Next Article in Journal
Influence of Nitrate Supplementation on Endurance Cyclic Sports Performance: A Systematic Review
Next Article in Special Issue
Anti-Heartburn Effects of Sugar Cane Flour: A Double-Blind, Randomized, Placebo-Controlled Study
Previous Article in Journal
A Comparison of Vitamin and Lutein Concentrations in Breast Milk from Four Asian Countries
Previous Article in Special Issue
Anti-Pathogenic Functions of Non-Digestible Oligosaccharides In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Potential of Skin Microbiome, Pro- and/or Pre-Biotics to Affect Local Cutaneous Responses to UV Exposure

by
VijayKumar Patra
1,2,
Irène Gallais Sérézal
3,4 and
Peter Wolf
2,*
1
Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
2
Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, 8010 Graz, Austria
3
Department of Medicine, Unit of Rheumatology, Karolinska Institutet, 171 77 Solna, Sweden
4
Department of Dermatology, Besançon University Hospital, 25000 Besancon, France
*
Author to whom correspondence should be addressed.
Submission received: 4 June 2020 / Revised: 14 June 2020 / Accepted: 15 June 2020 / Published: 17 June 2020
(This article belongs to the Special Issue Role of Prebiotics and Probiotics in Health and Disease)

Abstract

:
The human skin hosts innumerable microorganisms and maintains homeostasis with the local immune system despite the challenges offered by environmental factors such as ultraviolet radiation (UVR). UVR causes cutaneous alterations such as acute (i.e., sunburn) and chronic inflammation, tanning, photoaging, skin cancer, and immune modulation. Phototherapy on the other hand is widely used to treat inflammatory skin diseases such as psoriasis, atopic dermatitis, polymorphic light eruption and graft-versus-host disease (GvHD), as well as neoplastic skin diseases such as cutaneous T cell lymphoma, among others. Previous work has addressed the use of pro- and pre-biotics to protect against UVR through anti-oxidative, anti-inflammatory, anti-aging, anti-carcinogenic and/or pro-and contra-melanogenic properties. Herein, we discuss and share perspectives of the potential benefits of novel treatment strategies using microbes and pro- and pre-biotics as modulators of the skin response to UVR, and how they could act both for protection against UVR-induced skin damage and as enhancers of the UVR-driven therapeutic effects on the skin.

1. Introduction

1.1. Skin Microbiome

The healthy human skin harbors multitude of diverse and complex communities of bacteria, fungi, viruses, archaea, and mites, collectively called the skin microbiome [1,2,3,4]. The skin displays striking variation of microbial composition across body sites, shaped by their physical, chemical, and biological features [5,6,7]. Unlike the nutrient rich intestinal environment, the skin surface lacks many nutrients beyond basic proteins and lipids. In order to survive in this hostile environment, the resident microbes have to adapt and utilize available resources present in the stratum corneum (sphingolipids, amino acids, peptides, nitric oxide), sweat (NaCl, H2O, HCO3+, glucose, amino acids, free fatty acids, urea, ammonia, lactate, vitamins, peptides, sterols, mucopolysaccharides) and sebum (free fatty acids, sterols, squalene, wax esters). In addition, the skin microbiome must maintain a constant interaction and healthy interplay with the skin’s immune system for its survival. Studies using germ-free animals have shown a crucial role of skin microbiome in physiological and immunological functions. For example, Staphylococcus epidermidis, one of the dominant skin-associated bacteria produces several antimicrobial compounds and proteases that can limit the formation of biofilms by pathogenic species. Colonizing the skin with Staphylococcus epidermidis remodels the skin immunity by inducing IL-17a+ CD8+ T cells that migrate to the epidermis, enhances the immunity and limits pathogen invasion [5,8]. Numerous studies suggest that the skin microbiome is intricately involved in a wide range of molecular and cellular processes within the skin and beyond [9,10]. Thus, the nutrients that play an important role in shaping the individual differences in microbial signature ultimately contribute to health and diseases.

1.2. Ultraviolet Radiation (UVR)

UVR, especially UV-A (315–400 nm) and UV-B (290–315 nm) is one of the most prominent external factors affecting the skin. UV-A rays are either UV-A1 (340–400 nm) or UV-A2 (320–340 nm). The penetration of UV-A through the skin layers (epidermis and dermis) is about 5–10 times higher than that of UV-B [11], and UV-A (like UV-B) is known to play a more major role in skin aging, wrinkling (photoaging) and takes part in initiating and promoting skin cancers. This carcinogenic effect is due to the action of UV-A on various endogenous photosensitizers to produce reactive oxygen species (ROS), which can damage both the DNA and its cellular repair machinery. In contrast to UV-A, UV-B can only penetrate through the epidermis and not the dermis. UV-B exposure is biologically more active by causing sunburn and other physiological changes in the skin, which includes the delayed and more long-lasting tanning. The intensity of UV-B radiation (unlike UV-A) vastly varies depending upon geographic location, time, and season. One of the prominent targets of UV-B is cellular DNA, which can absorb UV-B radiation leading to the formation of cyclobutane pyrimidine dimers [12], (6-4) photoproducts [13] and other lesions in the DNA. In addition, UV-B causes significant adverse effects on organisms such as bacteria [14,15], cyanobacteria, phytoplankton, algae [16], plants [17], animals and humans [18]. Some organisms have evolutionary developed UV-absorbing pigments as the first line of defense, though they cannot prevent UVR completely from reaching the DNA in superficial tissues [19]. DNA repair systems, such as nucleotide excision repair, superoxide dismutase (SOD), catalase (CAT) and peroxidase (POD), and vitamins, such as B, C, D and E, play a protective role against UV-B [19]. Vitamin D3 (cholecalciferol) is mainly generated in the skin as a response to UVR and is further converted to the principal biologically active form 1, 25 (OH)2D3 with the help of various enzymes. Vitamin D is one of the most important nutrients for optimal health and wellbeing. It is known to have a variety of potential benefits such as prevention of cancers and autoimmune diseases [20,21], reduction of high blood pressure [22], positive regulation of immune system [23] and antimicrobial activity [24,25,26]. UVR also causes systemic effects such as increasing blood concentrations of inflammatory (IL1β, TNF-α) and metabolic (low/high density lipoproteins) factors [27,28]. UVR impacts the skin-brain axis by releasing neuro-endocrine-immune factors such as β-endorphin and corticotropin-releasing hormones from the skin into the blood circulation. Then, these factors exert endocrine and opioidergic effects on the central nervous system [29]. Indeed, behavioral effects of therapies using UVR are described. Psoralen used together with UV-A (PUVA) treatment in early-stage mycosis fungoides seems to have a positive impact on patient’s quality of life and psychological wellbeing [30]. This opioidergic effect is thought be the result of UV-induced production of endorphins (which modulate itch, pain and reduce stress) by interacting with the neuroendocrine system [31,32,33,34]. Moreover, UVR is reported to induce the production of endocannabinoids in serum [35]. Together, those mediators may favor and support psychologic wellbeing. Furthermore, recent study shows a striking correlation between serum concentrations of 25(OH)D levels and abundance of various microbes in the gut, thereby suggesting an interplay between UVR exposure and the skin-gut axis [36]. Effects of UVR on the local and systemic levels have been very well reviewed elsewhere [29,37,38,39]. This article will focus on the potential of skin microbiome and pro- and prebiotics on local responses to UVR exposure.

