Next Article in Journal
Characterization and Antioxidant Activity of Exopolysaccharides Produced by Lysobacter soyae sp. nov Isolated from the Root of Glycine max L.
Next Article in Special Issue
Oxygen Exposure and Tolerance Shapes the Cell Wall-Associated Lipids of the Skin Commensal Cutibacterium acnes
Previous Article in Journal
Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products

by
Keir Nicholas-Haizelden
1,
Barry Murphy
2,
Michael Hoptroff
2 and
Malcolm J. Horsburgh
1,*
1
Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
2
Unilever Research & Development, Port Sunlight, Wirral CH63 3JW, UK
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(8), 1899; https://doi.org/10.3390/microorganisms11081899
Submission received: 20 June 2023 / Revised: 20 July 2023 / Accepted: 25 July 2023 / Published: 27 July 2023
(This article belongs to the Special Issue Human Skin Microbiota 2.0)

Abstract

:
Bioprospecting is the discovery and exploration of biological diversity found within organisms, genetic elements or produced compounds with prospective commercial or therapeutic applications. The human skin is an ecological niche which harbours a rich and compositional diversity microbiome stemming from the multifactorial interactions between the host and microbiota facilitated by exploitable effector compounds. Advances in the understanding of microbial colonisation mechanisms alongside species and strain interactions have revealed a novel chemical and biological understanding which displays applicative potential. Studies elucidating the organismal interfaces and concomitant understanding of the central processes of skin biology have begun to unravel a potential wealth of molecules which can exploited for their proposed functions. A variety of skin-microbiome-derived compounds display prospective therapeutic applications, ranging from antioncogenic agents relevant in skin cancer therapy to treatment strategies for antimicrobial-resistant bacterial and fungal infections. Considerable opportunities have emerged for the translation to personal care products, such as topical agents to mitigate various skin conditions such as acne and eczema. Adjacent compound developments have focused on cosmetic applications such as reducing skin ageing and its associated changes to skin properties and the microbiome. The skin microbiome contains a wealth of prospective compounds with therapeutic and commercial applications; however, considerable work is required for the translation of in vitro findings to relevant in vivo models to ensure translatability.

1. Introduction

Despite perceptions of being an arid wasteland, the human skin is a habitat occupied by a diverse array of microorganisms. Human skin microbiome diversity stems from microenvironment variations between body sites ranging from the Cutibacterium-dominated sebaceous regions, rich in lipids, to the halotolerant Staphylococcus that are abundant in sweat-rich areas [1,2]. The complement of these microorganisms, the microbiota, utilises host compounds as nutrient sources whilst conveying benefits to their host, such as pathogenic colonisation resistance, local immune system priming and the maintenance of lipid barriers [3]. Akin to macroecological biomes, the skin microbiota community structure consists of organisms specialised to corresponding niches that are susceptible to invasion by foreign species displacing resident organisms [2]. The composition and stability of the skin microbiome are constrained by physical factors that include pH, oxygen concentration, nutrient availability and competition, plus host symbiotic and commensal relationships [4]. Interactions between host microbiota are largely mediated by secreted biological effector compounds, such as digestive enzymes, antimicrobials and quorum sensing compounds [5]. The diversity of skin microbiota and associated effector compounds represent an underappreciated source of effectors potentially relevant to the treatment of a variety of diseases and conditions.
Bioprospecting is the discovery and exploration of the biological diversity of organisms, genetic components or compounds with potential commercial or therapeutic applications. The utility of beneficial biologically derived compounds is highlighted by the discovery of antibiotics, which has revolutionised modern healthcare and drastically increased global lifespan [6]. Novel drug discovery remains necessary for improving the financial viability of therapeutic and commercial compound pipelines and healthcare outcomes [7]. There is an overwhelming need for novel treatments for bacterial infections in a contemporary world where antimicrobial resistant (AMR) pathogens are an increasing concern, with over 1 million deaths in 2019 associated with such infections [8]. Several alternative treatment strategies are in development to combat AMR infections, such as bacteriophage therapy that has recently gained momentum in Western countries [9].
The skin microbiome provides a vast resource for the development of novel treatment strategies towards the control of AMR infections. Understanding the biochemistry at the interface between host and microbiota will unravel hitherto unknown biology. The coevolution of the human skin with pathogens has resulted in control mechanisms, such as sapienic acid and psoriasin capable of the growth inhibition of AMR Staphylococcus aureus and Escherichia coli, respectively [8,10,11]. The comparable gut microbiome has revealed a plethora of novel compounds with treatment potential for diseases such as chronic infection, cancer and obesity [12]. Whilst skin microbiota potentially harbours a greater species richness than the gut, its potential abundance of therapeutic compounds and their translational benefits are yet to be mined [3].
Advances in high-throughput genomic technologies have revealed a wealth of microbiome data, facilitating a conceptual shift whereby multicellular organisms’ functions are intrinsically linked to their microbiota [13]. Classically, microbiological investigations relied upon the direct culturing of isolates utilising historic techniques to infer organism identity. The true picture of microbiota diversity was confounded by fastidious organisms lacking the necessary growth conditions provided in vivo, such as complex metabolic interdependences [14,15]. Metagenomic techniques have facilitated more interrogative analyses of the total genomic material present within the skin, which continue to reveal ever more diversity within the skin microbiota [16,17,18,19]. Further development of artificial intelligence technologies has enabled a sophisticated analysis of metagenomic and physical data, allowing for inferences in the causal linkage of skin hydration, age and smoking status to microbiome composition and interpersonal variation [16].
Investigations utilising next-generation omics technologies have revealed a multitude of mechanisms and concepts from the skin microbiome. The human skin microbiome is a complex and dynamic field with varied applications, and many organisms and associated compounds have feasibly exploitable properties [3,4,17,18]
This review is distinct through a focus on the breadth and recent advances in the therapeutic and commercial application of biological molecules gained from bioprospecting the human skin microbiome. We further explore the viability of prospective compounds for their translational capacity in human application.

2. Skin Environment

The external skin represents a surface functioning primarily to minimise transepidermal evaporative water loss whilst simultaneously providing a physical barrier to extrinsic biological and environmental agents and regulating body temperature [19]. Structurally, skin is comprised of three layers: the innermost hypodermis which stores adipose fat for physical protection and energy, the connective dermis, containing blood and nerve vessels, and the external epidermis [20]. The epidermis is composed of a stratified network of keratinocytes which originate from lower levels and migrate through the epidermis eventually forming the outer stratum corneum of dead anucleate corneocytes [19]. Eventually, corneocytes desquamate and are released from the keratin and lipid matrix, facilitating skin renewal whilst providing nutrition for resident skin microorganisms (Figure 1) [4,21].
Human skin displays localised variability in secretion content, local topography and thickness. Oily skin areas, such as the forehead, contain an increased abundance of sebaceous glands which release sebum onto the skin through the hair follicle canal and holocrine secretions [22,23]. Sebum is composed of a mixture of nonpolar lipids, such as wax esters, squalene and triglycerides, with the latter lipolysed into free fatty acids by resident microbiota [24,25,26,27,28,29,30]. Sebum-derived fatty acids including sapienic acid maintain skin pH and exert a broad-spectrum antimicrobial action modulating skin microbiota composition. Community structure disruption is linked to inflammatory skin conditions like atopic dermatitis [31,32,33]. Epidermal lipids secreted directly by keratinocytes filling in areas between squamous corneocytes act directly to minimise transepidermal evaporative water loss [34].
Perspiration occurs across nearly all bodily regions via sweat glands secreting saline-rich liquid onto the skin to facilitate evaporative thermoregulation [35,36]. The regional variation in human skin sites is further evidenced through variations in thickness, invaginations, moisture content and hair follicle density, among other factors (Figure 1), [2].
Figure 1. Structure and microbial diversity of healthy human skin at distinct body areas. The microhabitats of the scalp, axilla (armpit), forearm and plantar heel are related to distinct secretion profiles associated with epidermal lipids, sebum and sweat. Lipid- and sebum-rich sites are associated with an increased proportion of lipophilic microorganism genera, namely, Cutibacterium and Malassezia. Sweat-rich regions display an abundance of halotolerant genera such as Staphylococcus and Corynebacterium [1,37]. Pie charts represent the abundance of bacterial and fungal genera at corresponding body sites adapted from several studies. Additionally, sites such as the plantar heel display a high degree of diversity across individuals [2,38,39]. Created with www.BioRender.com (accessed on 4 April 2023).
Figure 1. Structure and microbial diversity of healthy human skin at distinct body areas. The microhabitats of the scalp, axilla (armpit), forearm and plantar heel are related to distinct secretion profiles associated with epidermal lipids, sebum and sweat. Lipid- and sebum-rich sites are associated with an increased proportion of lipophilic microorganism genera, namely, Cutibacterium and Malassezia. Sweat-rich regions display an abundance of halotolerant genera such as Staphylococcus and Corynebacterium [1,37]. Pie charts represent the abundance of bacterial and fungal genera at corresponding body sites adapted from several studies. Additionally, sites such as the plantar heel display a high degree of diversity across individuals [2,38,39]. Created with www.BioRender.com (accessed on 4 April 2023).
Microorganisms 11 01899 g001

3. Drivers of Skin Microbiome Community Structure

The human skin microbiome is governed by fundamental ecological principles relating to habitat, species interaction and community disruption. The skin environment contains an abundance of biological niches that vary in factors, including topography, lipid content, pH, salinity and moisture content alongside temporal changes from ageing and gender [16,40,41]. Human skin is occupied by very diverse populations of bacteria, archaea, fungi, viruses and protists, with colonisation requiring specialisation and adaptation due to niche-specific microenvironments [42,43]. Resident skin microorganisms coevolved with competitors and their hosts, producing complex nutritional interdependencies and behavioural characteristics such as diverse antimicrobial biosynthesis [2,4,44]. Given the range and variation in skin environments alongside differences in external factors, microorganisms are under a variety of abiotic and biotic selective pressures driving variation in stable community structures (Figure 2), [3].

4. Pathogenic Invasion and Community Disruption

The skin microbiome is susceptible to invasion by foreign organisms capable of disrupting the community equilibrium. Many human skin pathogens are generally considered commensal, with their opportunistic pathogenic behaviour evident when introduced into a new subsurface niche through environmental disturbance, such as a laceration [46]. Commensals such as Staphylococcus epidermidis and Cutibacterium acnes provide host benefit through nutritional competition with pathogens, stimulating host antimicrobial peptide secretion and dampening nonpathogenic local immune system activation [47,48,49]. Yet S. epidermidis and C. acnes represent common aetiological agents of nosocomial infections of immunocompromised patients, generally following medical device implantation and prosthetic joint surgery [4,50]. Staphylococcus aureus asymptomatic colonisation of the anterior nares is often causative of future systemic infection, a threat compounded by the feasibility of AMR strains competing with susceptible strains for dominance [51]. The wide range of skin-relevant pathogens has been discussed in detail elsewhere [52,53,54]. Current antibiotic treatment strategies are often ineffective given the recalcitrant nature of skin-pathogen-associated biofilms alongside the prevalence and continued dissemination of multiple antibiotic resistance elements [55,56,57]. Mechanisms by which microorganisms interact directly influence the microbiota community structure, hence these mechanisms can be manipulated as a source of therapeutically and commercially relevant compounds.

5. Therapeutically Relevant Skin-Microbiome-Derived Compounds

5.1. Bacteriocins

Bacteriocins are a range of bacterial-derived peptides and proteins capable of exerting bacteriostatic and bactericidal activity. Bacteriocins act as competitive factors to inhibit the growth of adjacent organisms, akin to antibiotics, yet are associated with distinct resistance mechanisms and thus therapeutically relevant [58]. Diversity in the structure of bacteriocins has evolved with variations in their molecular targets and breadth of host range [59,60,61].
In contrast to broad-spectrum antibiotics, bacteriocin-based treatments could be tailored to the clearance of specific pathogenic organisms with minimal disruption to natural flora [62,63,64,65]. Hence, they are relevant in the treatment of nosocomial infections which are linked to unstable patient microbiomes and increased colonisation by reservoirs of highly antimicrobial-resistant pathogenic bacteria in the healthcare environment [65,66].
Bacteriocins have been identified as effector agents of bacterial antagonism, directing research to exploit their commercial and therapeutic activity [67]. Commercial applications of bacteriocins have focused primarily on the preservation of meat, vegetables and dairy products through the prevention of the bacterial degradation of food [68]. To date, Nisin derived from the dairy fermentative lactic acid bacterium Lactococcus lactis remains the sole Food and Drug Administration-approved bacteriocin with licensing in food preservation [69]. The scope for the therapeutic use of bacteriocins is broad, ranging from infection treatments to oral hygiene products and spermicidal compounds. The therapeutic bacteriocin field has yet to fully target the potential of skin microbiome diversity, with studies needed to further the elucidation of the biosynthetic pathways, mechanisms of action and cost of production alongside tolerance in clinical trials [70,71,72].
Studies of the nasal microbiome identified that coagulase-negative commensal staphylococci produce bacteriocins active against niche competitors [73]. The skin and nasopharynx commensal S. lugdunensis synthesises lugdunin, which represents the first nonribosomal-produced antimicrobial identified within the human skin microbiome. Classified as a novel class of thiazolidine cyclic peptide bacteriocin termed fibupeptides, lugdunin inhibits the growth of several Gram-positive bacteria, including methicillin-resistant S. aureus (MRSA) [74,75,76]. Lugdunin is proposed to exert antimicrobial activity through the disruption of the transmembrane pH gradient likely leading to intracellular protein denaturation and reduced proton motive force, directly inhibiting respiration [74,77,78]. Lugdunin is further evidenced to increase the gene expression of several cutaneous antimicrobial peptides and induce the phagocytic recruitment of neutrophils and monocytes [75]. A key driver of lugdunin research is the low propensity for resistance development in exposed bacteria, with one study identifying S. aureus as not developing resistance after several subinhibitory passages [76].
The therapeutic translation of purified bacteriocins remains difficult due to several factors, such as serum half-life and immunogenicity. Protein engineering represents a promising area for redesigning bacteriocins towards their human use. An example comes from a study of the domesticated ruminant commensal Staphylococcus simulans that is associated with rare opportunistic infections in humans, such as endocarditis and erythema [79,80,81]. S. simulans secretes the bacteriocin lysostaphin, an endopeptidase enzyme capable of pentaglycine bridge cleavage within the peptidoglycan of staphylococci. Lysostaphin exerts anti-S. aureus bactericidal activity regardless of planktonic, quiescent and biofilm-associated lifestyles [82]. Lysostaphin immunogenicity represents a major barrier to clinical application. Protein engineering reduced lysostaphin immunogenicity through T-cell epitope removal alongside an increased serum half-life through polyethylene glycol and albumin conjugation [83,84,85]. Various lysostaphin application vehicles have been effective versus S. aureus in vivo via intravaginal dissolvable tablets, emulsion-based topical gels and intravenous injection.
The viricidal, virostatic and food-preservative applications of certain bacteriocins have further been reported. For example, the gut commensal and rare skin pathogen Enterococcus faecium produces the bacteriocin CRL35, capable of the considerable inhibition of herpes simplex virus (HSV) 1/2 through late-stage protein-synthesis retardation in vitro [86,87,88]. Antimicrobial properties of CRL35 have been demonstrated in vivo and been explored for use as a dairy food preservative via chitosan microencapsulation with a retainment of bactericidal activity [89]. Bacillus subtilis can be found on skin and its bacteriocin subtilosin, a macrocyclic lantibiotic, is capable of the inhibition of both HSV-1 and 2 late-stage replication in addition to several bacteria associated with urinary tract infections [90,91,92]. Further nonskin-bacteria-derived bacteriocins have potent antiviral activity, which highlights the scope for further investigation of the skin microbiome in this area [90].
Most skin-relevant bacteriocins remain to be explored in preclinical development stages to assess pharmacological benefits. Several considerations must be addressed prior to the therapeutic and commercial applications of bacteriocins, including toxicity, biological half-life and the capacity for resistance development [58]. Indeed, given the range of susceptibility spectrums for bacteriocins, there is a need to collect temporal data as a means to disfavour those with the potential to disrupt the skin microbiota community, where it may exacerbate or perpetuate inflammatory disorders [67].

