives. Cells 2024, 13, 425. [CrossRef]
[PubMed]
55. Fania, L.; Moretta, G.; Antonelli, F.; Scala, E.; Abeni, D.; Albanesi, C.; Madonna, S. Multiple Roles for Cytokines in Atopic
Dermatitis: From Pathogenic Mediators to Endotype-Specific Biomarkers to Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 2684.
[CrossRef] [PubMed]
56. Braun, C.; Patra, V.; Lina, G.; Nicolas, J.F.; Vocanson, M.; Nosbaum, A. The Role of Skin Dysbiosis in Atopic Dermatitis. Eur. J.
Dermatol. 2022, 32, 439–444. [CrossRef]
57. Nowowiejska, J.; Baran, A.; Flisiak, I. Lipid Alterations and Metabolism Disturbances in Selected Inflammatory Skin Diseases. Int.
J. Mol. Sci. 2023, 24, 7053. [CrossRef] [PubMed]
58. Alam, M.J.; Xie, L.; Yap, Y.-A.; Marques, F.Z.; Robert, R. Manipulating Microbiota to Treat Atopic Dermatitis: Functions and
Therapies. Pathogens 2022, 11, 642. [CrossRef] [PubMed]
59. Kim, H.J.; Kim, Y.H. Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies. Int. J. Mol. Sci.
2024, 25, 5302. [CrossRef] [PubMed]
60. Gruszczyńska, M.; Sadowska-Przytocka, A.; Szybiak, W.; Wi˛eckowska, B.; Lacka, K. Insulin Resistance in Patients with Acne
Vulgaris. Biomedicines 2023, 11, 2294. [CrossRef]
61. Hasrat, N.H.; Al-Yassen, A.Q. The Relationship Between Acne Vulgaris and Insulin Resistance. Cureus 2023, 15, e34241. [CrossRef]
[PubMed]
62. Taieb, A.; Feryel, A. Deciphering the Role of Androgen in the Dermatologic Manifestations of Polycystic Ovary Syndrome
Patients: A State-of-the-Art Review. Diagnostics 2024, 14, 2578. [CrossRef] [PubMed]
63. Del Rosso, J.Q.; Kircik, L. The Primary Role of Sebum in the Pathophysiology of Acne Vulgaris and Its Therapeutic Relevance in
Acne Management. J. Dermatolog. Treat. 2024, 35, 2296855. [CrossRef]
64. Biagi, L.G.; Sañudo, A.; Bagatin, E. Severe Acne and Metabolic Syndrome: A Possible Correlation. Dermatology 2019, 235, 456–462.
[CrossRef] [PubMed]
Microorganisms 2025, 13, 161 31 of 32
65. Andreadi, A.; Muscoli, S.; Tajmir, R.; Meloni, M.; Minasi, A.; Muscoli, C.; Ilari, S.; Mollace, V.; Della Morte, D.; Bellia, A.; et al.
Insulin Resistance and Acne: The Role of Metformin as Alternative Therapy in Men. Pharmaceuticals 2023, 16, 27. [CrossRef]
[PubMed]
66. Szefler, L.; Szybiak-Skora, W.; Sadowska-Przytocka, A.; Zaba, R.; Wieckowska, B.; Lacka, K. Metformin Therapy for Acne Vulgaris:
A Meta-Analysis. Pharmaceuticals 2024, 17, 728. [CrossRef] [PubMed]
67. Dessinioti, C.; Katsambas, A. The Microbiome and Acne: Perspectives for Treatment. Dermatol. Ther. 2024, 14, 31–44. [CrossRef]
68. Maden, S. Rosacea: An Overview of Its Etiological Factors, Pathogenesis, Classification, and Therapy Options. Dermato 2023, 3,
241–262. [CrossRef]
69. Woo, Y.R.; Lee, S.H.; Cho, S.H.; Lee, J.D.; Kim, H.S. Characterization and Analysis of the Skin Microbiota in Rosacea: Impact of
Systemic Antibiotics. J. Clin. Med. 2020, 9, 185. [CrossRef] [PubMed]
70. Chudzicka-Strugała, I.; Goł˛ebiewska, I.; Brudecki, G.; Elamin, W.; Zwoździak, B. Demodicosis in Different Age Groups and
Alternative Treatment Options—A Review. J. Clin. Med. 2023, 12, 1649. [CrossRef]
71. Habeebuddin, M.; Karnati, R.K.; Shiroorkar, P.N.; Nagaraja, S.; Asdaq, S.M.B.; Khalid Anwer, M.; Fattepur, S. Topical Probiotics:
More Than a Skin Deep. Pharmaceutics 2022, 14, 557. [CrossRef]
72. Medgyesi, B.; Dajnoki, Z.; Béke, G.; Gáspár, K.; Szabó, I.L.; Janka, E.A.; Póliska, S.; Hendrik, Z.; Méhes, G.; Törőcsik, D.; et al.