2. Perspectives

2.1. UVR-induced Pigmentation and Antioxidation

Skin pigmentation has long been believed to be the most important photoprotective factor, with melanin (especially eumelanin) functioning both as broadband UVR absorbent (of 50–70% of UVR) and possessing antioxidant and radical scavenging properties [40]. The underlying regulatory mechanisms leading to pigmentation are not well understood. Extensive data, however, suggests that UVR-induced DNA damage initiates signals that increases the melanogenesis. Melanin is produced in dendritic melanocytes (in specific ovoid organelles knowns as melanosomes), which account for mere 1% of the epidermal cells. Melanogenesis is regulated via various mechanisms such as transcriptional regulation, intracellular signal transduction pathways with cAMP responsive element being the critical factor [41] and reaction substrates such L-tyrosine, L-dihydroxyphenylalanine (L-DOPA), which also serve as bioregulators of melanogenesis [42,43]. Interestingly, the melanin precursors L-tyrosine and L-DOPA serve an additional role as hormonelike bioregulators, which is governed by melanocytes [44]. Darker skin contains more eumelanin than fair skin and is better protected against UV-induced skin damage. Furthermore, the epidermis of the darker skin allows only 7.4% of UV-B and 17.5% of UV-A to penetrate, in contrast to 24% UV-B and 55% UV-A that passes through fair (white) skin [45,46]. The capacity of several microbial strains to produce melanin could be relevant for skin health [47]. For instance, melanin harvested from cultures of Streptomyces glaucescens [48] exhibits anti-proliferative effects on human fibroblasts [23]. Melanin or melanin-like pigments can also be produced by fungi, but as their melanin production goes together with tissue invasion, their transplantation onto the skin might trigger an infection [49,50]. Fungi from the malassezia genus could be an exception, as some species can cause pityriasis versicolor, a condition with hyper- or hypopigmented skin patches, depending on the background color of the skin, while some others colonize the skin without symptoms [51,52]. Bacteria and fungi produce melanin as protection against UVR, solar or gamma radiation. For instance, melanization protects fungi such as Cladosporium spp. [53], Sporothrix Schenckii [54] and Cryptococcus neoformans [55] from UVR. With increasing evidence of UV-induced damage to skin microbiome, one could envision to use melanin as photoprotection not only for the skin, but also for the resident cutaneous microbes. Escherichia and Enterococcus species can produce serotonin, which is involved in skin pigmentation [56,57]. Indeed, inducing the production of melanin or transferring this capacity to non-melanogenic microbes using genetic modification has been discussed [58]. Interestingly, recombinant strains of Bacillus thuringiensis and Pseudomonas putida, which could produce increased levels of melanin, showed higher survival rate and resistance to UVR exposure [59,60]. Such (recombinant) microbes capable of producing and/or inducing melanin could be used for preventing the damaging effects of UVR on the resident microbiome and beyond. Apart from melanin, microorganisms can produce other compounds such as a large array of antioxidants [61]. Malassezia furfur, a human skin saprophyte, can produce pityriacitrin, a UV-absorbing indole alkaloid that protects the colonies from UVR [62]. The skin colonization with selected malassezia strains optimized for pityriacitrin production could have a clinical benefit in term of UV protection. On the other hand, toxins of Malassezia furfur may also lead to loss of pigment in the skin by complex mechanisms, including production of toxins such as dicarboxylic acids and inflammatory response [63,64]. Lastly, on the more classical probiotic front, the topical use of Lactobacillus helveticus supernatant on the skin has antioxidant effects on rodents [65]. Therefore, a novel approach to safely modulate pigmentation by microbes, or utilizing their antioxidation properties, could be beneficial for cutaneous photoprotection and preventing skin cancers (Figure 1).

2.2. Protection from Photoaging

The skin consists of a complex network of cells and structures connecting cutaneous nerves, local neuroendocrine elements and the immune system [66,67]. UVR exposure accounts for 80% of the visible signs of skin aging, which includes dryness, scalping, wrinkling, disturbed pigmentation and solar freckles [67,68]. Multiple studies have shown that long-term exposure to UV-A leads to photoaging and free radical production, by damaging the dermis more significantly than UV-B [66,69]. The skin’s response to external stress such as UVR involves coordination between various local and systemic responses, which are mediated by the skin neuroendocrine system [70,71]. Photoaging is known to correlate with cancer risk, as for instance shown in a study from Central Europe where subjects with early signs of wrinkling on the neck were over four times more susceptible to melanoma compared to subjects from general population. Furthermore, subjects with skin freckling on the back also showed over three times the risk of developing melanoma (compared to control population) [68]. The risk of developing melanoma is known to correlate with intermittent UV exposure [68,72]. That said, the skin microbiome is known to change overtime, and is largely influenced by chronological and physiological skin aging [73]. Moreover, UVR is known to cause local (skin) [74] and systemic (gut) [36,75] changes in the microbial landscape. With increasing understanding of multidirectional interplay between gut microbiome and neuroendocrine system (microbiome-gut-brain axis) [76], it is tempting to speculate on its role in photoaging.
Probiotics are emerging as potential therapeutic means to mitigate the damaging effects of UVR on the skin. Oral administration of Bifidobacterium breve in hairless mice prevented UVR-induced transepidermal water loss and suppressed UV-induced increase in hydrogen peroxide levels, oxidation of proteins and xanthine oxidase activity in the skin (Figure 1) [42]. Studies involving humans have also substantiated the role of probiotics in attenuating UV-induced skin damage. The probiotic Lactobacillus johnsonii was orally administered to UVR exposed-subjects and prevented the UVR-induced decrease in Langerhans cells and improved the recovery of immune homeostasis [77]. Other probiotics such as Faecalibacterium prausnitzii [45,46], Lactobacillus plantarum [47], Bifidobacterium Breve Strain Yakult [48], Bifidobacterium longum [47] and Lactobacillus plantarum HY7714 [78] are known to have beneficial effects on photoaging in humans and mice. In depth reviews of probiotics in photoaging have been published elsewhere [72,79,80]. Moreover, orally administered prebiotics such as oligosaccharides are known to prevent transepidermal water loss, reduce erythema and prevent skin damage [81].

2.3. Anti-Tumor Effects of the Microbiome

UVR exposure has been associated with the development of different types of skin cancers. Chronic cumulative exposure is commonly associated with basal cell carcinoma (BCC) and squamous cell carcinoma (SCC). SCC can develop from actinic keratosis (AK), which is a typical lesion of photodamaged skin. Interestingly a recent study has shown higher relative abundances of commensal strains such as Propionibacterium and Malassezia in non-lesional skin compared to tissue of AK and SCC skin lesions. Staphylococcus aureus was significantly more abundant in lesional AK and SCC skin than non-lesional skin [53,54,55]. Moreover, Staphylococcus aureus, Chlamydophila pneumoniae and Borrelia burgdorferi have been associated with cutaneous T- or B-cell lymphoma [82,83]. Although studies on the role of skin microbiome in cancers is still in its infancy, initial reports, however, suggest an altered microbial landscape in cancerous skin that could be partly playing a role in disease pathogenesis and/or progression. Skin resident T cells play a role in the defense against formation and recurrence of skin neoplasia [84]. As the presence of bacteria on the skin surface can alter its T cell population [85], it is tempting to speculate that topical application of certain bacterial strains could act as secondary treatment against skin neoplasms. Skin exposure to UVR triggers inflammation [86] and studies suggests local (skin) and systemic (intestine) changes in microbial landscape after UVR exposure [36]. UVR-induced impairments of the immune system reduce the capacity of the host to reject skin cancers, and rather promote carcinogenesis. Moreover, microbial exposure plays a crucial role in cancer immunobiology by limiting chronic inflammation in early stages [87] and any disruptions of homeostatic skin microbiome could initiate inflammatory mechanisms that may lead to carcinogenesis. Overall, the response to UVR in the presence of microbiome on mice skin held in SPF conditions, compared to germ-free mice seem to be pro-inflammatory and protective [10]. Thus, the modification of certain strains could potentially be useful in fighting the development of cancerous cells locally. For instance, the colonization of mouse skin by strains of S. epidermidis that produce the antiproliferative agent 6-N-hydroxyaminopurine (6-HAP) diminishes the incidence of UV-induced skin neoplasms [88]. The systemic use of probiotics to control UV-B-induced immunosuppression has also gained interest, and oral intake of lipoteichoic acid from Lactobacillus rhamnosus decreased the number of UV-induced skin tumor in SKH-1 hairless mice [89]. Prebiotics, capable of inducing or limiting the growth of certain microbes, are now considered to modulate the growth of certain pathogenic skin microbes. Oral intake of prebiotics such as inulin and mucin have been reported to induce Bifidobacterium spp. [90] and Akkermansia muciniphila [91,92], which are involved in inhibiting melanoma growth [93]. Addition of inulin and mucin to the diet induces anti-tumor immune responses and inhibits subcutaneously implanted BRAF mutant melanoma in a syngeneic mouse model [94]. There is a growing body of evidence that microbiome enhances the effectiveness of chemotherapy, radiotherapy and immunotherapy [95,96]. However, the interplay between photo(chemo)therapy, used to treat various inflammatory diseases such as psoriasis, atopic eczema, lichen planus and graft-versus-host disease (GvHD) as well as neoplastic diseases such as cutaneous T cell lymphoma and other immunoproliferative conditions, and the local microbial environment needs to be addressed. Finally, recent reports have shown that antibiotic treatment or selective enrichment of certain microbial species in the gut can indeed limit the effectiveness of immune check point therapy in cutaneous melanoma [97,98]. Interestingly, a recent study demonstrated targeted depletion of bacteria from a mixed population by programmed inhibitor cells that can direct the antibacterial activity against specified target cells using the type VI secretion system (T6SS) [99]. Such an approach where topical treatment selectively enriches skin commensals or targets specified microbes (pathogenic), known to be involved in immune responses [100], could also be envisioned for modulating UVR-induced immune suppression and the efficacy of phototherapy (Figure 1).