5.2. Bacteriophages

The virome remains an underappreciated area of the skin microbiome, with attention mostly limited to bacteriophages. In natural environments, bacteriophages are considered to modulate bacterial communities through the proliferation inhibition of dominant species, thereby maintaining diversity via predator–prey coevolution [93]. Research of the species-rich gut virome has revealed 1010 bacteriophages per gram of faecal material, with variations indicating complex ecological interactions [94,95]. Skin metagenome analyses have highlighted the breadth of phages associated with the predominant skin bacterial genera Staphylococcus, Cutibacterium and Streptococcus [96,97]. This repertoire represents a rich source of potentially therapeutic bacteriophages.
A principal translational pathway for the therapeutic use of lytic bacteriophages is to clear pathogens from infected tissue [98]. Phages exert bactericidal activity with typical strain specificity for infection through the interaction with bacterial cell surface structures, such as lipopolysaccharide or teichoic acid [99,100]. The genetic manipulation of phage–bacteria determinant regions has been extensively studied, thereby improving the effectiveness of this strategy for the treatment of AMR infections [101,102,103].
Multiple bacteriophages are reportedly being explored for phage therapy for various manifestations of skin-associated pathogens. Support for clinical research comes from phage therapy success for skin infections of Mycobacterium chelonae and multiple-drug resistant (MDR) Pseudomonas aeruginosa [104,105]. Topical bacteriophage therapy targeting C. acnes has advanced to phase I clinical trials after displaying efficacy in reducing C. acnes abundance without significant safety concerns [106]. Screening studies isolated seven distinct bacteriophages effective against S. epidermidis albeit with highly variable strain-specific resistance profiles [107]. Combinations of S. epidermidis with S. aureus-specific phages were demonstrated to suppress phage-resistant mutants of S. aureus in a skin model of atopic dermatitis [108].
More than 10 clinical trials were in various stages of completion by early 2023 investigating the efficacy of bacteriophage therapy for the treatment of musculoskeletal, skin and soft tissue infections [109]. Previous trials supported the high safety of phage therapy with varying efficacy arising from delivery mechanisms and phage stability [110]. For example, PhagoBurn is a phage-treated wound dressing found safe yet ineffective in phase I/II clinical trials at reducing Escherichia coli and Pseudomonas aeruginosa burdens, likely through insufficient dosing. The foundation is established, however, for future phage-mediated skin infection trials using phage lysins, liposome-encapsulated phage cocktails and antibiotic phage combinatorial therapy [111].
A topical bacteriophage cocktail therapy for acne, BX001, demonstrated significant C. acnes abundance reduction in phase I cosmetic clinical trials. The evidence implicates the IA-1 C. acnes phylotype as an opportunistic pathogen of the pilosebaceous unit by stimulating a localised proinflammatory response leading to acne lesions, meaning such treatment has utility [112]. Direct modulation of skin microbiome composition through targeted phage therapy may yield novel skin treatment strategies from wound healing to eczema [108,113,114,115]. A sufficient knowledge of multiple phages with distinct cellular targets is required to mitigate the capacity for phage resistance alongside a physiochemical knowledge of product formulations slowing product development [116].
An appreciation of safety concerns is necessary for the responsible translation of bacteriophages into clinical use. Wildtype phages are classified in relation to their infection cycles, with temperate phages associated with prophage genomic integration and maintenance, whilst obligate lytic phages are associated with direct cellular lysis following infection [117,118]. Phage therapy focuses on obligate lytic phages to circumvent toxin production, integration and insertional mutagenesis [119]. Temperate phage insertions are experimentally evidenced in some cases to facilitate the mobilisation of pathogenicity islands and associated antibiotic resistance and superantigen genes in S. aureus, thereby limiting their translation by promoting virulence and treatment resistance [120,121]. Strategies to circumvent inherent phage limitations include the modification of phage DNA to encode CRISPR-Cas9 systems that resensitise resistant bacteria to antibiotic treatments or bactericidal toxins [122]. Bacteriophage endolysins targeting specific cell wall lysis are a promising alternative to hindrances associated with bacteriophage therapy by removing the replicative cycle of phages, albeit with higher production costs [123]. Endolysins available commercially include the eczema treatment Gladskin Micreobalance® with considerable mitigation of S. aureus-associated atopic dermatitis [124].

5.3. Cutaneous Lipids

Cutaneous fatty acids impart multiple functions for the host, namely, microbial growth inhibition through a reduction in skin pH alongside direct antimicrobial activity, whilst simultaneously promoting commensal adherence [2]. Skin fatty acids are produced through the hydrolysis of epidermal and sebaceous triglycerides, with activity predominantly facilitated through the extracellular lipase secretion of commensals, such as Cutibacterium spp. and Staphylococcus spp. [125,126,127]. Fatty acids further function as signalling molecules capable of diminishing proinflammatory cytokine induction in keratinocytes associated with allergic dermis reactions through free fatty acid receptor 1 activation [128]. Triglyceride degradation liberates glycerol, a potent humectant that supports corneocyte desquamation from corneodesosome cleavage, thereby stimulating the recovery of irritated skin [26,129,130]. The microbial fermentation of glycerol with the production of fatty acids, namely lactic acid, promotes an acidic pH and the upregulated gene expression of essential skin barrier proteins in keratinocytes [131]. Topical formulations of certain skin lipids have the potential for therapeutic use due to their range of activity and antimicrobial activity. Sphingosine and sapienic acid show potent activity towards staphylococci [11,31,132]. Skin lipids represent an avenue for therapeutic application, and studies indicate a potential use for sapienic acid as a stable antimicrobial in cosmetic products [31,132,133].

5.4. Biofilm Inhibitors

Microorganisms aggregate into biofilms through the secretion of matrix proteins, polysaccharides and DNA, with the structures considered a dominant growth mechanism by members of the skin microbiome [134,135]. Biofilms convey many benefits to their community with decreases in both immune system stimulation and antimicrobial uptake through metabolic dormancy [136]. Many opportunistic skin pathogens form monomicrobial biofilms, e.g., S. aureus, C. acnes and S. epidermidis. Notably, S. aureus biofilms are prevalent in atopic dermatitis, with a correlation between abundance and disease severity [137]. C. acnes and S. epidermidis biofilms are associated with medical-implant-associated infection, often leading to systemic bacteraemia in immunocompromised individuals [138,139,140]. Given the clinical relevance of biofilm-associated infection, novel therapeutics are necessary to circumvent the treatment resistance of these bacterial aggregates.
Resident skin flora exerts antagonistic interactions to limit biofilm formation and integrity through the production of antibiofilm agents. S. epidermidis secretes a serine protease, Esp, evidenced to disassemble MRSA biofilms by degrading several proteins for cell wall and biofilm formation. Esp can degrade human receptors utilised by S. aureus for adherence, such as fibronectin to convey colonisation resistance [141,142,143]. Moreover, a S. epidermidis protease-independent biofilm inhibitor is effective against a range of MRSA and methicillin-susceptible S. aureus (MSSA) without reducing cell viability. This uncharacterised biofilm inhibitor was proposed to be a phenol-soluble modulin (PSM) likely functioning through the inhibition of the polysaccharide adhesion operon [144,145,146].
The lipophilic, aerotolerant commensal Cutibacterium acnes can reside in pilosebaceous units, and its secreted lipases hydrolyse triglycerides, releasing short-chain fatty acids (SCFAs) resulting in local pH reduction and the inhibition of competing bacteria, stabilising niche occupation [147]. Additional beneficial functions of C. acnes include the secretion of antioxidant enzymes and in vivo antitumoral properties [49,148]. However, sebum-derived SCFAs may also induce proinflammatory gene expression in keratinocytes and sebocytes within the pilosebaceous unit that likely drive acne vulgaris manifestations [149,150]. SCFAs further inhibit the polysaccharide biofilm formation of S. epidermidis at physiological concentrations whilst simultaneously increasing susceptibility to both ampicillin and doxycycline [151]. SCFAs are being explored within the gut microbiome given their significant potential for inflammatory and immune diseases, with a likely relevance to cutaneous homeostasis [152,153,154].

5.5. Quorum Sensing Modulators

The survival of bacterial communities requires constant environmental adaptation to selective pressures [155]. Quorum sensing (QS) systems mediate cell-to-cell communication to bring about a coordinated change in gene expression producing a community beneficial behaviour [156]. QS relies upon the secretion of autoinducer compounds at subthreshold levels that diffuse into the extracellular environment where the autoinducer concentration is linked with bacterial population density. The post-threshold autoinducer activates signal transduction to a transcription factor(s) that drive(s) regulon expression [157,158,159,160].
QS products such as extracellular proteases have a metabolic expense but provide community level fitness benefits, hence collaborative production acts to limit individual cost [161]. Many QS-mediated products are virulence factors that facilitate processes such as biofilm formation, integrity promotion and dispersal associated with systemic disease progression [160,162]. In recent years, analogous quorum-sensing-like systems were identified in fungi and bacteriophages, highlighting the significance of microbial communication in microbiota present on the skin [163,164].
The secretion of QS inhibitors represents a form of antagonism common within the skin microbiome that has potential therapeutic translatability. Various commensal staphylococci, including S. epidermidis, S. hominis and S. simulans, secrete inhibitors of the S. aureus accessory gene regulator (agr) that interfere with colonisation and virulence [165,166]. S. hominis autoinducer peptides (AIPs) were shown to quench accessory gene regulatory (agr) QS systems of MRSA, with AIP-2 providing protection from S. aureus necrosis and skin damage [167]. Staphylococcus warneri produces AIP-1 and AIP-2 capable of the dose-dependent inhibition of S. epidermidis agr-1 and all agr of S. aureus, reducing virulence factor production and providing protection against associated skin barrier damage [168].
Since pathogenic bacteria often utilise QS to stimulate biofilm formation, many QS inhibitors have a translational application in the treatment of recalcitrant biofilm infections derived from the human skin [169,170]. The skin commensal Staphylococcus xylosus expresses an RNAIII-inhibiting peptide (RIP) that inhibits QS-associated signal transduction pathways. RIP is a potent inhibitor of S. aureus and S. epidermidis biofilm formation proteins [171,172,173]. Moreover, RIP treatment displayed both an in vitro inhibition of S. aureus adhesion and efficacy in the treatment of S. aureus murine infection models [173,174].

5.6. Fungicidal Compounds

The cutaneous mycobiome is less well studied as the fungal component that colonises human skin. The mycobiome becomes established following birth with compositional changes associated with extrinsic and intrinsic factors comparable with the microbiome as a whole. Skin fungal dysbiosis correlates with common cutaneous disorders, such as dandruff, atopic dermatitis and pityriasis versicolor [175,176,177]. Fungal pathogen emergence is underrecognised in contrast to the antimicrobial resistance of bacterial pathogens and viral pathogen resurgence [178,179]. Problematically, fungal infection treatment is generally limited to just four classes of compounds, which can often be unsuccessful due to resistance development that highlights the necessity of novel antifungal compounds [180,181].
The predominant genus of the human cutaneous mycobiome is Malassezia, which was recently identified to have a greater breadth of species from metagenomic studies, albeit species that remain uncultured [182]. Malassezia can produce indole compounds for nitrogen acquisition through tryptophan metabolism [183]. Malassezia indoles display broad-spectrum fungicidal properties, effective against pathogenic yeasts and moulds at skin-relevant concentrations in vitro [184]. Malassezia furfur produces several indoles, such as indolo [2,3] carbazole, which inhibits Candida spp. and other Malassezia spp. at concentrations less than 6 ug/mL [184,185]. Known ligands of aryl hydrocarbon receptors, these indoles can result in the induction of proinflammatory responses, drastically limiting their therapeutic prospects [186].
Several commensal bacteria inhibit Candida albicans colonisation within the cutaneous environment. S. epidermidis stimulates the production of specific CD8+ T cells via localised dendritic cell activation, resulting in an improved protection against Candida albicans infection through enhanced innate defence systems [187]. S. epidermidis stimulates the primary keratinocyte production of human antimicrobial peptides beta defensin 1–3 in a Toll-like receptor-2-associated manner [177,188]. Lactobacillus spp. that are abundant within the vaginal microbiome produce lactic acid and SCFA resulting in vaginal acidification that inhibits the C. albicans yeast–hyphae transition [189]. Such hyphal transition represents a key virulence strategy of C. albicans required for mucosal invasion and systemic dissemination [190]. Several postulated antifungal benefits of vaginal lactobacilli are related to colonisation resistance through strong adhesion and biofilm formation upon mucosal membranes and free-radical secretion increasing mucus cohesion [191]. Indeed, vaginal probiotics are commercialised with Lactobacillus-based Canesten Canesflor®, demonstrating the potential of developing probiotics for fungal infections. The antifungal potential of Saccharomyces cerevisiae and Candida spp. was demonstrated in vitro to inhibit biofilm formation and hyphal transition in C. albicans, highlighting the need for future probiotic research [192].

5.7. Skin Cancer Treatments

The development of novel cancer treatments represents an unequivocally imperative task for modern health science. The significance of cancer is highlighted through the rapid growth in global morbidity and mortality, representing a leading cause of premature deaths in an estimated 60% of countries as of 2021 [193,194].
Within cancerous tumours exists a distinct microenvironment, and the microbiome can alter both progression and treatment resistance. Multiple associations were identified between the prevalence of individual skin microbiome members and specific skin cancers, such as S. aureus within squamous cell carcinoma skin biopsies [195]. A causative relationship between skin microbiome members and skin cancers is proposed to occur through the induction of proinflammatory responses, which links to an apparent increased risk of skin cancer with skin-microbiome-associated disorders [196,197,198,199]. There is a necessity to investigate the intentional modification of skin tumour microbiomes to mitigate the risks associated with skin cancer development given the previously described rich source of bactericidal products [195]. The characterisation of the skin microbiome has so far revealed several compounds displaying potential antioncogenic activity capable of increasing the available therapies to clinical professionals.
The metabolic analysis of S. epidermidis skin isolates revealed the trait variable production of 6-N-hydroxyaminopurine (6-HAP), a hydroxylamine adenine analogue with mutagenic and teratogenic activity [200,201]. 6-HAP displayed an in vitro selective inhibition of lymphoma and melanoma tumour cell line proliferation whilst exerting relatively little effect on an epidermal keratinocyte cell line. S. epidermidis-producing 6-HAP is prevalent in the human skin microbiome, and these strains exerted a significant inhibition of ultraviolet-associated neoplasia generation in vivo [202]. The antioncogenic properties of 6-HAP have been questioned based on the characterisation of commercially sourced versus commensal-produced 6-HAP [203,204]. The therapeutic potential of 6-HAP is contingent on the further translation to in vivo models alongside the elucidation of its mechanism of action, prior to its application in cancer treatment and prevention.
Commensal Malassezia spp. synthesise indirubin, a compound capable of potent aryl hydrocarbon receptor (AHR) agonism, with a proposed competitive inhibition of cyclin-dependent kinases associated with cell cycle modulation, resulting in cell cycle arrest [205]. Further antioncogenic mechanisms of indirubin have been discussed in detail elsewhere [206]. Despite the apparent commensality of Malassezia spp., opportunistic pathogenesis is associated with several conditions, such as dermatitis, supporting the application of topical probiotic formulations, i.e., for treatment of skin cancers [207].
Bacteriocins represent an attractive source of antioncogenic compounds arising from their specificity, nontoxicity and abundance within the skin microbiome [208]. Pyocins (bacteriocins produced by the genus Pseudomonas) of the transient opportunistic pathogen Pseudomonas aeruginosa selectively inhibit the proliferation of human hepatoma, B lymphocyte and murine fibroblast cell lines [209,210]. The antioncogenic activity of bacteriocins produced by skin-relevant pathogens has been described, such as Klebsiella pneumoniae-derived microcin E492; however, the therapeutic potential of skin-microbiome-derived bacteriocins remains unexplored [208]. Moreover, investigations of the antioncogenic activity of bacteriocins are largely limited to in vitro cell lines, illustrating the necessity for in vivo studies and considerations of the reported limitations of the therapeutic utilisation of bacteriocins.
Many anticancer therapeutic compounds are limited by the development of resistance in target cells alongside nonselective cell targeting, resulting in multiple adverse side effects. The selective and distinct functional mechanisms of bacteriocins highlight their therapeutic application potential [209]. Several methods were discussed to feasibly increase bacteriocin therapeutic activity, for example, the conjugation to nanoparticles for synergistic drug delivery facilitating a lower therapeutic dosage [211]. Additionally, many common oral probiotic compounds are associated with the production of bacteriocins with considerable anticancer activity, highlighting the need for parallel investigations with topical probiotics or purified compounds [209].