Rosacea Is Characterized by a Profoundly Diminished Skin Barrier. J. Investig. Dermatol. 2020, 140, 1938–1950.e5. [CrossRef]
73. Akin Belli, A.; Ozbas Gok, S.; Akbaba, G.; Etgu, F.; Dogan, G. The Relationship Between Rosacea and Insulin Resistance and
Metabolic Syndrome. Eur. J. Dermatol. 2016, 26, 260–264. [CrossRef] [PubMed]
74. Chang, C.H.; Chovatiya, R. More Yeast, More Problems? Reevaluating the Role of Malassezia in Seborrheic Dermatitis. Arch.
Dermatol. Res. 2024, 316, 100. [CrossRef]
75. Adalsteinsson, J.A.; Kaushik, S.; Muzumdar, S.; Guttman-Yassky, E.; Ungar, J. An Update on the Microbiology, Immunology, and
Genetics of Seborrheic Dermatitis. Exp. Dermatol. 2020, 29, 481–489. [CrossRef] [PubMed]
76. Sobhan, M.; Gholampoor, G.; Firozian, F.; Mohammadi, Y.; Mehrpooya, M. Comparison of Efficacy and Safety of Atorvastatin 5%
Lotion and Betamethasone 0.1% Lotion in the Treatment of Scalp Seborrheic Dermatitis. Clin. Cosmet. Investig. Dermatol. 2019, 12,
267–275. [CrossRef] [PubMed]
77. Truglio, M.; Sivori, F.; Cavallo, I.; Abril, E.; Licursi, V.; Fabrizio, G.; Cardinali, G.; Pignatti, M.; Toma, L.; Valensise, F.; et al.
Modulating the Skin Mycobiome-Bacteriome and Treating Seborrheic Dermatitis with a Probiotic-Enriched Oily Suspension. Sci.
Rep. 2024, 14, 2722. [CrossRef] [PubMed]
78. Polak, K.; Jobbágy, A.; Muszyński, T.; Wojciechowska, K.; Fratczak,
˛ A.; Bánvölgyi, A.; Bergler-Czop, B.; Kiss, N. Microbiome
Modulation as a Therapeutic Approach in Chronic Skin Diseases. Biomedicines 2021, 9, 1436. [CrossRef] [PubMed]
79. Diaz, M.J.; Aflatooni, S.; Abdi, P.; Li, R.; Anthony, M.R.; Neelam, S.; Farkouh, C.; Tran, J.T.; Svoboda, S.; Forouzandeh, M.; et al.
Hidradenitis Suppurativa: Molecular Etiology, Pathophysiology, and Management—A Systematic Review. Curr. Issues Mol. Biol.