2.4. Enhancement of the UVR-induced Immune Suppression

UVR has a profound effect on the skin’s immune system, which can manifest both locally and systemically in a dose-dependent manner. The role of UVR in modulating adaptive immunity was shown in landmark studies by Kripke et al., where the mice exposed to sub-carcinogenic doses of UVR developed skin tumors due to further suppression of the anti-tumor immune response [101]. Several subsequent studies in mice and humans established that UVR-induced immune suppression was a result of activation of suppressor T cells (now called as regulatory T cells (Tregs)), modulation in the number and function of antigen presenting cells, activation and recruitment of neutrophils and increased expression of anti-inflammatory cytokines such as IL-4 and IL-10 [37]. The role of skin microbiome in UVR-induced immune suppression has also now been addressed. We have recently shown, using the contact hypersensitivity (CHS) model, that absence of microbiome enhanced UVR-induced immune suppression in mice lacking microbiome (germ-free), with predominant expression of anti-inflammatory cytokines such as IL-10, and increased numbers of monocytes/macrophages in the skin. In contrast, mice with a microbiome showed diminished UVR-induced immune suppression with higher expression of pro-inflammatory cytokines such as IL-1β, epidermal hyperplasia and neutrophilic infiltration [10]. Interestingly, administration of fermented milk containing Lactobacillus casei DN-114 001 in a CHS model reduced skin inflammation by inhibiting the hapten-specific CD8+ T cells [102], and enhancing the frequency of FoxP3+ regulatory T cells and IL-10 producing CD4+CD25+ effector T cells [103]. These studies in T cell-mediated allergic skin inflammation model demonstrate that certain microbes or pro-biotics can indeed reduce severity of the disease by regulating the immune response (Figure 1). Other skin commensals could show similar effects, which remains to be addressed.

2.5. Protection in UVR-induced Skin Inflammation

Several skin diseases are triggered by UVR, such as cutaneous lupus [104], or polymorphic light eruption (PLE) [105]. In cutaneous lupus, UVR exposure induces long-lasting molecular changes in keratinocytes, including increased inducible nitric oxide (NO) synthase (iNOS) and interferon-stimulated transcripts [106]. For these diseases, the use of topical anti-inflammatory probiotics could be beneficial. For instance, Lactobacillus reuteri showed anti-inflammatory properties on a reconstructed human epidermis upon UVR-induced inflammation [107]. Oral intake of fermented milk, exopolysaccharide or Lactobacillus planterum in mice reduced UVR-induced epidermal thickness and activity of metalloprotease [108]. That said, oral administration of probiotic supplements containing lycopene, beta-carotene and Lactobacillus johnsonii [109] or natural extracts from fern leaves, such as Polypodium leucotomos, has been previously described to prevent and/or diminish PLE symptoms [110,111]. Patients with PLE have been reported to have low levels of vitamin D, due to sun avoidance. UVR-induced vitamin D and its metabolites participate in various immune pathways [112], leading to immune suppression by influencing innate and adaptive immune cells [113]. High doses of vitamin D3 can also alter the microbial composition in the gut [114]. Recently, it has been shown that exposure to narrow band UV-B increases serum vitamin D levels, which then modulates intestinal microbiome [36]. In this context, PLE pathogenesis, which is (hypothetically) linked with altered skin microbiome or microbial elements, could be treated/prevented using vitamin D supplements [105]. A clinical study has in fact shown that topical pre-treatment with a 1,25-dihydroxyvitamin D3 analogue (calcipotriol) significantly reduced photo-provoked PLE symptoms [115]. Though the mechanism of this has remained elusive, this finding might be partly due to microbial modulation by calcipotriol and/or its metabolites. Moreover, UVR-induced vitamin D3 can help “program” the migration of T cells within the skin [25].

2.6. Controlling Inflammatory Diseases in UV-exposed Skin Areas

Acne, rosacea and seborrheic dermatitis are an interesting group of mostly facial diseases that occur in UVR-exposed skin, but in which no direct effect of UVR is proven in the pathophysiology. It is not known whether the UVR effects on skin possibly participate in an inappropriate immune response to local microbes. UV-B triggers inflammasome activation, which is potentialized by the presence of the antimicrobial peptide LL-37 [116]. LL-37 is produced by the skin following the exposure to P. acnes and Malassezia [117], and is induced by vitamin D in sebocytes [118], which could be a mechanism through which saprophytes might become proinflammatory on UV-exposed areas. In this context, microbes with anti-inflammatory effect could be of interest. In mice, the application of the short chain fatty acid (succinic acid) produced by S. epidermidis onto ears injected with P. acnes decreases local inflammation [119]. In humans, the scarcity of randomized controlled trials of pre- and probiotics in facial dermatosis makes it difficult to draw any conclusions. Among the few randomized trials, the local application of Vitreoscilla filiformis decreased the clinical score in seborrheic dermatitis in one trial [120]. In mice, this bacterium impacted dendritic cells and induced IL-10+ regulatory T cells and reduced AD-like inflammation [121]. In many other skin diseases, such as psoriasis and eczema, the exposure to natural light or to UVA/B is, however, used as a treatment. For these diseases, the question is merely if the use of anti-inflammatory pre- or probiotics could potentiate the benefit [122]. Hence, in the last decade, elegant mice data have shown that the skin microbiome has been able to modify local T cell population and poise its function [85,123]. As T cells can dwell and persist in the skin after infections [124,125], using anti-inflammatory bacteria to maintain the balance of local pool of cutaneous immune cells will hopefully open interesting perspectives for better protection against UVR (Figure 1). Moreover, skin microbiome transplantation (live-biotherapeutic approach) has been successfully demonstrated to reduce atopic dermatitis (AD) symptoms. Culturable gram-negative bacteria (Roseomonas mucosa) collected from healthy humans were associated with activating the innate immune system, enhanced the barrier function and controlled the growth of S. aureus in AD-like mouse model (MC903) [126]. This approach of topical application of live-biotherapeutics also successfully reduced AD disease severity and S. aureus burden in AD patients [127]. A similar approach of using live-biotherapeutics should be envisioned to control UV-induced inflammatory diseases.

3. Conclusions and Outlook

Overall, probiotics and prebiotics are promising in protecting the skin against UVR-induced skin damage. On the other side of the spectrum, pro- and prebiotics may also modulate the efficacy of phototherapy. With growing knowledge of the human microbiome and patients’ unique microbial “fingerprint,” personalized oral or skin care regimen should be envisioned using special formulations to promote commensal microbiome that can protects against UVR and suppress growth of pathogens. Using live microbes pose certain side effects such as infection, deleterious metabolic activity, excessive immune stimulation, and dysregulation in gene expression in susceptible individuals [128,129,130]. On the prebiotic front, although they are well tolerated, undesirable effects after oral administration may occur, with allergic reactions and abdominal discomfort, and even potential distant effects on the skin [129,131]. To succeed in using probiotics and prebiotics, many questions need to be answered: what is the best route of administration of a supplementation? Will a combination approach of pro- or prebiotic supplementation be most effective? Are live microbes more advantageous than using pro- or pre-biotics, and how do we successfully implant them without side-effects? To address these issues, further studies are warranted that will enhance our understanding of the potential of skin microbiome, pro- and pre-biotics and help in developing novel strategies to control the cutaneous interplay with UVR.