6. Skin Microbiome Applications in Personal Care Products

Personal care products represent a considerable proportion of the global economy, with a total estimated market valuation of over USD 500 billion in 2022 and a predicted annual growth rate of over 6% [212]. Within this growth potential, the skin microbiome has many demonstratable applications for personal care product development and many areas for future investigations.
The composition and stability of an individual’s skin microbiota is paramount for the maintenance of cutaneous homeostasis through functions such as pathogenic colonisation resistance [2]. A growing body of evidence correlates skin microbiota composition with biophysical skin properties alongside the perpetuation and onset of skin conditions [16,40,213,214,215,216,217]. Hence, the reshaping of the skin microbiota community structure and utilisation of isolated effector compounds represent a significant area for commercial product development [218]. The viability of such products is shown by the prevalence of novel topical probiotics in the personal care market alongside the renowned success of parallel gut probiotics.

6.1. Acne Vulgaris

The key microbial component of acne pathophysiology is linked to the obstruction of the sebaceous gland leading to sebum accumulation, resulting in C. acnes extracellular lipases creating proinflammatory SCFAs [219,220,221]. Several clinical trials have demonstrated probiotics cause C. acnes inhibition leading to reduced acne severity. A topical mixture of E. faecalis and Lactobacillus sp. applied bidaily resulted in reductions in acne lesions, proposedly resultant of C. acnes inhibition and reduced proinflammatory factor production. Similarly, trials utilising aqueous Lactobacillus plantarum probiotics demonstrated dosage-dependent reductions in lesion size [222]. To date, these trials have had a small cohort of less than 100 people, inviting larger studies to support the confidence and reliability of such products [223,224]. Skin probiotics capable of reducing acne manifestations include products recently released to the market, such as those from Yun probiotherapy, indicating the growth of such probiotic markets [225].

6.2. Atopic Dermatitis

Atopic dermatitis (AD) is the most common inflammatory skin disorder globally and is associated with pruritic and desiccated inflamed lesions, arising from various inheritable mutations in skin barrier proteins and allergen-specific immunological defects [226,227]. AD cutaneous dysbiosis is characterised by an increased abundance of staphylococci such as S. aureus and Malassezia spp. which perpetuates AD manifestation and its severity through the secretion of virulence factors (e.g., toxins) and cytokine-inducing nanovesicles, respectively [228,229,230]. Nakatsuji et al. identified that in a small cohort study, the transplantation of specific antimicrobial peptide-producing S. epidermidis and S. hominis strains yielded considerable reductions in S. aureus abundance in AD patients [228]. Such promise means further investigations with a larger cohort size and an understanding of the longer-term persistence of transplanted strains with the effect of repeat applications is necessary to further unlock their clear potential. The effects of bacterial extracts of Vitreoscilla filiformis, Streptococcus thermophilus and Lactobacillus johnsonii have yielded a promising mitigation of AD severity, likely through an antagonistic action upon S. aureus and Malassezia [231].

6.3. Anti-Ageing

Skin ageing corresponds with physiological changes altering elasticity, thickness and moisture. These changes are linked to multiple biological processes, including mutation accumulation, cellular senescence and microbiome dysbiosis [232]. Physiological manifestations of cutaneous ageing are accelerated by environmental factors, such as UV-mediated photodamage [233]. The composition of the skin microbiome varies throughout life, with aged skin having increased overall diversity. Ageing is further correlated to increased antimicrobial production and physiological reductions in the production of cutaneous factors, notably reduced collagen and sebum [40,234].
Correlations between ageing and cutaneous skin microbiome changes are well established. Elderly skin microbiomes are associated with consistently reduced Cutibacterium spp. and Lactobacillus spp. across multiple bodily sites. [40,235]. A conjectural association with reduced C. acnes abundance is reduced glycerol, fatty acids and antioxidant production perpetuating physiological changes associated with cutaneous ageing [131,236]. Streptococcus spp. abundance increases until puberty and correlates with younger biophysical properties, such as high elasticity [235]. The exogenous treatment of human fibroblasts with a supernatant from facial-skin-swab-derived strains of Streptococcus sp. resulted in an increased gene expression of collagen, filaggrin and lipid synthesis proteins [237]. The causative compound within Streptococcus supernatants is accepted to be spermidine, a polyamine capable of inducing cytoprotective autophagy associated with an increased turnover of cells, proteins and organelles. Spermidine is further associated with chemotherapy potentiation and tumorigenesis suppression in murine models [238,239]. Moreover, spermidine levels are shown to decrease with biological ageing, further supporting its role in ageing, and this polyamine displays low toxicity in mice and humans [240]. The supplementation of spermidine or the promotion of producing streptococci in naturally deficient older humans may provide improved phenotypes associated with biological ageing and warrant further investigation.
Wide-ranging prospects of personal care treatments aimed to mitigate physiological alterations associated with ageing are feasible. For example, Lactobacillus spp. correlate with reduced photo-ageing through an ultraviolet protective effect, thus reducing collagen degradation [17]. Further, C. acnes RoxP is a free-radical oxygenase capable of potent antioxidant activity relevant for the mitigation of cutaneous ageing processes in purified or pre- and probiotic formulations [236,241,242].

6.4. Skin Rejuvenation

Topical probiotics produced with the aim of establishing stable communities of beneficial microorganisms represent a key direction for skin-microbiome-derived development. S. epidermidis may contribute to the production of ceramide on human skin through sphingomyelinase production leading to the digestion of antimicrobial amino alcohol sphingosines [132,243] S. epidermidis sphingomyelinase is sufficient to significantly increase murine ceramide content on skin and conveys no apparent pathology associated with keratinocyte cytolysis or biofilm formation [244]. Ceramides provide a diverse and vital range of functions through forming a major component of the lipid barrier to the regulation of keratinocyte proliferation and the modulation of localised immune responses [244,245]. The further potential of streptococci producing hyaluronic acid and streptococcal lysates increasing skin ceramide production highlights potential avenues that might direct skin care product development [246,247,248]. Sphingomyelinase expression appears to provide growth advantages for S. epidermidis by facilitating colonisation through nutrient acquisition and lipid osmoprotection [249]. Indeed, a correlative relationship was reported with reduced spingomyelinase activity in atopic dermatitis, highlighting a further direction for product development [250]. The exploration into the commercial viability of products utilising purified sphingomyelinase, lysate or probiotic formulations derived from strains with optimal activity is warranted.
Lactobacillus spp. were explored as topical cutaneous probiotics [225,251]. Lactobacilli are evidenced in primary human keratinocytes to aid skin rejuvenation through stimulating the expression of skin-junction proteins via Toll-like receptor 2 activation, thus promoting the integrity of the lipid barrier. Similarly, Lactobacillus ferment lysate stimulated keratinocyte migration leading to increased skin repair; however, the direct effector compounds are unknown [252].

6.5. Moisturisers

Topical moisturisers are a staple in the cosmetic industry, aiming to hydrate the skin to visually improve skin smoothness through restoration of the skin lipid barrier [253]. Since glycerol represents a potent humectant facilitating skin moisture and hydration retention, cosmetic products that aim to increase glycerol liberation from sebaceous triglycerides are a feasible option [131,254]. Hence, formulations containing either commensal-derived extracellular lipases or those that promote producer growth may yield moisturising potential. Both prebiotic and postbiotic moisturisers are on the market, with one study identifying visual improvements in skin moisture and increased skin microbiome diversity over the 4-week period studied [131,213,255]. Moisturisers containing probiotic lysates are currently commercially available containing Lactobacillus spp. and generally aim to stimulate the expression and production of skin barrier proteins as reported from in vivo and in vitro models [217,256,257].

6.6. Cutaneous Hyperpigmentation

The superficial skin infection termed pityriasis versicolor is caused by several Malassezia spp. and can result in hypopigmented and hyperpigmented macules [258]. Notably, the Malassezia sp.-produced indole derivative compound malassezin can induce melanocyte apoptosis through aryl hydrocarbon receptor agonist activity [259,260,261,262]. This activity means malassezin was explored as a novel cosmetic treatment for facial hyperpigmentation and produces clear decreases in melanin and visible skin lightening within two to four weeks following oil–water emulsion skin treatment [261,263].

6.7. Rosacea

The chronic inflammatory skin disorder rosacea is characterised by persistent facial flushing, sensitive dry skin and inflamed cutaneous plaques and is associated with genetic, neurological and immune system dysregulation. Correlations between rosacea and microbial dysbiosis were identified with an overgrowth of S. epidermidis, Bacillus oleronius and reduced C. acnes growth [217]. However, only one known study with one participant has utilised a topical probiotic and low-dose doxycycline combinatory therapy, albeit with apparent effective results 6 months post treatment [264]; the study signposts a future scope for treatment.

7. Future Perspectives

The human skin microbiome contains a wealth of compounds that can be exploited for therapeutic and personal care applications. Further research by both the public and private sectors will bring skin-microbiome-derived products to the market and importantly elucidate mechanisms of action, efficacy and safety. Simultaneously, improvements to known compounds through protein engineering that aid translation represent another promising avenue for the application of skin-microbiome-derived compounds. Such products have the capability to improve healthcare outcomes for a variety of disorders as well as improve the quality of life for a significant portion of the population.
A wealth of knowledge exists pertaining to the skin microbiome representing an incredibly well-characterised human–microbiome association. There is a plethora of commercial and therapeutic compounds evidenced to modulate the skin microbiome to provide holistic benefits. Yet many investigations of the skin microbiome are reliant on correlative studies and deductions from in vitro investigations on bacterial isolates. Hence, many microbiome investigations are only able to reveal rudimentary relationships between a limited number of species, which may not be translatable in vivo [265]. There is a necessity for the development of models more closely reflecting the natural skin environment and the diversity associated with physiological factors. Several skin models have been developed, such as stem-cell-derived reconstructed human epidermis, skin explants and NativeSkin®, albeit associated with a high cost and short-lived stability [266].
The advent of next-generation sequencing technologies has unequivocally transformed microbiome research. Robust metagenomic pipelines have proved pivotal for the untargeted creation of detailed compositional maps of the microbiome, facilitating procedures such as nosocomial MDR pathogen surveillance [267]. Metagenomics provides only a limited picture of the microbiome, with investigations unable to confidently identify rare species or nonviable cells [16]. Indeed, microbiome investigations are increasingly reliant on multiomics approaches facilitating revelations of nuanced microbial behaviours with potential human translatability [268].

8. Conclusions

The rapid growth in the development of therapeutic and commercial skin-microbiome-derived and probiotic products illustrates the enormous potential of the field. The skin microbiome contains a vast source of cosmetic or therapeutically relevant compounds which have been shown to be translatable for the treatment of many diseases from antimicrobial-resistant pathogen infections to cancers. Many applications further exist for commercial exploitation in the personal care sector, with the potential for the development of novel cosmetic drug formulations. Similarly, the intentional modulation of the native skin flora composition through topical pre and probiotics may alleviate symptoms of many skin disorders through the promotion of microorganisms associated with healthy individuals. Skin microbiome research has recently displayed a surge in interest, yet considerable developments in areas such as in vivo modelling are required to ensure coherency and translatability to the human skin environment.

Author Contributions

K.N.-H. writing and original draft preparation, visualisation; B.M., writing, review and editing; M.H., writing, review and editing; M.J.H. conceptualisation, writing, review and editing; All authors have read and agreed to the published version of the manuscript.

Funding

K.N.-H. is funded by BBSRC Collaborative Training Partnership grant BB/X511481/1.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Jordan Thomas for the critical review of the manuscript.

Conflicts of Interest

BM and MH are employed by Unilever Plc.