2023, 45, 4400–4415. [CrossRef]
80. Abu Rached, N.; Gambichler, T.; Dietrich, J.W.; Ocker, L.; Seifert, C.; Stockfleth, E.; Bechara, F.G. The Role of Hormones in
Hidradenitis Suppurativa: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 15250. [CrossRef]
81. Świerczewska, Z.; Lewandowski, M.; Surowiecka, A.; Barańska-Rybak, W. Microbiome in Hidradenitis Suppurativa—What We
Know and Where We Are Heading. Int. J. Mol. Sci. 2022, 23, 11280. [CrossRef] [PubMed]
82. Krajewski, P.K.; Złotowska, A.; Szepietowski, J.C. The Therapeutic Potential of GLP-1 Receptor Agonists in the Management
of Hidradenitis Suppurativa: A Systematic Review of Anti-Inflammatory and Metabolic Effects. J. Clin. Med. 2024, 13, 6292.
[CrossRef] [PubMed]
83. Theodorakis, N.; Feretzakis, G.; Hitas, C.; Kreouzi, M.; Kalantzi, S.; Spyridaki, A.; Boufeas, I.Z.; Sakagianni, A.; Paxinou, E.;
Verykios, V.S.; et al. Antibiotic Resistance in the Elderly: Mechanisms, Risk Factors, and Solutions. Microorganisms 2024, 12, 1978.
[CrossRef]
84. Garg, A.; Neuren, E.; Cha, D.; Kirby, J.S.; Ingram, J.R.; Jemec, G.B.E.; Esmann, S.; Thorlacius, L.; Villumsen, B.; Marmol, V.D.; et al.
Evaluating Patients’ Unmet Needs in Hidradenitis Suppurativa: Results from the Global Survey of Impact and Healthcare Needs
(VOICE) Project. J. Am. Acad. Dermatol. 2020, 82, 366–376. [CrossRef]
85. Theodorakis, N.; Feretzakis, G.; Hitas, C.; Kreouzi, M.; Kalantzi, S.; Spyridaki, A.; Kollia, Z.; Verykios, V.S.; Nikolaou, M.
Immunosenescence: How Aging Increases Susceptibility to Bacterial Infections and Virulence Factors. Microorganisms 2024, 12,
2052. [CrossRef] [PubMed]
86. Da Silva Vale, A.; de Melo Pereira, G.V.; de Oliveira, A.C.; de Carvalho Neto, D.P.; Herrmann, L.W.; Karp, S.G.; Soccol, V.T.;
Soccol, C.R. Production, Formulation, and Application of Postbiotics in the Treatment of Skin Conditions. Fermentation 2023, 9,
264. [CrossRef]
87. Bousbaine, D.; Bauman, K.D.; Chen, Y.E.; Lalgudi, P.V.; Nguyen, T.T.D.; Swenson, J.M.; Yu, V.K.; Tsang, E.; Conlan, S.; Li, D.B.;
et al. Discovery and Engineering of the Antibody Response to a Prominent Skin Commensal. Nature 2024. [CrossRef] [PubMed]
Microorganisms 2025, 13, 161 32 of 32
88. Gribonika, I.; Band, V.I.; Chi, L.; Perez-Chaparro, P.J.; Link, V.M.; Ansaldo, E.; Oguz, C.; Bousbaine, D.; Fischbach, M.A.; Belkaid,
Y. Skin Autonomous Antibody Production Regulates Host-Microbiota Interactions. Nature 2024. [CrossRef] [PubMed]
89. Hernández Medina, R.; Kutuzova, S.; Nielsen, K.N.; Johansen, J.; Hansen, L.H.; Nielsen, M.; Rasmussen, S. Machine Learning and
Deep Learning Applications in Microbiome Research. ISME Commun. 2022, 2, 98. [CrossRef]
90. Gilbert, J.A.; Quinn, R.A.; Debelius, J.; Xu, Z.Z.; Morton, J.; Garg, N.; Jansson, J.K.; Dorrestein, P.C.; Knight, R. Microbiome-Wide
Association Studies Link Dynamic Microbial Consortia to Disease. Nature 2016, 535, 94–103. [CrossRef] [PubMed]
91. Tan, I.J.; Podwojniak, A.; Parikh, A.; Cohen, B.A. Precision Dermatology: A Review of Molecular Biomarkers and Personalized
Therapies. Curr. Issues Mol. Biol. 2024, 46, 2975–2990. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.