Author Contributions

Conceptualization, V.P. and I.G.S.; writing—original draft preparation, V.P and I.G.S.; writing—review and editing, P.W.; visualization, V.P.; All authors have read and agreed to the published version of the manuscript.

Funding

V.P. was supported by the Austrian Science Fund FWF (W1241) and the Medical University of Graz through the Ph.D. Program Molecular Fundamentals of Inflammation (DK-MOLIN); funding from Scientific Association of Styrian Dermatology and City of Graz, Graz, Austria.

Acknowledgments

We thank Honnavara N. Ananthaswamy, Houston, TX, for critical reading and editing of the manuscript. Open Access Funding by the Austrian Science Fund (FWF).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Grice, E.A.; Kong, H.H.; Renaud, G.; Young, A.C.; Program, N.C.S.; Bouffard, G.G.; Blakesley, R.W.; Wolfsberg, T.G.; Turner, M.L.; Segre, J.A. A diversity profile of the human skin microbiota. Genome Res. 2008, 18, 1043–1050. [Google Scholar] [CrossRef] [Green Version]
  2. Gilbert, J.A.; Blaser, M.J.; Caporaso, J.G.; Jansson, J.K.; Lynch, S.V.; Knight, R. Current understanding of the human microbiome. Nat. Med. 2018, 24, 392–400. [Google Scholar] [CrossRef]
  3. Probst, A.J.; Auerbach, A.K.; Moissl-Eichinger, C. Archaea on human skin. PLoS ONE 2013, 8, e65388. [Google Scholar] [CrossRef] [Green Version]
  4. Lacey, N.; Ni Raghallaigh, S.; Powell, F.C. Demodex mites--commensals, parasites or mutualistic organisms? Dermatology 2011, 222, 128–130. [Google Scholar] [CrossRef]
  5. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The human skin microbiome. Nat. Rev. Microbiol.. 2018, 16, 143–155. [Google Scholar] [CrossRef]
  6. Costello, E.K.; Lauber, C.L.; Hamady, M.; Fierer, N.; Gordon, J.I.; Knight, R. Bacterial community variation in human body habitats across space and time. Science 2009, 326, 1694–1697. [Google Scholar] [CrossRef] [Green Version]
  7. 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, 3575–3581. [Google Scholar] [CrossRef] [Green Version]
  8. 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]
  9. 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, 20. [Google Scholar] [CrossRef]
  10. Patra, V.; Wagner, K.; Arulampalam, V.; Wolf, P. Skin Microbiome Modulates the Effect of Ultraviolet Radiation on Cellular Response and Immune Function. iScience 2019, 15, 211–222. [Google Scholar] [CrossRef] [Green Version]
  11. Hoffmann, K.; Kaspar, K.; Altmeyer, P.; Gambichler, T. UV transmission measurements of small skin specimens with special quartz cuvettes. Dermatology 2000, 201, 307–311. [Google Scholar] [CrossRef] [PubMed]
  12. Setlow, R.B.; Carrier, W.L. Pyrimidine dimers in ultraviolet-irradiated DNA’s. J. Mol. Biol. 1966, 17, 237–254. [Google Scholar] [CrossRef]
  13. Mitchell, D.L.; Nairn, R.S. The biology of the (6–4) photoproduct. Photochem. Photobiol. 1989, 49, 805–819. [Google Scholar] [CrossRef]
  14. Peak, M.J.; Peak, J.G. Single-strand breaks induced in Bacillus subtilis DNA by ultraviolet light: Action spectrum and properties. Photochem. Photobiol. 1982, 35, 675–680. [Google Scholar] [CrossRef]
  15. Peak, M.J.; Peak, J.G.; Moehring, M.P.; Webb, R.B. Ultraviolet action spectra for DNA dimer induction, lethality, and mutagenesis in Escherichia coli with emphasis on the UVB region. Photochem. Photobiol. 1984, 40, 613–620. [Google Scholar] [CrossRef]
  16. Rozema, J.; Bjorn, L.O.; Bornman, J.F.; Gaberscik, A.; Hader, D.P.; Trost, T.; Germ, M.; Klisch, M.; Groniger, A.; Sinha, R.P.; et al. The role of UV-B radiation in aquatic and terrestrial ecosystems--an experimental and functional analysis of the evolution of UV-absorbing compounds. J. Photochem. Photobiol. B 2002, 66, 2–12. [Google Scholar] [CrossRef]
  17. Quaite, F.E.; Sutherland, B.M.; Sutherland, J.C. Quantitation of pyrimidine dimers in DNA from UVB-irradiated alfalfa (Medicago sativa L.) seedlings. Appl. Theor. Electrophor. 1992, 2, 171–175. [Google Scholar]
  18. Sinha, R.P.; Hader, D.P. UV-induced DNA damage and repair: A review. Photochem. Photobiol. Sci. 2002, 1, 225–236. [Google Scholar] [CrossRef]
  19. Xie, Z.; Wang, Y.; Liu, Y.; Liu, Y. Ultraviolet-B exposure induces photo-oxidative damage and subsequent repair strategies in a desert cyanobacterium Microcoleus vaginatus Gom. Eur. J. Soil Boil. 2009, 45, 377–382. [Google Scholar] [CrossRef]
  20. Ponsonby, A.L.; Lucas, R.M.; van der Mei, I.A. UVR, vitamin D and three autoimmune diseases--multiple sclerosis, type 1 diabetes, rheumatoid arthritis. Photochem. Photobiol. 2005, 81, 1267–1275. [Google Scholar] [CrossRef]
  21. Grant, W.B.; Garland, C.F.; Holick, M.F. Comparisons of estimated economic burdens due to insufficient solar ultraviolet irradiance and vitamin D and excess solar UV irradiance for the United States. Photochem. Photobiol. 2005, 81, 1276–1286. [Google Scholar] [CrossRef] [PubMed]
  22. Pilz, S.; Tomaschitz, A.; Ritz, E.; Pieber, T.R. Vitamin D status and arterial hypertension: A systematic review. Nat. Rev. Cardiol. 2009, 6, 621–630. [Google Scholar] [CrossRef] [PubMed]
  23. Bell, E. Vitamin D3 promotes immune function in the skin. Nat. Rev. Immunol. 2007, 7, 174–175. [Google Scholar] [CrossRef]
  24. Liu, P.T.; Stenger, S.; Li, H.; Wenzel, L.; Tan, B.H.; Krutzik, S.R.; Ochoa, M.T.; Schauber, J.; Wu, K.; Meinken, C.; et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 2006, 311, 1770–1773. [Google Scholar] [CrossRef]
  25. Sigmundsdottir, H.; Pan, J.; Debes, G.F.; Alt, C.; Habtezion, A.; Soler, D.; Butcher, E.C. DCs metabolize sunlight-induced vitamin D3 to ’program’ T cell attraction to the epidermal chemokine CCL27. Nat. Immunol. 2007, 8, 285–293. [Google Scholar] [CrossRef]
  26. Schauber, J.; Dorschner, R.A.; Coda, A.B.; Buchau, A.S.; Liu, P.T.; Kiken, D.; Helfrich, Y.R.; Kang, S.; Elalieh, H.Z.; Steinmeyer, A.; et al. Injury enhances TLR2 function and antimicrobial peptide expression through a vitamin D-dependent mechanism. J. Clin. Investig. 2007, 117, 803–811. [Google Scholar] [CrossRef] [Green Version]
  27. Banerjee, S.; Leptin, M. Systemic response to ultraviolet radiation involves induction of leukocytic IL-1beta and inflammation in zebrafish. J. Immunol. 2014, 193, 1408–1415. [Google Scholar] [CrossRef] [Green Version]
  28. Gorman, S.; de Courten, B.; Lucas, R.M. Systematic Review of the Effects of Ultraviolet Radiation on Markers of Metabolic Dysfunction. Clin. Biochem. Rev. 2019, 40, 147–162. [Google Scholar] [CrossRef]
  29. Slominski, A.T.; Zmijewski, M.A.; Plonka, P.M.; Szaflarski, J.P.; Paus, R. How UV Light Touches the Brain and Endocrine System Through Skin, and Why. Endocrinology 2018, 159, 1992–2007. [Google Scholar] [CrossRef] [Green Version]