References

  1. Carmona-Cruz, S.; Orozco-Covarrubias, L.; Sáez-de-Ocariz, M. The Human Skin Microbiome in Selected Cutaneous Diseases. Front. Cell Infect. Microbiol. 2022, 12, 834135. [Google Scholar] [CrossRef] [PubMed]
  2. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The Human Skin Microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef]
  3. Boxberger, M.; Cenizo, V.; Cassir, N.; La Scola, B. Challenges in Exploring and Manipulating the Human Skin Microbiome. Microbiome 2021, 9, 1–14. [Google Scholar] [CrossRef]
  4. Swaney, M.H.; Kalan, L.R. Living in Your Skin: Microbes, Molecules, and Mechanisms. Infect. Immun. 2021, 89, e00695-20. [Google Scholar] [CrossRef] [PubMed]
  5. Gorter, F.A.; Manhart, M.; Ackermann, M. Understanding the Evolution of Interspecies Interactions in Microbial Communities. Philos. Trans. R. Soc. B 2020, 375, 20190256. [Google Scholar] [CrossRef] [Green Version]
  6. Hutchings, M.; Truman, A.; Wilkinson, B. Antibiotics: Past, Present and Future. Curr. Opin. Microbiol. 2019, 51, 72–80. [Google Scholar] [CrossRef] [PubMed]
  7. Mohs, R.C.; Greig, N.H. Drug Discovery and Development: Role of Basic Biological Research. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2017, 3, 651. [Google Scholar] [CrossRef]
  8. Thompson, T. The Staggering Death Toll of Drug-Resistant Bacteria. Nature 2022. [Google Scholar] [CrossRef]
  9. Strathdee, S.A.; Hatfull, G.F.; Mutalik, V.K.; Schooley, R.T. Phage Therapy: From Biological Mechanisms to Future Directions. Cell 2023, 186, 17–31. [Google Scholar] [CrossRef]
  10. Gläser, R.; Harder, J.; Lange, H.; Bartels, J.; Christophers, E.; Schröder, J.M. Antimicrobial Psoriasin (S100A7) Protects Human Skin from Escherichia coli Infection. Nat. Immunol. 2004, 6, 57–64. [Google Scholar] [CrossRef]
  11. Moran, J.C.; Alorabi, J.A.; Horsburgh, M.J. Comparative Transcriptomics Reveals Discrete Survival Responses of S. aureus and S. epidermidis to Sapienic Acid. Front. Microbiol. 2017, 8, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Anjana; Tiwari, S.K. Bacteriocin-Producing Probiotic Lactic Acid Bacteria in Controlling Dysbiosis of the Gut Microbiota. Front. Cell Infect. Microbiol. 2022, 12, 415. [Google Scholar] [CrossRef]
  13. Berg, G.; Rybakova, D.; Fischer, D.; Cernava, T.; Vergès, M.C.C.; Charles, T.; Chen, X.; Cocolin, L.; Eversole, K.; Corral, G.H.; et al. Microbiome Definition Re-Visited: Old Concepts and New Challenges. Microbiome 2020, 8, 1–22. [Google Scholar] [CrossRef]
  14. Arnold, J.W.; Roach, J.; Azcarate-Peril, M.A. Emerging Technologies for Gut Microbiome Research. Trends Microbiol. 2016, 24, 887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kong, H.H.; Segre, J.A. Skin Microbiome: Looking Back to Move Forward. J. Investig. Dermatol. 2012, 132, 933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Carrieri, A.P.; Haiminen, N.; Maudsley-Barton, S.; Gardiner, L.J.; Murphy, B.; Mayes, A.E.; Paterson, S.; Grimshaw, S.; Winn, M.; Shand, C.; et al. Explainable AI Reveals Changes in Skin Microbiome Composition Linked to Phenotypic Differences. Sci. Rep. 2021, 11, 4565. [Google Scholar] [CrossRef]
  17. Yu, J.; Ma, X.; Wang, X.; Cui, X.; Ding, K.; Wang, S.; Han, C. Application and Mechanism of Probiotics in Skin Care: A Review. J. Cosmet. Dermatol. 2022, 21, 886–894. [Google Scholar] [CrossRef]
  18. Grice, E.A.; Segre, J.A. The Skin Microbiome. Nat. Rev. Microbiol. 2011, 9, 244. [Google Scholar] [CrossRef] [PubMed]
  19. Yousef, H.; Alhajj, M.; Sharma, S. Anatomy, Skin (Integument), Epidermis; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK470464/ (accessed on 19 June 2023).
  20. Kim, J.Y.; Dao, H. Physiology, Integument; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK554386/ (accessed on 19 June 2023).
  21. Meisel, J.S.; Sfyroera, G.; Bartow-McKenney, C.; Gimblet, C.; Bugayev, J.; Horwinski, J.; Kim, B.; Brestoff, J.R.; Tyldsley, A.S.; Zheng, Q.; et al. Commensal Microbiota Modulate Gene Expression in the Skin. Microbiome 2018, 6, 1–15. [Google Scholar] [CrossRef]
  22. Fischer, H.; Fumicz, J.; Rossiter, H.; Napirei, M.; Buchberger, M.; Tschachler, E.; Eckhart, L. Holocrine Secretion of Sebum Is a Unique DNase2-Dependent Mode of Programmed Cell Death. J. Investig. Dermatol. 2017, 137, 587–594. [Google Scholar] [CrossRef] [Green Version]
  23. Atsugi, T.; Yokouchi, M.; Hirano, T.; Hirabayashi, A.; Nagai, T.; Ohyama, M.; Abe, T.; Kaneko, M.; Zouboulis, C.C.; Amagai, M.; et al. Holocrine Secretion Occurs Outside the Tight Junction Barrier in Multicellular Glands: Lessons from Claudin-1-Deficient Mice. J. Investig. Dermatol. 2020, 140, 298–308.e5. [Google Scholar] [CrossRef] [PubMed]
  24. Freinkel, R.K. The Origin of Free Fatty Acids in Sebum. I. Role of Coagulase Negative Staphylococci. J. Investig. Dermatol. 1968, 50, 186–188. [Google Scholar] [CrossRef] [Green Version]
  25. Lacey, R.W. Loss of the Antibacterial Action of Skin After Topical Neomycin. Br. J. Dermatol. 1969, 81, 435–439. [Google Scholar] [CrossRef]
  26. Marples, R.R.; Kligman, A.M.; Lantis, L.R.; Downing, D.T. The Role of the Aerobic Microflora in the Genesis of Fatty Acids in Human Surface Lipids. J. Investig. Dermatol. 1970, 55, 173–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. McGinley, K.J.; Webster, G.F.; Ruggieri, M.R.; Leyden, J.J. Regional Variations in Density of Cutaneous Propionibacteria: Correlation of Propionibacterium acnes Populations with Sebaceous Secretion. J. Clin. Microbiol. 1980, 12, 672–675. [Google Scholar] [CrossRef] [PubMed]
  28. Nicolaides, N.; Wells, G.C. On the Biogenesis of the Free Fatty Acids in Human Skin Surface Fat. J. Investig. Dermatol. 1957, 29, 423–433. [Google Scholar] [CrossRef] [Green Version]
  29. Shalita, A.R. Genesis of Free Fatty Acids. J. Investig. Dermatol. 1974, 62, 332–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Stewart, M.E.; Downing, D.T.; Pochi, P.E.; Strauss, J.S. The Fatty Acids of Human Sebaceous Gland Phosphatidylcholine. Biochim. Biophys. Acta (BBA)-Lipids Lipid Metab. 1978, 529, 380–386. [Google Scholar] [CrossRef]
  31. Takigawa, H.; Nakagawa, H.; Kuzukawa, M.; Mori, H.; Imokawa, G. Deficient Production of Hexadecenoic Acid in the Skin Is Associated in Part with the Vulnerability of Atopic Dermatitis Patients to Colonization by Staphylococcus aureus. Dermatology 2005, 211, 240–248. [Google Scholar] [CrossRef]
  32. Lovászi, M.; Szegedi, A.; Zouboulis, C.C.; Törőcsik, D. Sebaceous-Immunobiology Is Orchestrated by Sebum Lipids. Derm.-Endocrinol. 2017, 9, e1375636. [Google Scholar] [CrossRef] [Green Version]
  33. Makrantonaki, E.; Ganceviciene, R.; Zouboulis, C. An Update on the Role of the Sebaceous Gland in the Pathogenesis of Acne. Derm.-Endocrinol. 2011, 3, 41. [Google Scholar] [CrossRef] [Green Version]
  34. Vietri Rudan, M.; Watt, F.M. Mammalian Epidermis: A Compendium of Lipid Functionality. Front. Physiol. 2022, 12, 2444. [Google Scholar] [CrossRef] [PubMed]
  35. Mark, H.; Harding, C.R. Amino Acid Composition, Including Key Derivatives of Eccrine Sweat: Potential Biomarkers of Certain Atopic Skin Conditions. Int. J. Cosmet. Sci. 2013, 35, 163–168. [Google Scholar] [CrossRef] [PubMed]
  36. Murota, H.; Matsui, S.; Ono, E.; Kijima, A.; Kikuta, J.; Ishii, M.; Katayama, I. Sweat, the Driving Force behind Normal Skin: An Emerging Perspective on Functional Biology and Regulatory Mechanisms. J. Dermatol. Sci. 2015, 77, 3–10. [Google Scholar] [CrossRef] [PubMed]
  37. Belvoncikova, P.; Splichalova, P.; Videnska, P.; Gardlik, R. The Human Mycobiome: Colonization, Composition and the Role in Health and Disease. J. Fungi 2022, 8, 1046. [Google Scholar] [CrossRef]
  38. Findley, K.; Oh, J.; Yang, J.; Conlan, S.; Deming, C.; Meyer, J.A.; Schoenfeld, D.; Nomicos, E.; Park, M.; Becker, J.; et al. Topographic diversity of fungal and bacterial communities in human skin. Nature 2013, 498, 367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Gao, Z.; Perez-Perez, G.I.; Chen, Y.; Blaser, M.J. Quantitation of Major Human Cutaneous Bacterial and Fungal Populations. J. Clin. Microbiol. 2010, 48, 3575. [Google Scholar] [CrossRef] [Green Version]
  40. 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. [Google Scholar] [CrossRef]
  41. Flowers, L.; Grice, E.A. The Skin Microbiota: Balancing Risk and Reward. Cell Host Microbe 2020, 28, 190. [Google Scholar] [CrossRef]
  42. Sanford, J.A.; Gallo, R.L. Functions of the Skin Microbiota in Health and Disease. Semin. Immunol. 2013, 25, 370. [Google Scholar] [CrossRef] [Green Version]
  43. Kobayashi, T.; Voisin, B.; Kim, D.Y.; Kennedy, E.A.; Jo, J.H.; Shih, H.Y.; Truong, A.; Doebel, T.; Sakamoto, K.; Cui, C.Y.; et al. Homeostatic Control of Sebaceous Glands by Innate Lymphoid Cells Regulates Commensal Bacteria Equilibrium. Cell 2019, 176, 982. [Google Scholar] [CrossRef] [Green Version]
  44. Swaney, M.H.; Sandstrom, S.; Kalan, L.R. Cobamide Sharing Is Predicted in the Human Skin Microbiome. mSystems 2022, 7, e00677-22. [Google Scholar] [CrossRef] [PubMed]
  45. Skowron, K.; Bauza-kaszewska, J.; Kraszewska, Z.; Wiktorczyk-kapischke, N.; Grudlewska-buda, K.; Kwiecińska-piróg, J.; Wałecka-zacharska, E.; Radtke, L.; Gospodarek-komkowska, E. Human Skin Microbiome: Impact of Intrinsic and Extrinsic Factors on Skin Microbiota. Microorganisms 2021, 9, 543. [Google Scholar] [CrossRef] [PubMed]
  46. Sachdeva, C.; Satyamoorthy, K.; Murali, T.S. Microbial Interplay in Skin and Chronic Wounds. Curr. Clin. Microbiol. Rep. 2022, 9, 21–31. [Google Scholar] [CrossRef]
  47. Nguyen, T.H.; Park, M.D.; Otto, M. Host Response to Staphylococcus epidermidis Colonization and Infections. Front. Cell Infect. Microbiol. 2017, 7, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Rozas, M.; de Ruijter, A.H.; Fabrega, M.J.; Zorgani, A.; Guell, M.; Paetzold, B.; Brillet, F. From Dysbiosis to Healthy Skin: Major Contributions of Cutibacterium acnes to Skin Homeostasis. Microorganisms 2021, 9, 628. [Google Scholar] [CrossRef] [PubMed]
  49. Fournière, M.; Latire, T.; Souak, D.; Feuilloley, M.G.J.; Bedoux, G. Staphylococcus epidermidis and Cutibacterium acnes: Two Major Sentinels of Skin Microbiota and the Influence of Cosmetics. Microorganisms 2020, 8, 1752. [Google Scholar] [CrossRef]
  50. Tabaja, H.; Tai, D.B.G.; Beam, E.; Abdel, M.P.; Tande, A.J. Clinical Profile of Monomicrobial Corynebacterium Hip and Knee Periprosthetic Joint Infections. Open Forum Infect. Dis. 2022, 9, ofac193. [Google Scholar] [CrossRef]
  51. Sakr, A.; Brégeon, F.; Mège, J.L.; Rolain, J.M.; Blin, O. Staphylococcus aureus Nasal Colonization: An Update on Mechanisms, Epidemiology, Risk Factors, and Subsequent Infections. Front. Microbiol. 2018, 9, 2419. [Google Scholar] [CrossRef]
  52. Clebak, K.T.; Malone, M.A. Skin Infections. Prim. Care 2018, 45, 433–454. [Google Scholar] [CrossRef]
  53. Gunaydin, S.D.; Arikan-Akdagli, S.; Akova, M. Fungal Infections of the Skin and Soft Tissue. Curr. Opin. Infect. Dis. 2020, 33, 130–136. [Google Scholar] [CrossRef] [PubMed]
  54. Marques, S.A.; Abbade, L.P.F. Severe Bacterial Skin Infections. An. Bras. Dermatol. 2020, 95, 407–417. [Google Scholar] [CrossRef] [PubMed]
  55. Murray, C.J.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Robles Aguilar, G.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E.; et al. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef] [PubMed]
  56. Jo, J.H.; Harkins, C.P.; Schwardt, N.H.; Portillo, J.A.; Zimmerman, M.D.; Carter, C.L.; Hossen, M.A.; Peer, C.J.; Polley, E.C.; Dartois, V.; et al. Alterations of Human Skin Microbiome and Expansion of Antimicrobial Resistance after Systemic Antibiotics. Sci. Transl. Med. 2021, 13, eabd8077. [Google Scholar] [CrossRef]
  57. Wozniak, J.M.; Mills, R.H.; Olson, J.; Caldera, J.R.; Sepich-Poore, G.D.; Carrillo-Terrazas, M.; Tsai, C.M.; Vargas, F.; Knight, R.; Dorrestein, P.C.; et al. Mortality Risk Profiling of Staphylococcus aureus Bacteremia by Multi-Omic Serum Analysis Reveals Early Predictive and Pathogenic Signatures. Cell 2020, 182, 1311–1327.e14. [Google Scholar] [CrossRef]
  58. Benítez-Chao, D.F.; León-Buitimea, A.; Lerma-Escalera, J.A.; Morones-Ramírez, J.R. Bacteriocins: An Overview of Antimicrobial, Toxicity, and Biosafety Assessment by in Vivo Models. Front. Microbiol. 2021, 12, 677. [Google Scholar] [CrossRef]
  59. Darbandi, A.; Asadi, A.; Mahdizade Ari, M.; Ohadi, E.; Talebi, M.; Halaj Zadeh, M.; Darb Emamie, A.; Ghanavati, R.; Kakanj, M. Bacteriocins: Properties and Potential Use as Antimicrobials. J. Clin. Lab. Anal. 2022, 36, e24093. [Google Scholar] [CrossRef]
  60. Telhig, S.; Ben Said, L.; Zirah, S.; Fliss, I.; Rebuffat, S. Bacteriocins to Thwart Bacterial Resistance in Gram Negative Bacteria. Front. Microbiol. 2020, 11, 2807. [Google Scholar] [CrossRef]
  61. Acedo, J.Z.; Chiorean, S.; Vederas, J.C.; van Belkum, M.J. The Expanding Structural Variety among Bacteriocins from Gram-Positive Bacteria. FEMS Microbiol. Rev. 2018, 42, 805–828. [Google Scholar] [CrossRef]
  62. Grada, A.; Bunick, C.G. Spectrum of Antibiotic Activity and Its Relevance to the Microbiome. JAMA Netw. Open 2021, 4, e215357. [Google Scholar] [CrossRef]