  30. Graier, T.; Fink-Puches, R.; Porkert, S.; Lang, R.; Pöchlauer, S.; Ratzinger, G.; Tanew, A.; Selhofer, S.; Sator, P.-G.; Hofer, A.; et al. Quality of life, anxiety and depression in patients with early-stage mycosis fungoides and the effect of oral psoralen plus UV-A (PUVA) photochemotherapy on it. Front. Med. 2020, 7, 330. [Google Scholar] [CrossRef]
  31. Jozic, I.; Stojadinovic, O.; Kirsner, R.S.F.; Tomic-Canic, M. Skin under the (Spot)-Light: Cross-Talk with the Central Hypothalamic-Pituitary-Adrenal (HPA) Axis. J. Investig. Dermatol. 2015, 135, 1469–1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Fell, G.L.; Robinson, K.C.; Mao, J.; Woolf, C.J.; Fisher, D.E. Skin beta-endorphin mediates addiction to UV light. Cell 2014, 157, 1527–1534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tejeda, H.A.; Bonci, A. Shedding "UV" light on endogenous opioid dependence. Cell 2014, 157, 1500–1501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Bigliardi, P.L.; Dancik, Y.; Neumann, C.; Bigliardi-Qi, M. Opioids and skin homeostasis, regeneration and ageing—What’s the evidence? Exp. Dermatol. 2016, 25, 586–591. [Google Scholar] [CrossRef]
  35. Felton, S.J.; Kendall, A.C.; Almaedani, A.F.; Urquhart, P.; Webb, A.R.; Kift, R.; Vail, A.; Nicolaou, A.; Rhodes, L.E. Serum endocannabinoids and N-acyl ethanolamines and the influence of simulated solar UVR exposure in humans in vivo. Photochem. Photobiol. Sci. 2017, 16, 564–574. [Google Scholar] [CrossRef] [Green Version]
  36. Bosman, E.S.; Albert, A.Y.; Lui, H.; Dutz, J.P.; Vallance, B.A. Skin Exposure to Narrow Band Ultraviolet (UVB) Light Modulates the Human Intestinal Microbiome. Front. Microbiol. 2019, 10, 2410. [Google Scholar] [CrossRef]
  37. Bernard, J.J.; Gallo, R.L.; Krutmann, J. Photoimmunology: How ultraviolet radiation affects the immune system. Nat. Rev. Immunol. 2019, 19, 688–701. [Google Scholar] [CrossRef]
  38. Yu, Z.; Wolf, P. How It Works: The Immunology Underlying Phototherapy. Dermatol. Clin. 2020, 38, 37–53. [Google Scholar] [CrossRef]
  39. Vieyra-Garcia, P.A.; Wolf, P. From Early Immunomodulatory Triggers to Immunosuppressive Outcome: Therapeutic Implications of the Complex Interplay Between the Wavebands of Sunlight and the Skin. Front. Med. 2018, 5, 232. [Google Scholar] [CrossRef]
  40. Brenner, M.; Hearing, V.J. The protective role of melanin against UV damage in human skin. Photochem. Photobiol. 2008, 84, 539–549. [Google Scholar] [CrossRef] [Green Version]
  41. Romero-Graillet, C.; Aberdam, E.; Biagoli, N.; Massabni, W.; Ortonne, J.P.; Ballotti, R. Ultraviolet B radiation acts through the nitric oxide and cGMP signal transduction pathway to stimulate melanogenesis in human melanocytes. J. Biol. Chem. 1996, 271, 28052–28056. [Google Scholar] [CrossRef] [Green Version]
  42. Slominski, A.; Paus, R. Towards defining receptors for L-tyrosine and L-dopa. Mol. Cell Endocrinol. 1994, 99, C7–C11. [Google Scholar] [CrossRef]
  43. Slominski, A.; Tobin, D.J.; Shibahara, S.; Wortsman, J. Melanin pigmentation in mammalian skin and its hormonal regulation. Physiol. Rev. 2004, 84, 1155–1228. [Google Scholar] [CrossRef] [PubMed]
  44. Slominski, A.; Paus, R. Are L-tyrosine and L-dopa hormone-like bioregulators? J. Theor. Biol. 1990, 143, 123–138. [Google Scholar] [CrossRef]
  45. Heymann, W.R. Skin cancer in African Americans. J. Am. Acad. Dermatol. 2005, 53, 485–486. [Google Scholar] [CrossRef] [PubMed]
  46. Halder, R.M.; Bridgeman-Shah, S. Skin cancer in African Americans. Cancer 1995, 75, 667–673. [Google Scholar] [CrossRef]
  47. Rastogi, R.P.; Incharoensakdi, A. Analysis of UV-absorbing photoprotectant mycosporine-like amino acid (MAA) in the cyanobacterium Arthrospira sp. CU2556. Photochem. Photobiol. Sci. 2014, 13, 1016–1024. [Google Scholar] [CrossRef]
  48. El-Naggar, N.E.; El-Ewasy, S.M. Bioproduction, characterization, anticancer and antioxidant activities of extracellular melanin pigment produced by newly isolated microbial cell factories Streptomyces glaucescens NEAE-H. Sci. Rep. 2017, 7, 42129. [Google Scholar] [CrossRef]
  49. Chongkae, S.; Nosanchuk, J.D.; Pruksaphon, K.; Laliam, A.; Pornsuwan, S.; Youngchim, S. Production of melanin pigments in saprophytic fungi in vitro and during infection. J. Basic Microbiol. 2019, 59, 1092–1104. [Google Scholar] [CrossRef]
  50. Youngchim, S.; Pornsuwan, S.; Nosanchuk, J.D.; Dankai, W.; Vanittanakom, N. Melanogenesis in dermatophyte species in vitro and during infection. Microbiology 2011, 157, 2348–2356. [Google Scholar] [CrossRef] [Green Version]
  51. Morishita, N.; Sei, Y.; Takiuchi, I.; Sugita, T. Examination of the causative agent of pityriasis versicolor. Nihon. Ishinkin Gakkai Zasshi 2005, 46, 169–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Findley, K.; Program, N.I.S.C.C.S.; Oh, J.; Yang, J.; Conlan, S.; Deming, C.; Meyer, J.A.; Schoenfeld, D.; Nomicos, E.; Park, M.; et al. Topographic diversity of fungal and bacterial communities in human skin. Nature 2013, 498, 367–370. [Google Scholar] [CrossRef] [PubMed]
  53. Zhdanova, N.N.; Gavriushina, A.I.; Vasilevskaia, A.I. Effect of gamma and UV irradiation on the survival of Cladosporium sp. and Oidiodendron cerealis. Mikrobiol. Zhurnal 1973, 35, 449–452. [Google Scholar]
  54. Romero-Martinez, R.; Wheeler, M.; Guerrero-Plata, A.; Rico, G.; Torres-Guerrero, H. Biosynthesis and functions of melanin in Sporothrix schenckii. Infect. Immun. 2000, 68, 3696–3703. [Google Scholar] [CrossRef] [Green Version]
  55. Wang, Y.; Casadevall, A. Decreased susceptibility of melanized Cryptococcus neoformans to UV light. Appl. Environ. Microbiol. 1994, 60, 3864–3866. [Google Scholar] [CrossRef] [Green Version]
  56. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef]
  57. Lee, H.J.; Park, M.K.; Kim, S.Y.; Park Choo, H.Y.; Lee, A.Y.; Lee, C.H. Serotonin induces melanogenesis via serotonin receptor 2A. Br. J. Dermatol. 2011, 165, 1344–1348. [Google Scholar] [CrossRef]
  58. Martinez, L.M.; Martinez, A.; Gosset, G. Production of Melanins With Recombinant Microorganisms. Front. Bioeng. Biotechnol. 2019, 7, 285. [Google Scholar] [CrossRef]
  59. Ruan, L.; Yu, Z.; Fang, B.; He, W.; Wang, Y.; Shen, P. Melanin pigment formation and increased UV resistance in Bacillus thuringiensis following high temperature induction. Syst. Appl. Microbiol. 2004, 27, 286–289. [Google Scholar] [CrossRef] [Green Version]
  60. Nikodinovic-Runic, J.; Martin, L.B.; Babu, R.; Blau, W.; O’Connor, K.E. Characterization of melanin-overproducing transposon mutants of Pseudomonas putida F6. FEMS Microbiol. Lett. 2009, 298, 174–183. [Google Scholar] [CrossRef] [Green Version]
  61. Peyrat, L.A.; Tsafantakis, N.; Georgousaki, K.; Ouazzani, J.; Genilloud, O.; Trougakos, I.P.; Fokialakis, N. Terrestrial Microorganisms: Cell Factories of Bioactive Molecules with Skin Protecting Applications. Molecules 2019, 24, 1836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Mayser, P.; Schafer, U.; Kramer, H.J.; Irlinger, B.; Steglich, W. Pityriacitrin—An ultraviolet-absorbing indole alkaloid from the yeast Malassezia furfur. Arch. Dermatol. Res. 2002, 294, 131–134. [Google Scholar] [CrossRef] [PubMed]
  63. Inamadar, A.C.; Palit, A. The genus Malassezia and human disease. Indian J. Dermatol. Venereol. Leprol. 2003, 69, 265–270. [Google Scholar] [PubMed]
  64. Gupta, P.; Bansal, A.; Singh, A. Resurvey of symptomatics of the Jaipur district population and suggestion for alternative diagnostic criteria of asthma for epidemiological surveys. Pigment. Int. 2014, 1, 32–35. [Google Scholar] [CrossRef] [PubMed]
  65. Rong, J.; Shan, C.; Liu, S.; Zheng, H.; Liu, C.; Liu, M.; Jin, F.; Wang, L. Skin resistance to UVB-induced oxidative stress and hyperpigmentation by the topical use of Lactobacillus helveticus NS8-fermented milk supernatant. J. Appl. Microbiol. 2017, 123, 511–523. [Google Scholar] [CrossRef]
  66. Bocheva, G.; Slominski, R.M.; Slominski, A.T. Neuroendocrine Aspects of Skin Aging. Int. J. Mol. Sci. 2019, 20, 2798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Grant, W.B. The effect of solar UVB doses and vitamin D production, skin cancer action spectra, and smoking in explaining links between skin cancers and solid tumours. Eur. J. Cancer 2008, 44, 12–15. [Google Scholar] [CrossRef]
  68. Wendt, J.; Schanab, O.; Binder, M.; Pehamberger, H.; Okamoto, I. Site-dependent actinic skin damage as risk factor for melanoma in a central European population. Pigment. Cell Melanoma Res. 2012, 25, 234–242. [Google Scholar] [CrossRef]
  69. Marionnet, C.; Tricaud, C.; Bernerd, F. Exposure to non-extreme solar UV daylight: Spectral characterization, effects on skin and photoprotection. Int. J. Mol. Sci. 2014, 16, 68–90. [Google Scholar] [CrossRef]
  70. Slominski, A.; Wortsman, J. Neuroendocrinology of the skin. Endocr. Rev. 2000, 21, 457–487. [Google Scholar] [CrossRef]
  71. Slominski, A.T.; Zmijewski, M.A.; Skobowiat, C.; Zbytek, B.; Slominski, R.M.; Steketee, J.D. Introduction. In Sensing the Environment: Regulation of Local and Global Homeostasis by the Skin’s Neuroendocrine System; Springer: Berlin/Heidelberg, Germany, 2012; pp. 1–6. [Google Scholar] [CrossRef]
  72. Amaro-Ortiz, A.; Yan, B.; D’Orazio, J.A. Ultraviolet radiation, aging and the skin: Prevention of damage by topical cAMP manipulation. Molecules 2014, 19, 6202–6219. [Google Scholar] [CrossRef] [PubMed]
  73. Shibagaki, N.; Suda, W.; Clavaud, C.; Bastien, P.; Takayasu, L.; Iioka, E.; Kurokawa, R.; Yamashita, N.; Hattori, Y.; Shindo, C.; et al. Aging-related changes in the diversity of women’s skin microbiomes associated with oral bacteria. Sci. Rep. 2017, 7, 10567. [Google Scholar] [CrossRef]
  74. Burns, E.M.; Ahmed, H.; Isedeh, P.N.; Kohli, I.; Van Der Pol, W.; Shaheen, A.; Muzaffar, A.F.; Al-Sadek, C.; Foy, T.M.; Abdelgawwad, M.S.; et al. Ultraviolet radiation, both UVA and UVB, influences the composition of the skin microbiome. Exp. Dermatol. 2019, 28, 136–141. [Google Scholar] [CrossRef] [PubMed]
  75. Ghaly, S.; Kaakoush, N.O.; Lloyd, F.; Gordon, L.; Forest, C.; Lawrance, I.C.; Hart, P.H. Ultraviolet Irradiation of Skin Alters the Faecal Microbiome Independently of Vitamin D in Mice. Nutrients 2018, 10, 1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Farzi, A.; Frohlich, E.E.; Holzer, P. Gut Microbiota and the Neuroendocrine System. Neurotherapeutics 2018, 15, 5–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Gueniche, A.; Philippe, D.; Bastien, P.; Blum, S.; Buyukpamukcu, E.; Castiel-Higounenc, I. Probiotics for photoprotection. Dermatoendocrinol 2009, 1, 275–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Kim, H.M.; Lee, D.E.; Park, S.D.; Kim, Y.T.; Kim, Y.J.; Jeong, J.W.; Jang, S.S.; Ahn, Y.T.; Sim, J.H.; Huh, C.S.; et al. Oral administration of Lactobacillus plantarum HY7714 protects hairless mouse against ultraviolet B-induced photoaging. J. Microbiol. Biotechnol. 2014, 24, 1583–1591. [Google Scholar] [CrossRef]
  79. Ouwehand, A.C.; Tiihonen, K.; Lahtinen, S. The Potential of Probiotics and Prebiotics for Skin Health. In Textbook of Aging Skin; Farage, M.A., Miller, K.W., Maibach, H.I., Eds.; Springer: Berlin/Heidelberg, Germany, 2010; pp. 799–809. [Google Scholar] [CrossRef]
  80. Kober, M.M.; Bowe, W.P. The effect of probiotics on immune regulation, acne, and photoaging. Int. J. Womens Dermatol. 2015, 1, 85–89. [Google Scholar] [CrossRef] [Green Version]
  81. Hong, K.B.; Jeong, M.; Han, K.S.; Hwan Kim, J.; Park, Y.; Suh, H.J. Photoprotective effects of galacto-oligosaccharide and/or Bifidobacterium longum supplementation against skin damage induced by ultraviolet irradiation in hairless mice. Int. J. Food Sci. Nutr. 2015, 66, 923–930. [Google Scholar] [CrossRef]
  82. Mirvish, J.J.; Pomerantz, R.G.; Falo, L.D., Jr.; Geskin, L.J. Role of infectious agents in cutaneous T-cell lymphoma: Facts and controversies. Clin. Dermatol. 2013, 31, 423–431. [Google Scholar] [CrossRef]
  83. Nguyen, V.; Huggins, R.H.; Lertsburapa, T.; Bauer, K.; Rademaker, A.; Gerami, P.; Guitart, J. Cutaneous T-cell lymphoma and Staphylococcus aureus colonization. J. Am. Acad. Dermatol. 2008, 59, 949–952. [Google Scholar] [CrossRef] [PubMed]
  84. Malik, B.T.; Byrne, K.T.; Vella, J.L.; Zhang, P.; Shabaneh, T.B.; Steinberg, S.M.; Molodtsov, A.K.; Bowers, J.S.; Angeles, C.V.; Paulos, C.M.; et al. Resident memory T cells in the skin mediate durable immunity to melanoma. Sci. Immunol. 2017, 2, eaam6346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Naik, S.; Bouladoux, N.; Wilhelm, C.; Molloy, M.J.; Salcedo, R.; Kastenmuller, W.; Deming, C.; Quinones, M.; Koo, L.; Conlan, S.; et al. Compartmentalized control of skin immunity by resident commensals. Science 2012, 337, 1115–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Hruza, L.L.; Pentland, A.P. Mechanisms of UV-induced inflammation. J. Investig. Dermatol. 1993, 100, 35S–41S. [Google Scholar] [CrossRef] [Green Version]
  87. Gallimore, A.M.; Simon, A.K. Positive and negative influences of regulatory T cells on tumour immunity. Oncogene 2008, 27, 5886–5893. [Google Scholar] [CrossRef] [Green Version]
  88. 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]