  63. Strati, F.; Pujolassos, M.; Burrello, C.; Giuffrè, M.R.; Lattanzi, G.; Caprioli, F.; Troisi, J.; Facciotti, F. Antibiotic-Associated Dysbiosis Affects the Ability of the Gut Microbiota to Control Intestinal Inflammation upon Fecal Microbiota Transplantation in Experimental Colitis Models. Microbiome 2021, 9, 1–15. [Google Scholar] [CrossRef] [PubMed]
  64. Mills, S.; Ross, R.P.; Hill, C. Bacteriocins and Bacteriophage; a Narrow-Minded Approach to Food and Gut Microbiology. FEMS Microbiol. Rev. 2017, 41, S129–S153. [Google Scholar] [CrossRef]
  65. Mirande, C.; Bizine, I.; Giannetti, A.; Picot, N.; van Belkum, A. Epidemiological Aspects of Healthcare-Associated Infections and Microbial Genomics. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 823–831. [Google Scholar] [CrossRef]
  66. Tozzo, P.; Delicati, A.; Caenazzo, L. Human Microbiome and Microbiota Identification for Preventing and Controlling Healthcare-Associated Infections: A Systematic Review. Front. Public Health 2022, 10, 989496. [Google Scholar] [CrossRef] [PubMed]
  67. Simons, A.; Alhanout, K.; Duval, R.E. Bacteriocins, Antimicrobial Peptides from Bacterial Origin: Overview of Their Biology and Their Impact against Multidrug-Resistant Bacteria. Microorganisms 2020, 8, 639. [Google Scholar] [CrossRef] [PubMed]
  68. Raj, T.; Chandrasekhar, K.; Kumar, A.N.; Kim, S.H. Recent Biotechnological Trends in Lactic Acid Bacterial Fermentation for Food Processing Industries. Syst. Microbiol. Biomanuf. 2021, 2, 14–40. [Google Scholar] [CrossRef]
  69. Lahiri, D.; Nag, M.; Dutta, B.; Sarkar, T.; Pati, S.; Basu, D.; Abdul Kari, Z.; Wei, L.S.; Smaoui, S.; Wen Goh, K.; et al. Bacteriocin: A Natural Approach for Food Safety and Food Security. Front. Bioeng. Biotechnol. 2022, 10, 1978. [Google Scholar] [CrossRef]
  70. Gallo, G.; Renzone, G.; Palazzotto, E.; Monciardini, P.; Arena, S.; Faddetta, T.; Giardina, A.; Alduina, R.; Weber, T.; Sangiorgi, F.; et al. Elucidating the Molecular Physiology of Lantibiotic NAI-107 Production in Microbispora ATCC-PTA-5024. BMC Genom. 2016, 17, 42. [Google Scholar] [CrossRef] [Green Version]
  71. O’Sullivan, J.N.; O’Connor, P.M.; Rea, M.C.; O’Sullivan, O.; Walsh, C.J.; Healy, B.; Mathur, H.; Field, D.; Hill, C.; Paul Ross, R. Nisin J, a Novel Natural Nisin Variant, Is Produced by Staphylococcus capitis Sourced from the Human Skin Microbiota. J. Bacteriol. 2020, 202, e00639-19. [Google Scholar] [CrossRef] [Green Version]
  72. Negash, A.W.; Tsehai, B.A. Current Applications of Bacteriocin. Int. J. Microbiol. 2020, 2020, 4374891. [Google Scholar] [CrossRef]
  73. Janek, D.; Zipperer, A.; Kulik, A.; Krismer, B.; Peschel, A. High Frequency and Diversity of Antimicrobial Activities Produced by Nasal Staphylococcus Strains against Bacterial Competitors. PLoS Pathog. 2016, 12, e1005812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Barber, C.C.; Zhang, W. Small Molecule Natural Products in Human Nasal/Oral Microbiota. J. Ind. Microbiol. Biotechnol. 2021, 48, 10. [Google Scholar] [CrossRef] [PubMed]
  75. Bitschar, K.; Sauer, B.; Focken, J.; Dehmer, H.; Moos, S.; Konnerth, M.; Schilling, N.A.; Grond, S.; Kalbacher, H.; Kurschus, F.C.; et al. Lugdunin Amplifies Innate Immune Responses in the Skin in Synergy with Host- and Microbiota-Derived Factors. Nat. Commun. 2019, 10, 2730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Krauss, S.; Zipperer, A.; Wirtz, S.; Saur, J.; Konnerth, M.C.; Heilbronner, S.; Torres Salazar, B.O.; Grond, S.; Krismer, B.; Peschel, A. Secretion of and Self-Resistance to the Novel Fibupeptide Antimicrobial Lugdunin by Distinct Abc Transporters in Staphylococcus lugdunensis. Antimicrob. Agents Chemother. 2021, 65, e01734-20. [Google Scholar] [CrossRef] [PubMed]
  77. Krulwich, T.A.; Sachs, G.; Padan, E. Molecular Aspects of Bacterial PH Sensing and Homeostasis. Nat. Rev. Microbiol. 2011, 9, 330. [Google Scholar] [CrossRef] [Green Version]
  78. Farha, M.A.; Verschoor, C.P.; Bowdish, D.; Brown, E.D. Collapsing the Proton Motive Force to Identify Synergistic Combinations against Staphylococcus aureus. Chem. Biol. 2013, 20, 1168–1178. [Google Scholar] [CrossRef] [Green Version]
  79. Shields, B.E.; Tschetter, A.J.; Wanat, K.A. Staphylococcus simulans: An Emerging Cutaneous Pathogen. JAAD Case Rep. 2016, 2, 428. [Google Scholar] [CrossRef]
  80. Power, N.; Calisti, G.; Price, F.; Watt, V.; Gamlin, W.; Dobson, L.E.; Ray, S.G. Staphylococcus simulans Endocarditis of Native Aortic and Mitral Valves. Case Report and Literature Review. Clin. Infect. Pract. 2020, 7–8, 100044. [Google Scholar] [CrossRef]
  81. Drobeniuc, A.; Traenkner, J.; Rebolledo, P.A.; Ghazaryan, V.; Rouphael, N. Staphylococcus simulans: A Rare Uropathogen. IDCases 2021, 25, e01202. [Google Scholar] [CrossRef]
  82. Jayakumar, J.; Kumar, V.A.; Biswas, L.; Biswas, R. Therapeutic Applications of Lysostaphin against Staphylococcus aureus. J. Appl. Microbiol. 2021, 131, 1072–1082. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, H.; Brooks, S.A.; Eszterhas, S.; Heim, S.; Li, L.; Xiong, Y.Q.; Fang, Y.; Kirsch, J.R.; Verma, D.; Bailey-Kellogg, C.; et al. Globally Deimmunized Lysostaphin Evades Human Immune Surveillance and Enables Highly Efficacious Repeat Dosing. Sci. Adv. 2020, 6, eabb9011. [Google Scholar] [CrossRef] [PubMed]
  84. Blazanovic, K.; Zhao, H.; Choi, Y.; Li, W.; Salvat, R.S.; Osipovitch, D.C.; Fields, J.; Moise, L.; Berwin, B.L.; Fiering, S.N.; et al. Structure-Based Redesign of Lysostaphin Yields Potent Antistaphylococcal Enzymes That Evade Immune Cell Surveillance. Mol. Ther. Methods Clin. Dev. 2015, 2, 15021. [Google Scholar] [CrossRef]
  85. Tossavainen, H.; Raulinaitis, V.; Kauppinen, L.; Pentikäinen, U.; Maaheimo, H.; Permi, P. Structural and Functional Insights into Lysostaphin-Substrate Interaction. Front. Mol. Biosci. 2018, 5, 60. [Google Scholar] [CrossRef] [PubMed]
  86. Wachsman, M.B.; Castilla, V.; De Ruiz Holgado, A.P.; De Torres, R.A.; Sesma, F.; Coto, C.E. Enterocin CRL35 Inhibits Late Stages of HSV-1 and HSV-2 Replication in Vitro. Antivir. Res. 2003, 58, 17–24. [Google Scholar] [CrossRef] [PubMed]
  87. Medina Amado, C.; Minahk, C.J.; Cilli, E.; Oliveira, R.G.; Dupuy, F.G. Bacteriocin Enterocin CRL35 Is a Modular Peptide That Induces Non-Bilayer States in Bacterial Model Membranes. Biochim. Biophys. Acta (BBA)-Biomembr. 2020, 1862, 183135. [Google Scholar] [CrossRef] [PubMed]
  88. Krawczyk, B.; Wityk, P.; Gałęcka, M.; Michalik, M. The Many Faces of Enterococcus spp.—Commensal, Probiotic and Opportunistic Pathogen. Microorganisms 2021, 9, 1900. [Google Scholar] [CrossRef]
  89. Farizano, J.V.; Díaz Vergara, L.I.; Masias, E.; Baillo, A.A.; Torino, M.I.; Fadda, S.; Vanden Braber, N.L.; Montenegro, M.A.; Saavedra, L.; Minahk, C. Biotechnological Use of Dairy By-Products for the Production and Microencapsulation of the Food Preservative Enterocin CRL35. FEMS Microbiol. Lett. 2022, 369, fnac033. [Google Scholar] [CrossRef]
  90. Huang, F.; Teng, K.; Liu, Y.; Cao, Y.; Wang, T.; Ma, C.; Zhang, J.; Zhong, J. Bacteriocins: Potential for Human Health. Oxid. Med. Cell Longev. 2021, 2021, 5518825. [Google Scholar] [CrossRef]
  91. Quintana, V.M.; Torres, N.I.; Wachsman, M.B.; Sinko, P.J.; Castilla, V.; Chikindas, M. Anti-Herpes Simplex Virus Type 2 Activity of the Antimicrobial Peptide Subtilosin. J. Appl. Microbiol. 2014, 117, 1253. [Google Scholar] [CrossRef] [Green Version]
  92. Alajlani, M.M. Characterization of Subtilosin Gene in Wild Type Bacillus spp. and Possible Physiological Role. Sci. Rep. 2022, 12, 10521. [Google Scholar] [CrossRef]
  93. Chevallereau, A.; Pons, B.J.; van Houte, S.; Westra, E.R. Interactions between Bacterial and Phage Communities in Natural Environments. Nat. Rev. Microbiol. 2021, 20, 49–62. [Google Scholar] [CrossRef] [PubMed]
  94. Cao, Z.; Sugimura, N.; Burgermeister, E.; Ebert, M.P.; Zuo, T.; Lan, P. The Gut Virome: A New Microbiome Component in Health and Disease. EBioMedicine 2022, 81, 104113. [Google Scholar] [CrossRef] [PubMed]
  95. Nishijima, S.; Nagata, N.; Kiguchi, Y.; Kojima, Y.; Miyoshi-Akiyama, T.; Kimura, M.; Ohsugi, M.; Ueki, K.; Oka, S.; Mizokami, M.; et al. Extensive Gut Virome Variation and Its Associations with Host and Environmental Factors in a Population-Level Cohort. Nat. Commun. 2022, 13, 5252. [Google Scholar] [CrossRef] [PubMed]
  96. Oh, J.; Byrd, A.L.; Park, M.; Kong, H.H.; Segre, J.A. Temporal Stability of the Human Skin Microbiome. Cell 2016, 165, 854–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Keen, E.C. A Century of Phage Research: Bacteriophages and the Shaping of Modern Biology. Bioessays 2015, 37, 6. [Google Scholar] [CrossRef] [Green Version]
  98. Kasman, L.M.; Porter, L.D. Bacteriophages. In Brenner’s Encyclopedia of Genetics, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 280–283. [Google Scholar] [CrossRef]
  99. Koskella, B.; Meaden, S. Understanding Bacteriophage Specificity in Natural Microbial Communities. Viruses 2013, 5, 806–823. [Google Scholar] [CrossRef] [Green Version]
  100. Kortright, K.E.; Chan, B.K.; Turner, P.E. High-Throughput Discovery of Phage Receptors Using Transposon Insertion Sequencing of Bacteria. Proc. Natl. Acad. Sci. USA 2020, 117, 18670–18679. [Google Scholar] [CrossRef]
  101. Dunne, M.; Prokhorov, N.S.; Loessner, M.J.; Leiman, P.G. Reprogramming Bacteriophage Host Range: Design Principles and Strategies for Engineering Receptor Binding Proteins This Review Comes from a Themed Issue on Nanobiotechnology-Phage Therapy. Curr. Opin. Biotechnol. 2021, 68, 272–281. [Google Scholar] [CrossRef]
  102. Chen, Y.; Batra, H.; Dong, J.; Chen, C.; Rao, V.B.; Tao, P. Genetic Engineering of Bacteriophages against Infectious Diseases. Front. Microbiol. 2019, 10, 954. [Google Scholar] [CrossRef]
  103. Yehl, K.; Lemire, S.; Yang, A.C.; Ando, H.; Mimee, M.; Torres, M.D.T.; de la Fuente-Nunez, C.; Lu, T.K. Engineering Phage Host-Range and Suppressing Bacterial Resistance through Phage Tail Fiber Mutagenesis. Cell 2019, 179, 459–469.e9. [Google Scholar] [CrossRef]
  104. Little, J.S.; Dedrick, R.M.; Freeman, K.G.; Cristinziano, M.; Smith, B.E.; Benson, C.A.; Jhaveri, T.A.; Baden, L.R.; Solomon, D.A.; Hatfull, G.F. Bacteriophage Treatment of Disseminated Cutaneous Mycobacterium chelonae Infection. Nat. Commun. 2022, 13, 2313. [Google Scholar] [CrossRef]
  105. Wang, J.; Meng, W.; Zhang, K.; Wang, J.; Lu, B.; Wang, R.; Jia, K. Topically Applied Bacteriophage to Control Multi-Drug Resistant Pseudomonas aeruginosa-Infected Wounds in a New Zealand Rabbit Model. Front. Microbiol. 2022, 13, 4171. [Google Scholar] [CrossRef]
  106. Golembo, M.; Puttagunta, S.; Rappo, U.; Weinstock, E.; Engelstein, R.; Gahali-Sass, I.; Moses, A.; Kario, E.; Ben-Dor Cohen, E.; Nicenboim, J.; et al. Development of a Topical Bacteriophage Gel Targeting Cutibacterium acnes for Acne Prone Skin and Results of a Phase 1 Cosmetic Randomized Clinical Trial. Ski. Health Dis. 2022, 2, e93. [Google Scholar] [CrossRef] [PubMed]
  107. Valente, L.G.; Pitton, M.; Urholz, M.F.; Oberhaensli, S.; Bruggmann, R.; Leib, S.L.; Jakob, S.M.; Resch, G.; Que, Y.-A.; Cameron, D.R. Isolation and Characterization of Bacteriophages from the Human Skin Microbiome That Infect Staphylococcus epidermidis. FEMS Microbes 2022, 2, xtab003. [Google Scholar] [CrossRef] [PubMed]
  108. Shimamori, Y.; Mitsunaka, S.; Yamashita, H.; Suzuki, T.; Kitao, T.; Kubori, T.; Nagai, H.; Takeda, S.; Ando, H. Staphylococcal Phage in Combination with Staphylococcus epidermidis as a Potential Treatment for Staphylococcus aureus-Associated Atopic Dermatitis and Suppressor of Phage-Resistant Mutants. Viruses 2020, 13, 7. [Google Scholar] [CrossRef]
  109. Metsemakers, W.-J.; Onsea, J.; Fintan Moriarty, T.; Pruidze, N.; Nadareishvili, L.; Dadiani, M.; Kutateladze, M.; Eliava, G. Bacteriophage Therapy for Human Musculoskeletal and Skin/Soft Tissue Infections. Clin. Microbiol. Infect. 2023, 29, 695–701. [Google Scholar] [CrossRef]
  110. Stacey, H.J.; De Soir, S.; Jones, J.D. The Safety and Efficacy of Phage Therapy: A Systematic Review of Clinical and Safety Trials. Antibiotics 2022, 11, 1340. [Google Scholar] [CrossRef] [PubMed]
  111. Azevedo, M.M.; Pina-Vaz, C.; Rodrigues, A.G. The Role of Phage Therapy in Burn Wound Infections Management: Advantages and Pitfalls. J. Burn. Care Res. 2022, 43, 336–342. [Google Scholar] [CrossRef]
  112. Mayslich, C.; Grange, P.A.; Dupin, N. Cutibacterium acnes as an Opportunistic Pathogen: An Update of Its Virulence-Associated Factors. Microorganisms 2021, 9, 303. [Google Scholar] [CrossRef]
  113. Verbanic, S.; Deacon, J.M.; Chen, I.A. The Chronic Wound Virome: Phage Diversity and Associations with Wounds and Healing Outcomes. medRxiv 2022. [Google Scholar] [CrossRef]
  114. Khullar, L.; Harjai, K.; Chhibber, S. Therapeutic and Pro-Healing Potential of Advanced Wound Dressings Loaded with Bioactive Agents. Future Microbiol. 2023, 18, 43–63. [Google Scholar] [CrossRef] [PubMed]
  115. 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. [Google Scholar] [CrossRef]
  116. De Souza, C.M.; Tanir, T.; Orellana, M.; Escalante, A.; Koeris, M.S. Manufacturing Bacteriophages (Part 2 of 2): Formulation, Analytics and Quality Control Considerations. Pharmaceuticals 2021, 14, 895. [Google Scholar] [CrossRef]
  117. Zhang, M.; Zhang, T.; Yu, M.; Chen, Y.L.; Jin, M. The Life Cycle Transitions of Temperate Phages: Regulating Factors and Potential Ecological Implications. Viruses 2022, 14, 1904. [Google Scholar] [CrossRef] [PubMed]
  118. Hobbs, Z.; Abedon, S.T. Diversity of Phage Infection Types and Associated Terminology: The Problem with ‘Lytic or Lysogenic’. FEMS Microbiol. Lett. 2016, 363, 47. [Google Scholar] [CrossRef] [Green Version]
  119. Venturini, C.; Fabijan, A.P.; Lubian, A.F.; Barbirz, S.; Iredell, J. Biological Foundations of Successful Bacteriophage Therapy. EMBO Mol. Med. 2022, 14, e12435. [Google Scholar] [CrossRef] [PubMed]
  120. Fillol-Salom, A.; Rostøl, J.T.; Ojiogu, A.D.; Chen, J.; Douce, G.; Humphrey, S.; Penadés, J.R. Bacteriophages Benefit from Mobilizing Pathogenicity Islands Encoding Immune Systems against Competitors. Cell 2022, 185, 3248–3262.e20. [Google Scholar] [CrossRef] [PubMed]
  121. Fernández, L.; González, S.; Quiles-Puchalt, N.; Gutiérrez, D.; Penadés, J.R.; García, P.; Rodríguez, A. Lysogenization of Staphylococcus aureus RN450 by Phages Φ11 and Φ80α Leads to the Activation of the SigB Regulon. Sci. Rep. 2018, 8, 12662. [Google Scholar] [CrossRef]
  122. Guo, D.; Chen, J.; Zhao, X.; Luo, Y.; Jin, M.; Fan, F.; Park, C.; Yang, X.; Sun, C.; Yan, J.; et al. Genetic and Chemical Engineering of Phages for Controlling Multidrug-Resistant Bacteria. Antibiotics 2021, 10, 202. [Google Scholar] [CrossRef] [PubMed]
  123. Chang, Y. Bacteriophage-Derived Endolysins Applied as Potent Biocontrol Agents to Enhance Food Safety. Microorganisms 2020, 8, 724. [Google Scholar] [CrossRef] [PubMed]
  124. Moreau, M.; Seité, S.; Aguilar, L.; Da Cruz, O.; Puech, J.; Frieling, J.; Demessant, A.L. Topical S. aureus-Targeting Endolysin Significantly Improves Symptoms and QoL in Individuals with Atopic Dermatitis. J. Drugs Dermatol. 2021, 20, 1323–1328. [Google Scholar] [CrossRef]
  125. Townsend, E.C.; Kalan, L.R. The Dynamic Balance of the Skin Microbiome across the Lifespan. Biochem. Soc. Trans. 2023, 51, 71–86. [Google Scholar] [CrossRef] [PubMed]
  126. Bomar, L.; Brugger, S.D.; Yost, B.H.; Davies, S.S.; Lemon, K.P. Corynebacterium accolens Releases Antipneumococcal Free Fatty Acids from Human Nostril and Skin Surface Triacylglycerols. mBio 2016, 7, e01725-15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Chen, H.; Zhao, Q.; Zhong, Q.; Cheng, D.; Krutmann, J.; Wang, J.; Xia, J. Skin Microbiome, Metabolome and Skin Phenome, from the Perspectives of Skin as an Ecosystem. Phenomics 2022, 2, 363–382. [Google Scholar] [CrossRef] [PubMed]
  128. Kimura, I.; Ichimura, A.; Ohue-Kitano, R.; Igarashi, M. Free Fatty Acid Receptors in Health and Disease. Physiol. Rev. 2020, 100, 171–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Rawlings, A.; Sabin, R.; Harding, C.; Watkinson, A.; Banks, J.; Ackerman, C. The Effect of Glycerol and Humidity on Desmosome Degradation in Stratum Corneum. Arch. Dermatol. Res. 1995, 287, 457–464. [Google Scholar] [CrossRef]
  130. Atrux-Tallau, N.; Romagny, C.; Padois, K.; Denis, A.; Haftek, M.; Falson, F.; Pirot, F.; Maibach, H.I. Effects of Glycerol on Human Skin Damaged by Acute Sodium Lauryl Sulphate Treatment. Arch. Dermatol. Res. 2010, 302, 435–441. [Google Scholar] [CrossRef]
  131. Salgaonkar, N.; Kadamkode, V.; Kumaran, S.; Mallemala, P.; Christy, E.; Appavoo, S.; Majumdar, A.; Mitra, R.; Dasgupta, A. Glycerol Fermentation by Skin Bacteria Generates Lactic Acid and Upregulates the Expression Levels of Genes Associated with the Skin Barrier Function. Exp. Dermatol. 2022, 31, 1364–1372. [Google Scholar] [CrossRef]
  132. Chen, Y.; Moran, J.C.; Campbell-Lee, S.; Horsburgh, M.J. Transcriptomic Responses and Survival Mechanisms of Staphylococci to the Antimicrobial Skin Lipid Sphingosine. Antimicrob. Agents Chemother. 2022, 66, e00569-21. [Google Scholar] [CrossRef] [PubMed]
  133. Araki, H.; Hajime, M.; Tamura, E.; Hagihara, H.; Takigawa, H.; Araki, H.; Hajime, M.; Tamura, E.; Hagihara, H.; Takigawa, H. Human Sebaceous Cis-6-Hexadecenoic Acid: Possible Application of an Innate Antimicrobial Substance to Cosmetic Products for Mucous Membrane. J. Cosmet. Dermatol. Sci. Appl. 2017, 7, 133–147. [Google Scholar] [CrossRef] [Green Version]
  134. Brandwein, M.; Steinberg, D.; Meshner, S. Microbial Biofilms and the Human Skin Microbiome. npj Biofilms Microbiomes 2016, 2, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Domenico, E.G.D.; Cavallo, I.; Capitanio, B.; Ascenzioni, F.; Pimpinelli, F.; Morrone, A.; Ensoli, F. Staphylococcus aureus and the Cutaneous Microbiota Biofilms in the Pathogenesis of Atopic Dermatitis. Microorganisms 2019, 7, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Vestby, L.K.; Grønseth, T.; Simm, R.; Nesse, L.L. Bacterial Biofilm and Its Role in the Pathogenesis of Disease. Antibiotics 2020, 9, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Gonzalez, T.; Stevens, M.L.; Baatyrbek, A.K.; Alarcon, R.; He, H.; Kroner, J.W.; Spagna, D.; Grashel, B.; Sidler, E.; Martin, L.J.; et al. Biofilm Propensity of Staphylococcus aureus Skin Isolates Is Associated with Increased Atopic Dermatitis Severity and Barrier Dysfunction in the MPAACH Pediatric Cohort. Allergy 2021, 76, 302. [Google Scholar] [CrossRef]
  138. Kuehnast, T.; Cakar, F.; Weinhäupl, T.; Pilz, A.; Selak, S.; Schmidt, M.A.; Rüter, C.; Schild, S. Comparative Analyses of Biofilm Formation among Different Cutibacterium acnes Isolates. Int. J. Med. Microbiol. 2018, 308, 1027–1035. [Google Scholar] [CrossRef]
  139. Lu, Y.; Cai, W.J.; Ren, Z.; Han, P. The Role of Staphylococcal Biofilm on the Surface of Implants in Orthopedic Infection. Microorganisms 2022, 10, 1909. [Google Scholar] [CrossRef]
  140. Garcia, D.; Mayfield, C.K.; Leong, J.; Deckey, D.G.; Zega, A.; Glasser, J.; Daniels, A.H.; Eberson, C.; Green, A.; Born, C. Early Adherence and Biofilm Formation of Cutibacterium acnes (Formerly Propionibacterium acnes) on Spinal Implant Materials. Spine J. 2020, 20, 981–987. [Google Scholar] [CrossRef]
  141. Nowrouzian, F.L.; Ljung, A.; Hesselmar, B.; Nilsson, S.; Adlerberth, I.; Wolda, A.E. Bacterial Carriage of Genes Encoding Fibronectin-Binding Proteins Is Associated with Long-Term Persistence of Staphylococcus aureus in the Nasal and Gut Microbiota of Infants. Appl. Environ. Microbiol. 2021, 87, 1–14. [Google Scholar] [CrossRef]
  142. Sugimoto, S.; Iwamoto, T.; Takada, K.; Okuda, K.I.; Tajima, A.; Iwase, T.; Mizunoe, Y. Staphylococcus epidermidis Esp Degrades Specific Proteins Associated with Staphylococcus aureus Biofilm Formation and Host-Pathogen Interaction. J. Bacteriol. 2013, 195, 1645–1655. [Google Scholar] [CrossRef] [Green Version]
  143. Iwase, T.; Uehara, Y.; Shinji, H.; Tajima, A.; Seo, H.; Takada, K.; Agata, T.; Mizunoe, Y. Staphylococcus epidermidis Esp Inhibits Staphylococcus aureus Biofilm Formation and Nasal Colonization. Nature 2010, 465, 346–349. [Google Scholar] [CrossRef]
  144. Kumari, A.; Singh, R. Antibiofilm Activity of Small Molecules Produced by Staphylococcus epidermidis against Staphylococcus aureus. Appl. Environ. Microbiol. 2020, 86, e00627-20. [Google Scholar] [CrossRef] [PubMed]
  145. Glatthardt, T.; Curityba de Mello Campos, J.; Chamon, R.C.; Freitas de Sá Coimbra, T.; de Almeida Rocha, G.; Figueira de Melo, M.A.; Parente, T.E.; Lobo, L.A.; Antunes, L.C.M.; dos Santos, K.R.N.; et al. Small Molecules Produced by Commensal Staphylococcus epidermidis Disrupt Formation of Biofilms by Staphylococcus aureus. Appl. Environ. Microbiol. 2020, 86, e02539-19. [Google Scholar] [CrossRef]
  146. Brooks, J.L.; Jefferson, K.K. Phase Variation of Poly-N-Acetylglucosamine Expression in Staphylococcus aureus. PLoS Pathog. 2014, 10, e1004292. [Google Scholar] [CrossRef]
  147. Kim, H.J.; Lee, B.J.; Kwon, A.R. The Grease Trap: Uncovering the Mechanism of the Hydrophobic Lid in Cutibacterium acnes Lipase. J. Lipid Res. 2020, 61, 722–733. [Google Scholar] [CrossRef] [Green Version]
  148. Brüggemann, H.; Salar-Vidal, L.; Gollnick, H.P.M.; Lood, R. A Janus-Faced Bacterium: Host-Beneficial and -Detrimental Roles of Cutibacterium acnes. Front. Microbiol. 2021, 12, 673845. [Google Scholar] [CrossRef]
  149. Huang, T.Y.; Jiang, Y.E.; Scott, D.A. Culturable Bacteria in the Entire Acne Lesion and Short-Chain Fatty Acid Metabolites of Cutibacterium acnes and Staphylococcus epidermidis Isolates. Biochem. Biophys. Res. Commun. 2022, 622, 45–49. [Google Scholar] [CrossRef]
  150. Sanford, J.A.; O’Neill, A.M.; Zouboulis, C.C.; Gallo, R.L. Short-Chain Fatty Acids from Cutibacterium acnes Activate Both a Canonical and Epigenetic Inflammatory Response in Human Sebocytes. J. Immunol. 2019, 202, 1767–1776. [Google Scholar] [CrossRef] [PubMed]
  151. Nakamura, K.; O’Neill, A.M.; Williams, M.R.; Cau, L.; Nakatsuji, T.; Horswill, A.R.; Gallo, R.L. Short Chain Fatty Acids Produced by Cutibacterium acnes Inhibit Biofilm Formation by Staphylococcus epidermidis. Sci. Rep. 2020, 10, 21237. [Google Scholar] [CrossRef] [PubMed]
  152. Gill, P.A.; van Zelm, M.C.; Muir, J.G.; Gibson, P.R. Review Article: Short Chain Fatty Acids as Potential Therapeutic Agents in Human Gastrointestinal and Inflammatory Disorders. Aliment. Pharmacol. Ther. 2018, 48, 15–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Keshari, S.; Balasubramaniam, A.; Myagmardoloonjin, B.; Herr, D.R.; Negari, I.P.; Huang, C.M. Butyric Acid from Probiotic Staphylococcus epidermidis in the Skin Microbiome Down-Regulates the Ultraviolet-Induced Pro-Inflammatory IL-6 Cytokine via Short-Chain Fatty Acid Receptor. Int. J. Mol. Sci. 2019, 20, 4477. [Google Scholar] [CrossRef] [Green Version]
  154. Xiao, X.; Hu, X.; Yao, J.; Cao, W.; Zou, Z.; Wang, L.; Qin, H.; Zhong, D.; Li, Y.; Xue, P.; et al. The Role of Short-Chain Fatty Acids in Inflammatory Skin Diseases. Front. Microbiol. 2023, 13, 5417. [Google Scholar] [CrossRef] [PubMed]
  155. Scheuerl, T.; Hopkins, M.; Nowell, R.W.; Rivett, D.W.; Barraclough, T.G.; Bell, T. Bacterial Adaptation Is Constrained in Complex Communities. Nat. Commun. 2020, 11, 754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Abisado, R.G.; Benomar, S.; Klaus, J.R.; Dandekar, A.A.; Chandler, J.R. Bacterial Quorum Sensing and Microbial Community Interactions. mBio 2018, 9, e02331-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Miller, M.B.; Bassler, B.L. Quorum Sensing in Bacteria. Annu. Rev. Microbiol. 2001, 55, 165–199. [Google Scholar] [CrossRef] [Green Version]
  158. Abbamondi, G.R.; Tommonaro, G. Research Progress and Hopeful Strategies of Application of Quorum Sensing in Food, Agriculture and Nanomedicine. Microorganisms 2022, 10, 1192. [Google Scholar] [CrossRef]
  159. Haque, S.; Yadav, D.K.; Bisht, S.C.; Yadav, N.; Singh, V.; Dubey, K.K.; Jawed, A.; Wahid, M.; Dar, S.A. Quorum Sensing Pathways in Gram-Positive and -Negative Bacteria: Potential of Their Interruption in Abating Drug Resistance. J. Chem. 2019, 31, 161–187. [Google Scholar] [CrossRef]
  160. Rutherford, S.T.; Bassler, B.L. Bacterial Quorum Sensing: Its Role in Virulence and Possibilities for Its Control. Cold Spring Harb. Perspect. Med. 2012, 2, a012427. [Google Scholar] [CrossRef]
  161. Chen, L.; Ku, L.; Li, M. Editorial: Frontiers in Bacterial Quorum Sensing Research. Front. Cell Infect. Microbiol. 2022, 12, 1293. [Google Scholar] [CrossRef]
  162. Grando, K.; Nicastro, L.K.; Tursi, S.A.; De Anda, J.; Lee, E.Y.; Wong, G.C.L.; Tükel, Ç. Phenol-Soluble Modulins from Staphylococcus aureus Biofilms Form Complexes with DNA to Drive Autoimmunity. Front. Cell Infect. Microbiol. 2022, 12, 517. [Google Scholar] [CrossRef]
  163. Erez, Z.; Steinberger-Levy, I.; Shamir, M.; Doron, S.; Stokar-Avihail, A.; Peleg, Y.; Melamed, S.; Leavitt, A.; Savidor, A.; Albeck, S.; et al. Communication between Viruses Guides Lysis–Lysogeny Decisions. Nature 2017, 541, 488–493. [Google Scholar] [CrossRef] [Green Version]
  164. Whiteley, M.; Diggle, S.P.; Greenberg, E.P. Progress in and Promise of Bacterial Quorum Sensing Research. Nature 2017, 551, 313–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Parlet, C.P.; Brown, M.M.; Horswill, A.R. Commensal Staphylococci Influence Staphylococcus aureus Skin Colonization and Disease. Trends Microbiol. 2019, 27, 497. [Google Scholar] [CrossRef] [PubMed]
  166. Brown, M.M.; Kwiecinski, J.M.; Cruz, L.M.; Shahbandi, A.; Todd, D.A.; Cech, N.B.; Horswill, A.R. Novel Peptide from Commensal Staphylococcus simulans Blocks Methicillin-Resistant Staphylococcus aureus Quorum Sensing and Protects Host Skin from Damage. Antimicrob. Agents Chemother. 2020, 64, e00172-20. [Google Scholar] [CrossRef] [PubMed]
  167. Severn, M.M.; Williams, M.R.; Shahbandi, A.; Bunch, Z.L.; Lyon, L.M.; Nguyen, A.; Zaramela, L.S.; Todd, D.A.; Zengler, K.; Cech, N.B.; et al. The Ubiquitous Human Skin Commensal Staphylococcus hominis Protects against Opportunistic Pathogens. mBio 2022, 13, e00930-22. [Google Scholar] [CrossRef]