  89. Friedrich, A.D.; Campo, V.E.; Cela, E.M.; Morelli, A.E.; Shufesky, W.J.; Tckacheva, O.A.; Leoni, J.; Paz, M.L.; Larregina, A.T.; Gonzalez Maglio, D.H. Oral administration of lipoteichoic acid from Lactobacillus rhamnosus GG overcomes UVB-induced immunosuppression and impairs skin tumor growth in mice. Eur. J. Immunol. 2019, 49, 2095–2102. [Google Scholar] [CrossRef]
  90. Fehlbaum, S.; Prudence, K.; Kieboom, J.; Heerikhuisen, M.; van den Broek, T.; Schuren, F.H.J.; Steinert, R.E.; Raederstorff, D. In Vitro Fermentation of Selected Prebiotics and Their Effects on the Composition and Activity of the Adult Gut Microbiota. Int. J. Mol. Sci. 2018, 19, 3097. [Google Scholar] [CrossRef] [Green Version]
  91. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [Green Version]
  92. Singh, A.; Zapata, R.C.; Pezeshki, A.; Reidelberger, R.D.; Chelikani, P.K. Inulin fiber dose-dependently modulates energy balance, glucose tolerance, gut microbiota, hormones and diet preference in high-fat-fed male rats. J. Nutr. Biochem. 2018, 59, 142–152. [Google Scholar] [CrossRef]
  93. Li, Y.; Tinoco, R.; Elmen, L.; Segota, I.; Xian, Y.; Fujita, Y.; Sahu, A.; Zarecki, R.; Marie, K.; Feng, Y.; et al. Gut microbiota dependent anti-tumor immunity restricts melanoma growth in Rnf5(-/-) mice. Nat. Commun. 2019, 10, 1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Li, Y.; Elmen, L.; Segota, I.; Xian, Y.; Tinoco, R.; Feng, Y.; Fujita, Y.; Segura Munoz, R.R.; Schmaltz, R.; Bradley, L.M.; et al. Prebiotic-Induced Anti-tumor Immunity Attenuates Tumor Growth. Cell Rep. 2020, 30, 1753–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Alexander, J.L.; Wilson, I.D.; Teare, J.; Marchesi, J.R.; Nicholson, J.K.; Kinross, J.M. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 356–365. [Google Scholar] [CrossRef] [PubMed]
  96. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef] [PubMed]
  97. Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell 2018, 33, 570–580. [Google Scholar] [CrossRef]
  98. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [Green Version]
  99. Ting, S.Y.; Martinez-Garcia, E.; Huang, S.; Bertolli, S.K.; Kelly, K.A.; Cutler, K.J.; Su, E.D.; Zhi, H.; Tang, Q.; Radey, M.C.; et al. Targeted Depletion of Bacteria from Mixed Populations by Programmable Adhesion with Antagonistic Competitor Cells. Cell Host Microbe 2020. [Google Scholar] [CrossRef]
  100. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [Green Version]
  101. Kripke, M.L.; Fisher, M.S. Immunologic aspects of tumor induction by ultraviolet radiation. Natl. Cancer Inst. Monogr. 1978, 50, 179–183. [Google Scholar]
  102. Chapat, L.; Chemin, K.; Dubois, B.; Bourdet-Sicard, R.; Kaiserlian, D. Lactobacillus casei reduces CD8+ T cell-mediated skin inflammation. Eur. J. Immunol. 2004, 34, 2520–2528. [Google Scholar] [CrossRef] [PubMed]
  103. Hacini-Rachinel, F.; Gheit, H.; Le Luduec, J.B.; Dif, F.; Nancey, S.; Kaiserlian, D. Oral probiotic control skin inflammation by acting on both effector and regulatory T cells. PLoS ONE 2009, 4, e4903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Jatwani, S.; Hearth Holmes, M.P. Subacute Cutaneous Lupus Erythematosus; StatPearls: Treasure Island, FL, USA, 2020.
  105. Patra, V.; Wolf, P. Microbial elements as the initial triggers in the pathogenesis of polymorphic light eruption? Exp. Dermatol. 2016, 25, 999–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Katayama, S.; Panelius, J.; Koskenmies, S.; Skoog, T.; Mahonen, K.; Kisand, K.; Bondet, V.; Duffy, D.; Krjutskov, K.; Kere, J.; et al. Delineating the Healthy Human Skin UV Response and Early Induction of Interferon Pathway in Cutaneous Lupus Erythematosus. J. Investig. Dermatol. 2019, 139, 2058–2061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Khmaladze, I.; Butler, E.; Fabre, S.; Gillbro, J.M. Lactobacillus reuteri DSM 17938-A comparative study on the effect of probiotics and lysates on human skin. Exp. Dermatol. 2019, 28, 822–828. [Google Scholar] [CrossRef] [PubMed]
  108. Ra, J.; Lee, D.E.; Kim, S.H.; Jeong, J.W.; Ku, H.K.; Kim, T.Y.; Choi, I.D.; Jeung, W.; Sim, J.H.; Ahn, Y.T. Effect of oral administration of Lactobacillus plantarum HY7714 on epidermal hydration in ultraviolet B-irradiated hairless mice. J. Microbiol. Biotechnol. 2014, 24, 1736–1743. [Google Scholar] [CrossRef] [Green Version]
  109. Marini, A.; Jaenicke, T.; Grether-Beck, S.; Le Floc’h, C.; Cheniti, A.; Piccardi, N.; Krutmann, J. Prevention of polymorphic light eruption by oral administration of a nutritional supplement containing lycopene, beta-carotene, and Lactobacillus johnsonii: Results from a randomized, placebo-controlled, double-blinded study. Photodermatol. Photoimmunol. Photomed. 2014, 30, 189–194. [Google Scholar] [CrossRef]
  110. Tanew, A.; Radakovic, S.; Gonzalez, S.; Venturini, M.; Calzavara-Pinton, P. Oral administration of a hydrophilic extract of Polypodium leucotomos for the prevention of polymorphic light eruption. J. Am. Acad. Dermatol. 2012, 66, 58–62. [Google Scholar] [CrossRef]
  111. Caccialanza, M.; Percivalle, S.; Piccinno, R.; Brambilla, R. Photoprotective activity of oral polypodium leucotomos extract in 25 patients with idiopathic photodermatoses. Photodermatol. Photoimmunol. Photomed. 2007, 23, 46–47. [Google Scholar] [CrossRef]
  112. Hart, P.H.; Norval, M.; Byrne, S.N.; Rhodes, L.E. Exposure to Ultraviolet Radiation in the Modulation of Human Diseases. Annu. Rev. Pathol. 2019, 14, 55–81. [Google Scholar] [CrossRef]
  113. Clark, A.; Mach, N. Role of Vitamin D in the Hygiene Hypothesis: The Interplay between Vitamin D, Vitamin D Receptors, Gut Microbiota, and Immune Response. Front. Immunol. 2016, 7, 627. [Google Scholar] [CrossRef] [Green Version]
  114. Bashir, M.; Prietl, B.; Tauschmann, M.; Mautner, S.I.; Kump, P.K.; Treiber, G.; Wurm, P.; Gorkiewicz, G.; Hogenauer, C.; Pieber, T.R. Effects of high doses of vitamin D3 on mucosa-associated gut microbiome vary between regions of the human gastrointestinal tract. Eur. J. Nutr. 2016, 55, 1479–1489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Gruber-Wackernagel, A.; Bambach, I.; Legat, F.J.; Hofer, A.; Byrne, S.N.; Quehenberger, F.; Wolf, P. Randomized double-blinded placebo-controlled intra-individual trial on topical treatment with a 1,25-dihydroxyvitamin D(3) analogue in polymorphic light eruption. Br. J. Dermatol. 2011, 165, 152–163. [Google Scholar] [CrossRef] [PubMed]
  116. Salzer, S.; Kresse, S.; Hirai, Y.; Koglin, S.; Reinholz, M.; Ruzicka, T.; Schauber, J. Cathelicidin peptide LL-37 increases UVB-triggered inflammasome activation: Possible implications for rosacea. J. Dermatol. Sci. 2014, 76, 173–179. [Google Scholar] [CrossRef] [PubMed]
  117. Bandholtz, L.; Ekman, G.J.; Vilhelmsson, M.; Buentke, E.; Agerberth, B.; Scheynius, A.; Gudmundsson, G.H. Antimicrobial peptide LL-37 internalized by immature human dendritic cells alters their phenotype. Scand. J. Immunol. 2006, 63, 410–419. [Google Scholar] [CrossRef] [PubMed]
  118. Park, H.J.; Cho, D.H.; Kim, H.J.; Lee, J.Y.; Cho, B.K.; Bang, S.I.; Song, S.Y.; Yamasaki, K.; Di Nardo, A.; Gallo, R.L. Collagen synthesis is suppressed in dermal fibroblasts by the human antimicrobial peptide LL-37. J. Investig. Dermatol. 2009, 129, 843–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Wang, Y.; Kuo, S.; Shu, M.; Yu, J.; Huang, S.; Dai, A.; Two, A.; Gallo, R.L.; Huang, C.M. Staphylococcus epidermidis in the human skin microbiome mediates fermentation to inhibit the growth of Propionibacterium acnes: Implications of probiotics in acne vulgaris. Appl. Microbiol. Biotechnol. 2014, 98, 411–424. [Google Scholar] [CrossRef] [Green Version]
  120. Gueniche, A.; Cathelineau, A.C.; Bastien, P.; Esdaile, J.; Martin, R.; Queille Roussel, C.; Breton, L. Vitreoscilla filiformis biomass improves seborrheic dermatitis. J. Eur. Acad. Dermatol. Venereol. 2008, 22, 1014–1015. [Google Scholar] [CrossRef]
  121. Volz, T.; Skabytska, Y.; Guenova, E.; Chen, K.M.; Frick, J.S.; Kirschning, C.J.; Kaesler, S.; Rocken, M.; Biedermann, T. Nonpathogenic bacteria alleviating atopic dermatitis inflammation induce IL-10-producing dendritic cells and regulatory Tr1 cells. J. Investig. Dermatol. 2014, 134, 96–104. [Google Scholar] [CrossRef] [Green Version]
  122. Yu, Y.; Dunaway, S.; Champer, J.; Kim, J.; Alikhan, A. Changing our microbiome: Probiotics in dermatology. Br. J. Dermatol. 2020, 182, 39–46. [Google Scholar] [CrossRef]
  123. Harrison, O.J.; Linehan, J.L.; Shih, H.Y.; Bouladoux, N.; Han, S.J.; Smelkinson, M.; Sen, S.K.; Byrd, A.L.; Enamorado, M.; Yao, C.; et al. Commensal-specific T cell plasticity promotes rapid tissue adaptation to injury. Science 2019, 363, eaat6280. [Google Scholar] [CrossRef] [Green Version]
  124. Cheuk, S.; Wiken, M.; Blomqvist, L.; Nylen, S.; Talme, T.; Stahle, M.; Eidsmo, L. Epidermal Th22 and Tc17 cells form a localized disease memory in clinically healed psoriasis. J. Immunol. 2014, 192, 3111–3120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Gebhardt, T.; Wakim, L.M.; Eidsmo, L.; Reading, P.C.; Heath, W.R.; Carbone, F.R. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 2009, 10, 524–530. [Google Scholar] [CrossRef] [PubMed]
  126. Myles, I.A.; Williams, K.W.; Reckhow, J.D.; Jammeh, M.L.; Pincus, N.B.; Sastalla, I.; Saleem, D.; Stone, K.D.; Datta, S.K. Transplantation of human skin microbiota in models of atopic dermatitis. JCI Insight 2016, 1, e86955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Myles, I.A.; Earland, N.J.; Anderson, E.D.; Moore, I.N.; Kieh, M.D.; Williams, K.W.; Saleem, A.; Fontecilla, N.M.; Welch, P.A.; Darnell, D.A.; et al. First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis. JCI Insight 2018, 3, e120608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Borriello, S.P.; Hammes, W.P.; Holzapfel, W.; Marteau, P.; Schrezenmeir, J.; Vaara, M.; Valtonen, V. Safety of probiotics that contain lactobacilli or bifidobacteria. Clin. Infect. Dis. 2003, 36, 775–780. [Google Scholar] [CrossRef] [PubMed]
  129. Marteau, P.; Seksik, P. Tolerance of probiotics and prebiotics. J. Clin. Gastroenterol. 2004, 38, S67–S69. [Google Scholar] [CrossRef]
  130. Pfefferle, P.I.; Prescott, S.L.; Kopp, M. Microbial influence on tolerance and opportunities for intervention with prebiotics/probiotics and bacterial lysates. J. Allergy Clin. Immunol. 2013, 131, 1453–1463. [Google Scholar] [CrossRef]
  131. Hungin, A.P.S.; Mitchell, C.R.; Whorwell, P.; Mulligan, C.; Cole, O.; Agreus, L.; Fracasso, P.; Lionis, C.; Mendive, J.; Philippart de Foy, J.M.; et al. Systematic review: Probiotics in the management of lower gastrointestinal symptoms—an updated evidence-based international consensus. Aliment. Pharmacol. Ther. 2018, 47, 1054–1070. [Google Scholar] [CrossRef] [Green Version]
Figure 1. UV-induced effects on the skin and potential treatment strategies using microbes, pro- or pre-biotics. Topical formulations or application of recombinant microbes capable of inducing melanin and/or UV-absorbing compounds such as pityriacitrin could be used for pigmentation and antioxidation. Microbes or pro- or pre-biotics that can prevent and/or reduce UV-induced increase of transepidermal water loss (TEWL), hydrogen peroxide (H2O2) levels, oxidation of proteins and xanthine oxidase activity can have beneficial effects on photoaging. Novel strategies such as selective microbial enrichment using topical antibiotics, and/or application of anti-tumor, anti-inflammatory microbes or microbial metabolites/compounds could be beneficial to prevent UV-induced skin cancers and reduce UV-induced skin inflammation.
Figure 1. UV-induced effects on the skin and potential treatment strategies using microbes, pro- or pre-biotics. Topical formulations or application of recombinant microbes capable of inducing melanin and/or UV-absorbing compounds such as pityriacitrin could be used for pigmentation and antioxidation. Microbes or pro- or pre-biotics that can prevent and/or reduce UV-induced increase of transepidermal water loss (TEWL), hydrogen peroxide (H2O2) levels, oxidation of proteins and xanthine oxidase activity can have beneficial effects on photoaging. Novel strategies such as selective microbial enrichment using topical antibiotics, and/or application of anti-tumor, anti-inflammatory microbes or microbial metabolites/compounds could be beneficial to prevent UV-induced skin cancers and reduce UV-induced skin inflammation.
Nutrients 12 01795 g001

Share and Cite

MDPI and ACS Style

Patra, V.; Gallais Sérézal, I.; Wolf, P. Potential of Skin Microbiome, Pro- and/or Pre-Biotics to Affect Local Cutaneous Responses to UV Exposure. Nutrients 2020, 12, 1795. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12061795

AMA Style

Patra V, Gallais Sérézal I, Wolf P. Potential of Skin Microbiome, Pro- and/or Pre-Biotics to Affect Local Cutaneous Responses to UV Exposure. Nutrients. 2020; 12(6):1795. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12061795

Chicago/Turabian Style

Patra, VijayKumar, Irène Gallais Sérézal, and Peter Wolf. 2020. "Potential of Skin Microbiome, Pro- and/or Pre-Biotics to Affect Local Cutaneous Responses to UV Exposure" Nutrients 12, no. 6: 1795. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12061795

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