  168. Severn, M.M.; Cho, Y.S.K.; Manzer, H.S.; Bunch, Z.L.; Shahbandi, A.; Todd, D.A.; Cech, N.B.; Horswill, A.R. The Commensal Staphylococcus warneri Makes Peptide Inhibitors of MRSA Quorum Sensing That Protect Skin from Atopic or Necrotic Damage. J. Investig. Dermatol. 2022, 142, 3349–3352.e5. [Google Scholar] [CrossRef] [PubMed]
  169. Pena, R.T.; Blasco, L.; Ambroa, A.; González-Pedrajo, B.; Fernández-García, L.; López, M.; Bleriot, I.; Bou, G.; García-Contreras, R.; Wood, T.K.; et al. Relationship between Quorum Sensing and Secretion Systems. Front. Microbiol. 2019, 10, 1100. [Google Scholar] [CrossRef] [Green Version]
  170. Ghosh, A.; Jayaraman, N.; Chatterji, D. Small-Molecule Inhibition of Bacterial Biofilm. ACS Omega 2020, 5, 3108–3115. [Google Scholar] [CrossRef]
  171. Balaban, N.; Goldkorn, T.; Gov, Y.; Hirshberg, M.; Koyfman, N.; Matthews, H.R.; Nhan, R.T.; Singh, B.; Uziel, O. Regulation of Staphylococcus aureus Pathogenesis via Target of RNAIII-Activating Protein (TRAP). J. Biol. Chem. 2001, 276, 2658–2667. [Google Scholar] [CrossRef] [Green Version]
  172. Ciulla, M.; Di Stefano, A.; Marinelli, L.; Cacciatore, I.; Di Biase, G. RNAIII Inhibiting Peptide (RIP) and Derivatives as Potential Tools for the Treatment of S. aureus Biofilm Infections. Curr. Top. Med. Chem. 2018, 18, 2068–2079. [Google Scholar] [CrossRef]
  173. Ye, H.; Wu, L.; Zhang, W.; Li, J.; Sun, X.; Chen, C.; Sun, Y.; Han, J. The Use of RNAIII Inhibitory Peptide in the Treatment of Elderly Patients with Staphylococcus aureus Infection and Construction of Fuzzy Mathematical Model for Curative Effect Evaluation. Results Phys. 2021, 25, 104323. [Google Scholar] [CrossRef]
  174. Simonetti, O.; Cirioni, O.; Ghiselli, R.; Goteri, G.; Scalise, A.; Orlando, F.; Silvestri, C.; Riva, A.; Saba, V.; Madanahally, K.D.; et al. RNAIII-Inhibiting Peptide Enhances Healing of Wounds Infected with Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2008, 52, 2205–2211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Grimshaw, S.G.; Smith, A.M.; Arnold, D.S.; Xu, E.; Hoptroff, M.; Murphy, B. The Diversity and Abundance of Fungi and Bacteria on the Healthy and Dandruff Affected Human Scalp. PLoS ONE 2019, 14, e0225796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Leong, C.; Schmid, B.; Buttafuoco, A.; Glatz, M.; Bosshard, P.P. In Vitro Efficacy of Antifungal Agents Alone and in Shampoo Formulation against Dandruff-Associated Malassezia spp. and Staphylococcus spp. Int. J. Cosmet. Sci. 2019, 41, 221–227. [Google Scholar] [CrossRef] [PubMed]
  177. Nguyen, U.T.; Kalan, L.R. Forgotten Fungi: The Importance of the Skin Mycobiome. Curr. Opin. Microbiol. 2022, 70, 102235. [Google Scholar] [CrossRef] [PubMed]
  178. Denissen, J.; Reyneke, B.; Waso-Reyneke, M.; Havenga, B.; Barnard, T.; Khan, S.; Khan, W. Prevalence of ESKAPE Pathogens in the Environment: Antibiotic Resistance Status, Community-Acquired Infection and Risk to Human Health. Int. J. Hyg. Environ. Health 2022, 244, 114006. [Google Scholar] [CrossRef]
  179. Ka-Wai Hui, E. Reasons for the Increase in Emerging and Re-Emerging Viral Infectious Diseases. Microbes Infect. 2006, 8, 905. [Google Scholar] [CrossRef] [PubMed]
  180. Fisher, M.C.; Alastruey-Izquierdo, A.; Berman, J.; Bicanic, T.; Bignell, E.M.; Bowyer, P.; Bromley, M.; Brüggemann, R.; Garber, G.; Cornely, O.A.; et al. Tackling the Emerging Threat of Antifungal Resistance to Human Health. Nat. Rev. Microbiol. 2022, 20, 557–571. [Google Scholar] [CrossRef] [PubMed]
  181. Fisher, M.C.; Hawkins, N.J.; Sanglard, D.; Gurr, S.J. Worldwide Emergence of Resistance to Antifungal Drugs Challenges Human Health and Food Security. Science 2018, 360, 739–742. [Google Scholar] [CrossRef] [Green Version]
  182. Saheb Kashaf, S.; Proctor, D.M.; Deming, C.; Saary, P.; Hölzer, M.; Mullikin, J.; Thomas, J.; Young, A.; Bouffard, G.; Barnabas, B.; et al. Integrating Cultivation and Metagenomics for a Multi-Kingdom View of Skin Microbiome Diversity and Functions. Nat. Microbiol. 2021, 7, 169–179. [Google Scholar] [CrossRef]
  183. Vijaya Chandra, S.H.; Srinivas, R.; Dawson, T.L.; Common, J.E. Cutaneous Malassezia: Commensal, Pathogen, or Protector? Front. Cell Infect. Microbiol. 2020, 10, 1. [Google Scholar] [CrossRef]
  184. Gaitanis, G.; Magiatis, P.; Mexia, N.; Melliou, E.; Efstratiou, M.A.; Bassukas, I.D.; Velegraki, A. Antifungal Activity of Selected Malassezia Indolic Compounds Detected in Culture. Mycoses 2019, 62, 597–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Sparber, F.; LeibundGut-Landmann, S. Host Responses to Malassezia spp. in the Mammalian Skin. Front. Immunol. 2017, 8, 1614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Sparber, F.; De Gregorio, C.; Steckholzer, S.; Ferreira, F.M.; Dolowschiak, T.; Ruchti, F.; Kirchner, F.R.; Mertens, S.; Prinz, I.; Joller, N.; et al. The Skin Commensal Yeast Malassezia Triggers a Type 17 Response That Coordinates Anti-Fungal Immunity and Exacerbates Skin Inflammation. Cell Host Microbe 2019, 25, 389–403.e6. [Google Scholar] [CrossRef] [Green Version]
  187. Naik, S.; Bouladoux, N.; Linehan, J.L.; Han, S.J.; Harrison, O.J.; Wilhelm, C.; Conlan, S.; Himmelfarb, S.; Byrd, A.L.; Deming, C.; et al. Commensal–Dendritic-Cell Interaction Specifies a Unique Protective Skin Immune Signature. Nature 2015, 520, 104–108. [Google Scholar] [CrossRef] [Green Version]
  188. Wanke, I.; Steffen, H.; Christ, C.; Krismer, B.; Götz, F.; Peschel, A.; Schaller, M.; Schittek, B. Skin Commensals Amplify the Innate Immune Response to Pathogens by Activation of Distinct Signaling Pathways. J. Investig. Dermatol. 2011, 131, 382–390. [Google Scholar] [CrossRef] [Green Version]
  189. Höfs, S.; Mogavero, S.; Hube, B. Interaction of Candida Albicans with Host Cells: Virulence Factors, Host Defense, Escape Strategies, and the Microbiota. J. Microbiol. 2016, 54, 149–169. [Google Scholar] [CrossRef] [PubMed]
  190. Desai, J.V. Candida Albicans Hyphae: From Growth Initiation to Invasion. J. Fungi 2018, 4, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Lacroix, G.; Gouyer, V.; Gottrand, F.; Desseyn, J.L. The Cervicovaginal Mucus Barrier. Int. J. Mol. Sci. 2020, 21, 8266. [Google Scholar] [CrossRef] [PubMed]
  192. Wu, Y.; Hu, S.; Wu, C.; Gu, F.; Yang, Y. Probiotics: Potential Novel Therapeutics Against Fungal Infections. Front. Cell Infect. Microbiol. 2021, 11, 793419. [Google Scholar] [CrossRef]
  193. Mattiuzzi, C.; Lippi, G. Current Cancer Epidemiology. J. Epidemiol. Glob. Health 2019, 9, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  195. Woo, Y.R.; Cho, S.H.; Lee, J.D.; Kim, H.S. The Human Microbiota and Skin Cancer. Int. J. Mol. Sci. 2022, 23, 1813. [Google Scholar] [CrossRef] [PubMed]
  196. Zhang, M.; Qureshi, A.A.; Fortner, R.T.; Hankinson, S.E.; Wei, Q.; Wang, L.E.; Eliassen, A.H.; Willett, W.C.; Hunter, D.J.; Han, J. Teenage Acne and Cancer Risk in US Women: A Prospective Cohort Study. Cancer 2015, 121, 1681–1687. [Google Scholar] [CrossRef] [Green Version]
  197. Zhu, Y.; Wang, H.; He, J.; Yang, L.; Zhou, X.; Li, Z.; Zhou, H.; Zhao, H.; Li, Y. Atopic Dermatitis and Skin Cancer Risk: A Systematic Review. Dermatol. Ther. 2022, 12, 1167. [Google Scholar] [CrossRef] [PubMed]
  198. Butrón-Bris, B.; Daudén, E.; Rodríguez-Jiménez, P. Psoriasis Therapy and Skin Cancer: A Review. Life 2021, 11, 1109. [Google Scholar] [CrossRef]
  199. Li, W.Q.; Zhang, M.; Danby, F.W.; Han, J.; Qureshi, A.A. Personal History of Rosacea and Risk of Incident Cancer among Women in the US. Br. J. Cancer 2015, 113, 520. [Google Scholar] [CrossRef] [Green Version]
  200. Mercier-Parot, L.T.-D.H. Skeletal Malformations in Rats Produced by 6-Hydroxylaminopurine. Attempts at Prevention. C. R. Seances Soc. Biol. Fil. 1973, 167, 5–10. [Google Scholar]
  201. Stepchenkova, E.I.; Kozmin, S.G.; Alenin, V.V.; Pavlov, Y.I. Genome-Wide Screening for Genes Whose Deletions Confer Sensitivity to Mutagenic Purine Base Analogs in Yeast. BMC Genet. 2005, 6, 31. [Google Scholar] [CrossRef] [Green Version]
  202. Nakatsuji, T.; Chen, T.H.; Butcher, A.M.; Trzoss, L.L.; Nam, S.J.; Shirakawa, K.T.; Zhou, W.; Oh, J.; Otto, M.; Fenical, W.; et al. A Commensal Strain of Staphylococcus epidermidis Protects against Skin Neoplasia. Sci. Adv. 2018, 4, eaao4502. [Google Scholar] [CrossRef] [Green Version]
  203. Nakatsuji, T.; Fenical, W.; Gallo, R.L. Response to Comment on “A Commensal Strain of Staphylococcus epidermidis Protects against Skin Neoplasia” by Nakatsuji et Al. Sci. Adv. 2019, 5, 5611–5622. [Google Scholar] [CrossRef] [Green Version]
  204. Kozmin, S.G.; Rogozin, I.B.; Moore, E.A.; Abney, M.; Schaaper, R.M.; Pavlov, Y.I. Comment on “A Commensal Strain of Staphylococcus epidermidis Protects against Skin Neoplasia” by Nakatsuji et Al. Sci. Adv. 2019, 5, 3915–3926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Ding, L.; Cao, J.; Lin, W.; Chen, H.; Xiong, X.; Ao, H.; Yu, M.; Lin, J.; Cui, Q. The Roles of Cyclin-Dependent Kinases in Cell-Cycle Progression and Therapeutic Strategies in Human Breast Cancer. Int. J. Mol. Sci. 2020, 21, 1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Yang, L.; Li, X.; Huang, W.; Rao, X.; Lai, Y. Pharmacological Properties of Indirubin and Its Derivatives. Biomed. Pharmacother. 2022, 151, 113112. [Google Scholar] [CrossRef] [PubMed]
  207. Saunte, D.M.L.; Gaitanis, G.; Hay, R.J. Malassezia-Associated Skin Diseases, the Use of Diagnostics and Treatment. Front. Cell Infect. Microbiol. 2020, 10, 112. [Google Scholar] [CrossRef] [PubMed]
  208. Molujin, A.M.; Abbasiliasi, S.; Nurdin, A.; Lee, P.C.; Gansau, J.A.; Jawan, R. Bacteriocins as Potential Therapeutic Approaches in the Treatment of Various Cancers: A Review of In Vitro Studies. Cancers 2022, 14, 4758. [Google Scholar] [CrossRef] [PubMed]
  209. Sharma, P.; Tiwari, S.K. Bacteriocins of Probiotics as Potent Anticancer Agents. In Probiotic Research in Therapeutics; Springer: Berlin/Heidelberg, Germany, 2021; Volume 1, pp. 231–250. [Google Scholar] [CrossRef]
  210. Tran, N.B.V.; Truong, Q.M.; Nguyen, L.Q.A.; Nguyen, N.M.H.; Tran, Q.H.; Dinh, T.T.P.; Hua, V.S.; Nguyen, V.D.; Lambert, P.A.; Nguyen, T.T.H. Prevalence and Virulence of Commensal Pseudomonas aeruginosa Isolates from Healthy Individuals in Southern Vietnam (2018–2020). Biomedicines 2022, 11, 54. [Google Scholar] [CrossRef]
  211. Zgheib, H.; Drider, D.; Belguesmia, Y. Broadening and Enhancing Bacteriocins Activities by Association with Bioactive Substances. Int. J. Environ. Res. Public Health 2020, 17, 7835. [Google Scholar] [CrossRef]
  212. Liu, J.K. Natural Products in Cosmetics. Nat. Prod. Bioprospect. 2022, 12, 40. [Google Scholar] [CrossRef]
  213. Murphy, B.; Grimshaw, S.; Hoptroff, M.; Paterson, S.; Arnold, D.; Cawley, A.; Adams, S.E.; Falciani, F.; Dadd, T.; Eccles, R.; et al. Alteration of Barrier Properties, Stratum Corneum Ceramides and Microbiome Composition in Response to Lotion Application on Cosmetic Dry Skin. Sci. Rep. 2022, 12, 5223. [Google Scholar] [CrossRef]
  214. Mukherjee, S.; Mitra, R.; Maitra, A.; Gupta, S.; Kumaran, S.; Chakrabortty, A.; Majumder, P.P. Sebum and Hydration Levels in Specific Regions of Human Face Significantly Predict the Nature and Diversity of Facial Skin Microbiome. Sci. Rep. 2016, 6, 36062. [Google Scholar] [CrossRef] [Green Version]
  215. Roux, P.F.; Oddos, T.; Stamatas, G. Deciphering the Role of Skin Surface Microbiome in Skin Health: An Integrative Multiomics Approach Reveals Three Distinct Metabolite–Microbe Clusters. J. Investig. Dermatol. 2022, 142, 469–479.e5. [Google Scholar] [CrossRef] [PubMed]
  216. Lee, H.J.; Kim, M. Skin Barrier Function and the Microbiome. Int. J. Mol. Sci. 2022, 23, 13071. [Google Scholar] [CrossRef] [PubMed]
  217. Habeebuddin, M.; Karnati, R.K.; Shiroorkar, P.N.; Nagaraja, S.; Asdaq, S.M.B.; Anwer, M.K.; Fattepur, S. Topical Probiotics: More Than a Skin Deep. Pharmaceutics 2022, 14, 557. [Google Scholar] [CrossRef]
  218. Ellis, S.R.; Nguyen, M.; Vaughn, A.R.; Notay, M.; Burney, W.A.; Sandhu, S.; Sivamani, R.K. The Skin and Gut Microbiome and Its Role in Common Dermatologic Conditions. Microorganisms 2019, 7, 550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Leung, A.K.C.; Barankin, B.; Lam, J.M.; Leong, K.F.; Hon, K.L. Dermatology: How to Manage Acne vulgaris. Drugs Context 2021, 10. [Google Scholar] [CrossRef] [PubMed]
  220. Mavranezouli, I.; Welton, N.J.; Daly, C.H.; Wilcock, J.; Bromham, N.; Berg, L.; Xu, J.; Wood, D.; Ravenscroft, J.C.; Dworzynski, K.; et al. Cost-effectiveness of Topical Pharmacological, Oral Pharmacological, Physical and Combined Treatments for Acne vulgaris. Clin. Exp. Dermatol. 2022, 47, 2176–2187. [Google Scholar] [CrossRef] [PubMed]
  221. Sutaria, A.H.; Masood, S.; Schlessinger, J. Acne Vulgari; StatePearls Publishing: Treasure Island, FL USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK459173/ (accessed on 19 June 2023).
  222. Goodarzi, A.; Mozafarpoor, S.; Bodaghabadi, M.; Mohamadi, M. The Potential of Probiotics for Treating Acne vulgaris: A Review of Literature on Acne and Microbiota. Dermatol. Ther. 2020, 33, e13279. [Google Scholar] [CrossRef] [PubMed]
  223. Kang, B.S.; Seo, J.G.; Lee, G.S.; Kim, J.H.; Kim, S.Y.; Han, Y.W.; Kang, H.; Kim, H.O.; Rhee, J.H.; Chung, M.J.; et al. Antimicrobial Activity of Enterocins from Enterococcus faecalis SL-5 against Propionibacterium acnes, the Causative Agent in Acne vulgaris, and Its Therapeutic Effect. J. Microbiol. 2009, 47, 101–109. [Google Scholar] [CrossRef]
  224. Muizzuddin, N.; Maher, W.; Sullivan, M.; Schnittger, S.; Mammone, T. Physiological Effect of a Probiotic on Skin. J. Cosmet. Sci. 2012, 63, 385–395. [Google Scholar]
  225. Lebeer, S.; Oerlemans, E.F.M.; Claes, I.; Henkens, T.; Delanghe, L.; Wuyts, S.; Spacova, I.; van den Broek, M.F.L.; Tuyaerts, I.; Wittouck, S.; et al. Selective Targeting of Skin Pathobionts and Inflammation with Topically Applied Lactobacilli. Cell Rep. Med. 2022, 3, 100521. [Google Scholar] [CrossRef]
  226. Frazier, W.; Bhardwaj, N. Atopic Dermatitis: Diagnosis and Treatment. Am. Fam. Physician 2020, 101, 590–598. [Google Scholar] [PubMed]
  227. Wollenberg, A.; Thomsen, S.F.; Lacour, J.P.; Jaumont, X.; Lazarewicz, S. Targeting Immunoglobulin E in Atopic Dermatitis: A Review of the Existing Evidence. World Allergy Organ. J. 2021, 14, 100519. [Google Scholar] [CrossRef] [PubMed]
  228. Nakatsuji, T.; Chen, T.H.; Narala, S.; Chun, K.A.; Two, A.M.; Yun, T.; Shafiq, F.; Kotol, P.F.; Bouslimani, A.; Melnik, A.V.; et al. Antimicrobials from Human Skin Commensal Bacteria Protect against Staphylococcus aureus and Are Deficient in Atopic Dermatitis. Sci. Transl. Med. 2017, 9, eaah4680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Bjerre, R.D.; Holm, J.B.; Palleja, A.; Sølberg, J.; Skov, L.; Johansen, J.D. Skin Dysbiosis in the Microbiome in Atopic Dermatitis Is Site-Specific and Involves Bacteria, Fungus and Virus. BMC Microbiol. 2021, 21, 1–13. [Google Scholar] [CrossRef] [PubMed]
  230. Koh, L.F.; Ong, R.Y.; Common, J.E. Skin Microbiome of Atopic Dermatitis. Allergol. Int. 2022, 71, 31–39. [Google Scholar] [CrossRef]
  231. Knackstedt, R.; Knackstedt, T.; Gatherwright, J. The Role of Topical Probiotics in Skin Conditions: A Systematic Review of Animal and Human Studies and Implications for Future Therapies. Exp. Dermatol. 2020, 29, 15–21. [Google Scholar] [CrossRef] [Green Version]
  232. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of Aging: An Expanding Universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef]
  233. Zhang, S.; Duan, E. Fighting against Skin Aging: The Way from Bench to Bedside. Cell Transpl. 2018, 27, 729. [Google Scholar] [CrossRef]
  234. Zhou, W.; Fleming, E.; Legendre, G.; Roux, L.; Latreille, J.; Gendronneau, G.; Forestier, S.; Oh, J. Skin Microbiome Attributes Associate with Biophysical Skin Aging. bioRxiv 2023. [Google Scholar] [CrossRef]
  235. Murphy, B.; Hoptroff, M.; Arnold, D.; Cawley, A.; Smith, E.; Adams, S.E.; Mitchell, A.; Horsburgh, M.J.; Hunt, J.; Dasgupta, B.; et al. Compositional Variations between Adult and Infant Skin Microbiome: An Update. Microorganisms 2023, 11, 1484. [Google Scholar] [CrossRef]
  236. Andersson, T.; Ertürk Bergdahl, G.; Saleh, K.; Magnúsdóttir, H.; Stødkilde, K.; Andersen, C.B.F.; Lundqvist, K.; Jensen, A.; Brüggemann, H.; Lood, R. Common Skin Bacteria Protect Their Host from Oxidative Stress through Secreted Antioxidant RoxP. Sci. Rep. 2019, 9, 3596. [Google Scholar] [CrossRef] [Green Version]
  237. Kim, G.; Kim, M.; Kim, M.; Park, C.; Yoon, Y.; Lim, D.H.; Yeo, H.; Kang, S.; Lee, Y.G.; Beak, N.I.; et al. Spermidine-Induced Recovery of Human Dermal Structure and Barrier Function by Skin Microbiome. Commun. Biol. 2021, 4, 1–11. [Google Scholar] [CrossRef] [PubMed]
  238. Madeo, F.; Carmona-Gutierrez, D.; Kepp, O.; Kroemer, G. Spermidine Delays Aging in Humans. Aging 2018, 10, 2209. [Google Scholar] [CrossRef]
  239. Madeo, F.; Eisenberg, T.; Pietrocola, F.; Kroemer, G. Spermidine in Health and Disease. Science 2018, 359, eaan2788. [Google Scholar] [CrossRef] [Green Version]
  240. Madeo, F.; Bauer, M.A.; Carmona-Gutierrez, D.; Kroemer, G. Spermidine: A Physiological Autophagy Inducer Acting as an Anti-Aging Vitamin in Humans? Autophagy 2018, 15, 165–168. [Google Scholar] [CrossRef]
  241. Stødkilde, K.; Nielsen, J.T.; Petersen, S.V.; Paetzold, B.; Brüggemann, H.; Mulder, F.A.A.; Andersen, C.B.F. Solution Structure of the Cutibacterium acnes-Specific Protein RoxP and Insights Into Its Antioxidant Activity. Front. Cell Infect. Microbiol. 2022, 12, 100. [Google Scholar] [CrossRef] [PubMed]
  242. Allhorn, M.; Arve, S.; Brüggemann, H.; Lood, R. A Novel Enzyme with Antioxidant Capacity Produced by the Ubiquitous Skin Colonizer Propionibacterium acnes. Sci. Rep. 2016, 6, 36412. [Google Scholar] [CrossRef] [Green Version]
  243. Teichmann, P.; Both, A.; Wolz, C.; Hornef, M.W.; Rohde, H.; Yazdi, A.S.; Burian, M. The Staphylococcus epidermidis Transcriptional Profile During Carriage. Front. Microbiol. 2022, 13, 1537. [Google Scholar] [CrossRef] [PubMed]
  244. Zheng, Y.; Hunt, R.L.; Villaruz, A.E.; Fisher, E.L.; Liu, R.; Liu, Q.; Cheung, G.Y.C.; Li, M.; Otto, M. Commensal Staphylococcus epidermidis Contributes to Skin Barrier Homeostasis by Generating Protective Ceramides. Cell Host Microbe 2022, 30, 301–313.e9. [Google Scholar] [CrossRef]
  245. Li, Q.; Fang, H.; Dang, E.; Wang, G. The Role of Ceramides in Skin Homeostasis and Inflammatory Skin Diseases. J. Dermatol. Sci. 2020, 97, 2–8. [Google Scholar] [CrossRef]
  246. Di Marzio, L.; Cinque, B.; De Simone, C.; Cifone, M.G. Effect of the Lactic Acid Bacterium Streptococcus thermophilus on Ceramide Levels in Human KeratinocytesIn Vitro and Stratum Corneum In Vivo. J. Investig. Dermatol. 1999, 113, 98–106. [Google Scholar] [CrossRef] [Green Version]
  247. Di Marzio, L.; Cinque, B.; Cupelli, F.; De Simone, C.; Cifone, M.G.; Giuliani, M. Increase of Skin-Ceramide Levels in Aged Subjects Following a Short-Term Topical Application of Bacterial Sphingomyelinase from Streptococcus thermophilus. Int. J. Immunopathol. Pharmacol. 2008, 21, 137–143. [Google Scholar] [CrossRef] [PubMed]
  248. Izawa, N.; Hanamizu, T.; Iizuka, R.; Sone, T.; Mizukoshi, H.; Kimura, K.; Chiba, K. Streptococcus thermophilus Produces Exopolysaccharides Including Hyaluronic Acid. J. Biosci. Bioeng. 2009, 107, 119–123. [Google Scholar] [CrossRef] [PubMed]
  249. Swaney, M.H.; Kalan, L.R. Two-for-One: Dual Host-Microbe Functions of S. epidermidis Sph. Cell Host Microbe 2022, 30, 279–280. [Google Scholar] [CrossRef] [PubMed]
  250. Boer, D.E.C.; van Smeden, J.; Al-Khakany, H.; Melnik, E.; van Dijk, R.; Absalah, S.; Vreeken, R.J.; Haenen, C.C.P.; Lavrijsen, A.P.M.; Overkleeft, H.S.; et al. Skin of Atopic Dermatitis Patients Shows Disturbed β-Glucocerebrosidase and Acid Sphingomyelinase Activity That Relates to Changes in Stratum Corneum Lipid Composition. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2020, 1865, 158673. [Google Scholar] [CrossRef]
  251. Delanghe, L.; Spacova, I.; Van Malderen, J.; Oerlemans, E.; Claes, I.; Lebeer, S. The Role of Lactobacilli in Inhibiting Skin Pathogens. Biochem. Soc. Trans. 2021, 49, 617–627. [Google Scholar] [CrossRef]
  252. Mohammedsaeed, W.; Cruickshank, S.; McBain, A.J.; O’Neill, C.A. Lactobacillus rhamnosus GG Lysate Increases Re-Epithelialization of Keratinocyte Scratch Assays by Promoting Migration. Sci. Rep. 2015, 5, 16147. [Google Scholar] [CrossRef] [Green Version]
  253. Draelos, Z.D. The Science behind Skin Care: Moisturizers. J. Cosmet. Dermatol. 2018, 17, 138–144. [Google Scholar] [CrossRef] [PubMed]
  254. Chen, H.J.; Lee, P.Y.; Chen, C.Y.; Huang, S.L.; Huang, B.W.; Dai, F.J.; Chau, C.F.; Chen, C.S.; Lin, Y.S. Moisture Retention of Glycerin Solutions with Various Concentrations: A Comparative Study. Sci. Rep. 2022, 12, 10232. [Google Scholar] [CrossRef]
  255. Iglesia, S.; Kononov, T.; Zahr, A.S. A Multi-functional Anti-aging Moisturizer Maintains a Diverse and Balanced Facial Skin Microbiome. J. Appl. Microbiol. 2022, 133, 1791–1799. [Google Scholar] [CrossRef]
  256. Jung, Y.O.; Jeong, H.; Cho, Y.; Lee, E.O.; Jang, H.W.; Kim, J.; Nam, K.T.; Lim, K.M. Lysates of a Probiotic, Lactobacillus rhamnosus, Can Improve Skin Barrier Function in a Reconstructed Human Epidermis Model. Int. J. Mol. Sci. 2019, 20, 4289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Chandan, N.; Rajkumar, J.R.; Shi, V.Y.; Lio, P.A. A New Era of Moisturizers. J. Cosmet. Dermatol. 2021, 20, 2425–2430. [Google Scholar] [CrossRef]
  258. Bamford, J.T.M.; Flores-Genuino, R.N.S.; Ray, S.; Bigby, M.; Morales-Sánchez, M.A.; Arkoncel, M.; Realubit-Serrano, M.A.C. Interventions for the Treatment of Pityriasis Versicolor. Cochrane Database Syst. Rev. 2018, 2018, CD011208. [Google Scholar] [CrossRef]
  259. Gaitanis, G.; Magiatis, P.; Stathopoulou, K.; Bassukas, I.D.; Alexopoulos, E.C.; Velegraki, A.; Skaltsounis, A.L. AhR Ligands, Malassezin, and Indolo[3,2-b]Carbazole Are Selectively Produced by Malassezia furfur Strains Isolated from Seborrheic Dermatitis. J. Investig. Dermatol. 2008, 128, 1620–1625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Van den Bogaard, E.H.; Esser, C.; Perdew, G.H. The Aryl Hydrocarbon Receptor at the Forefront of Host-Microbe Interactions in the Skin: A Perspective on Current Knowledge Gaps and Directions for Future Research and Therapeutic Applications. Exp. Dermatol. 2021, 30, 1477–1483. [Google Scholar] [CrossRef] [PubMed]
  261. Grimes, P.; Bhawan, J.; Howell, M.; Desai, S.; Coryell, E.; Einziger, M.; Simpson, A.; Yaroshinsky, A.; McCraw, T. Histopathological Changes Induced by Malassezin: A Novel Natural Microbiome Indole for Treatment of Facial Hyperpigmentation. J. Drugs Dermatol. 2022, 21, 141–145. [Google Scholar] [CrossRef] [PubMed]
  262. Kurniadi, I.; Wijaya, W.H.; Timotius, K.H. Malassezia Virulence Factors and Their Role in Dermatological Disorders. Acta Dermatovenerol. Alp. Pannonica Adriat. 2022, 31, 65–70. [Google Scholar] [CrossRef] [PubMed]
  263. Grimes, P.E.; Bhawan, J.; Howell, M.D.; Desai, S.; Coryell, E.; Nashawati, R.; Einziger, M.; Simpson, A.M.; Yaroshinsky, A.; Mc Craw, T. A Novel Proof-of-Concept Study Assessing the Lightening Effects and Safety of Malassezin for Treatment of Facial Hyperpigmentation. J. Am. Acad. Dermatol. 2022, 87, 456–458. [Google Scholar] [CrossRef]
  264. Fortuna, M.C.; Garelli, V.; Pranteda, G.; Romaniello, F.; Cardone, M.; Carlesimo, M.; Rossi, A. A Case of Scalp Rosacea Treated with Low Dose Doxycycline and Probiotic Therapy and Literature Review on Therapeutic Options. Dermatol. Ther. 2016, 29, 249–251. [Google Scholar] [CrossRef]
  265. Smythe, P.; Wilkinson, H.N. The Skin Microbiome: Current Landscape and Future Opportunities. Int. J. Mol. Sci. 2023, 24, 3950. [Google Scholar] [CrossRef]
  266. Larson, P.J.; Chong, D.; Fleming, E.; Oh, J. Challenges Developing a Human Model System for Skin Microbiome Research. J. Investig. Dermatol. 2021, 141, 228. [Google Scholar] [CrossRef] [PubMed]
  267. Shen, J.; McFarland, A.G.; Blaustein, R.A.; Rose, L.J.; Perry-Dow, K.A.; Moghadam, A.A.; Hayden, M.K.; Young, V.B.; Hartmann, E.M. An Improved Workflow for Accurate and Robust Healthcare Environmental Surveillance Using Metagenomics. Microbiome 2022, 10, 1–18. [Google Scholar] [CrossRef] [PubMed]
  268. Shute, A.; Bihan, D.G.; Lewis, I.A.; Nasser, Y. Metabolomics: The Key to Unraveling the Role of the Microbiome in Visceral Pain Neurotransmission. Front. Neurosci. 2022, 16, 937. [Google Scholar] [CrossRef] [PubMed]
Figure 2. Drivers structuring the human skin microbiome structure. Intrinsic intrapersonal factors are associated with the differing physiological conditions across different microenvironments on the body. Extrinsic intrapersonal factors relate to environmentally derived variations, such as observations of cultural practices. Intrinsic interpersonal drivers are associated with the biological variation between individuals, such as hormonal differences between genders leading to altered skin physiology [45]. Created with www.BioRender.com.
Figure 2. Drivers structuring the human skin microbiome structure. Intrinsic intrapersonal factors are associated with the differing physiological conditions across different microenvironments on the body. Extrinsic intrapersonal factors relate to environmentally derived variations, such as observations of cultural practices. Intrinsic interpersonal drivers are associated with the biological variation between individuals, such as hormonal differences between genders leading to altered skin physiology [45]. Created with www.BioRender.com.
Microorganisms 11 01899 g002
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.

Share and Cite

MDPI and ACS Style

Nicholas-Haizelden, K.; Murphy, B.; Hoptroff, M.; Horsburgh, M.J. Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products. Microorganisms 2023, 11, 1899. https://doi.org/10.3390/microorganisms11081899

AMA Style

Nicholas-Haizelden K, Murphy B, Hoptroff M, Horsburgh MJ. Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products. Microorganisms. 2023; 11(8):1899. https://doi.org/10.3390/microorganisms11081899

Chicago/Turabian Style

Nicholas-Haizelden, Keir, Barry Murphy, Michael Hoptroff, and Malcolm J. Horsburgh. 2023. "Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products" Microorganisms 11, no. 8: 1899. https://doi.org/10.3390/microorganisms11081899

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop