Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4048 2022-09-28 16:51:15 |
2 format -5 word(s) 4043 2022-09-29 03:27:46 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Olunoiki, E.B.;  Rehner, J.;  Bischoff, M.;  Koshel, E.;  Vogt, T.;  Reichrath, J.;  Becker, S. Characteristics of Skin Microbiome in Selected Dermatological Conditions. Encyclopedia. Available online: https://encyclopedia.pub/entry/27908 (accessed on 19 April 2024).
Olunoiki EB,  Rehner J,  Bischoff M,  Koshel E,  Vogt T,  Reichrath J, et al. Characteristics of Skin Microbiome in Selected Dermatological Conditions. Encyclopedia. Available at: https://encyclopedia.pub/entry/27908. Accessed April 19, 2024.
Olunoiki, Esther Boluwatife, Jacqueline Rehner, Markus Bischoff, Elena Koshel, Thomas Vogt, Jörg Reichrath, Sören Becker. "Characteristics of Skin Microbiome in Selected Dermatological Conditions" Encyclopedia, https://encyclopedia.pub/entry/27908 (accessed April 19, 2024).
Olunoiki, E.B.,  Rehner, J.,  Bischoff, M.,  Koshel, E.,  Vogt, T.,  Reichrath, J., & Becker, S. (2022, September 28). Characteristics of Skin Microbiome in Selected Dermatological Conditions. In Encyclopedia. https://encyclopedia.pub/entry/27908
Olunoiki, Esther Boluwatife, et al. "Characteristics of Skin Microbiome in Selected Dermatological Conditions." Encyclopedia. Web. 28 September, 2022.
Characteristics of Skin Microbiome in Selected Dermatological Conditions
Edit

The skin is the largest and outermost organ of the human body. The microbial diversity of the skin can be influenced by several variable factors such as physiological state, lifestyle, and geographical locations. There are increased interest in research aiming at an improved understanding of the relationship between the human microbiota and several diseases. Albeit understudied, interesting correlations between the skin microbiota and several dermatological conditions have been observed. Studies have shown that a decrease or increase in the abundance of certain microbial communities can be implicated in several dermatological pathologies.

dermatology human microbiome metagenomics microbial diversity

1. Introduction

The skin is the external and the largest organ of the human body that functions as an important physical barrier (e.g., to protect against harmful external conditions and invasion of pathogens) and communication tool (e.g., to respond to external stimuli) [1][2][3]. The human skin is home to millions of microorganisms, which are referred to as skin microbiota [1][2][3]. When compared to the more extensively studied gut microbiota, the skin microbiota seem to have similar characteristics, e.g., they also confer protection against pathogens, maintain the skin homeostasis, stimulate, and activate various immune responses, metabolize natural products, and produce antimicrobial peptides [3][4][5][6]. The physiological composition of the skin can be divided into four major micro-environments, i.e., (1) sebaceous (face, chest, and back), (2) moist (elbow, knees, genitalia), (3) dry (palms), and (4) foot-specific [3][5][7]. These micro-environments are characterized by specific microbiota that may have a considerable impact in maintaining a ‘healthy’ skin, whereas microbiota ‘shifts’ can facilitate the development and progression of diseases [3][5][8][9]. Members of the genus Cutibacterium (previously known as Propionibacterium), for example, dominate the sebaceous (oily) sites of the human skin, while the moist sites are primarily colonized by members of the genera Staphylococcus and Corynebacterium, and the female genital tract is characterized by a predominance of Lactobacillus [3][8][10][11]. Although not as diverse as the bacterial community, viral and fungal commensals are also present on skin; for example, fungi of the genus Malassezia are widely distributed across the sebaceous sites of the skin [3][8][12][13].
The composition of the skin microbiota is assumed to be relatively stable. However, an imbalance in the proportion of ‘normal’ skin flora to pathogenic microorganisms, a phenomenon known as dysbiosis, results in the initiation and progression of several dermatological diseases [1][3][8]. Until recently, microbiological culture-based methods were primarily used for the isolation and characterization of skin microbial communities [1][3][8][14]. However, these methods underestimate the ‘true’ diversity of organ-specific microbiota because many bacterial species differ considerably with regard to nutritional requirements and growth rates on agar plates, and a number of species simply cannot be grown in culture [3][8][14][15][16][17][18]. Staphylococcus spp., for example, grows faster than Cutibacterium spp. and Corynebacterium spp. on agar plates inoculated by skin swabs, and the latter may thus be overlooked in culture-based studies. In an attempt to circumvent these biases and capture the total diversity of the skin microbiome, sequencing approaches are nowadays used to identify members of the skin microbial communities [1][3][15][18]. A commonly employed approach makes use of the targeted amplification of ‘molecular fingerprints’ of specific pathogen classes, e.g., the housekeeping 16S ribosomal RNA gene for bacterial communities [3][8][17] and the internal transcribed spacer 1 (ITS1) region of the eukaryotic ribosomal gene for fungi [3][8][15][16], which is then followed by nucleic acid sequencing of the amplified product for species identification. However, the constant evolution of sequencing technologies from conventional Sanger sequencing to more high-throughput techniques such as pyrosequencing and Illumina sequencing allows for an improved, more precise microbiota characterization, which is facilitated by increased read depths and shorter read lengths [3][8][19][20]. Amplicon sequencing, i.e., the sequencing of previously amplified nucleic acids from specific microbiota, can capture genetic variability in microbial communities efficiently [1][3][21][22][23]. Such amplicons can then be analyzed using different software tools such as mothur [3][24] and ‘Quantitative Insights Into Microbial Ecology’ (Qiime) [25]. However, amplicon sequencing is prone to different shortcomings and may cause an analytical bias, which might be overcome by the application of the more recent shotgun metagenomic sequencing [1][2][3][8]. Through shotgun metagenomics sequencing, all the genetic materials present in a given sample are analyzed simultaneously by untargeted (and unbiased) sequencing of nucleic acids without prior polymerase chain reaction (PCR) assays to amplify specific sequences. Many short sequences are being generated, which are then reconstructed by computational biology into consensus sequences [8][26]. Shotgun metagenomics provides sufficient information to differentiate microbial genetic materials into species and further into strains [8][21][26]. This ability for strain differentiation might be important to elucidate the functional differences within a species [8][21][26][27][28].

2. The Role of Microbiota in Healthy Skin

Structurally, the skin is composed of three distinct layers: the epidermis, the dermis, and the subcutaneous fat tissue [29][30]. The epidermis, which is the outer layer of the skin, is differentiated into the stratum corneum, stratum lucidum, stratum granulosum, and stratum basale [3][30]. The stratum corneum, which is the outermost layer, is composed of terminally differentiated and enucleated keratinocytes that are chemically cross-linked and act as a barrier [28][29][30]. The keratinocytes present in the stratum granulosum are cysteine-histidine rich and bind keratin [28][31]. Basal keratinocytes, immune cells (Langerhans cells, T cells, and melanocytes) can be found in the stratum basale [28][31]. The dermis is located beneath the epidermis. It is differentiated into the papillary sub-layer that facilitates the transport of nutrients [3][28][32] and the reticular sub-layer in which the hair follicles, sebaceous glands, and sweat glands are present. The dermis is also home to fibroblasts, myofibroblasts, and immune cells (macrophages, lymphocytes, and mast cells) [28][31][32][33]. The subcutaneous fat layer is composed of fibrocytes and adipocytes. This layer produces a wide variety of growth factors, adipokines, cytokines, and immune cells. The adipose tissue also stores energy and functions as an endocrine gland, which is crucial for glucose homeostasis and lipid metabolism [34][35][36][37]. The skin plays a vital role in stimulating and training the human immune system [38][39][40]. Studies have shown that the microbiome is influenced by many factors, such as environment, lifestyle, diet, and medication. Changes within these factors could lead to a decrease in the diversity of the human microbiota, thereby negatively impacting the host immune system and microbe communication, which may lead to an altered immunological tolerance and miseducation of the immune system. The sebum produced by the sebaceous gland lubricates the hair and skin. Sapienic acid produced by the hydrolysis of sebum by commensal microbes acts alongside other antimicrobial peptides such as cathelicidin, beta-defensins, and antimicrobial histones to control microbial colonization [1][38][39][40]. Eccrine sweat secreted directly onto the skin surface creates unfavorable conditions for the survival and proliferation of microbes [3][38][41]. The highly colonized dermal appendage is home to a variety of microbes. This environment allows for ample interaction between microorganisms and host cells [38]. Dendritic cells are found in the epidermis and act as antigen-presenting cells to T lymphocytes in the nearby lymphoid organs. T cells become activated in the presence of invading pathogens, and trigger an inflammatory response. However, a dysregulation in this mechanism can occur, which may result in the emergence of inflammatory skin diseases [38].
Extensive microbe-microbe interaction is expected between organisms that share a similar niche as they compete for nutrients and other growth factors [38]. For example, Staphylococcus aureus, which is a commensal skin colonizer in roughly 30 percent of the population, has been studied extensively with regard to its interaction with other microbes. S. aureus is an opportunistic pathogen that can cause severe infection, particularly in immunocompromised hosts. Other Staphylococcus spp. that inhabit the skin of humans, for example Staphylococcus hominis and Staphylococcus lugdunensis, may produce antimicrobial peptides that specifically inhibit colonization by S. aureus. Other species are also known for their potentially beneficial effect on the human skin, such as Lactobacillus spp., which prevent pathogen colonization and produce anti-inflammatory secondary metabolites [39][40][41]. The microbial diversity of the skin microbiota is influenced by several factors such as lifestyle, age, medications, etc. [3][8][38][42][43]. The immune system often recognizes the host’s commensal microbiota and establishes a mutualistic interaction. Some bacteria, such as Staphylococcus epidermidis, can also interact with the host’s keratinocytes directly, thereby inducing the production of antimicrobial peptides via immune cell signaling [40]. However, dysbiosis may occur and can trigger the initiation of chronic inflammatory skin disorders [38][42][43][44]. The incidence of specific pathologies such as atopic dermatitis and psoriasis have risen considerably in high-income countries, and it is speculated that dietary changes, lifestyle modifications, hygiene, and environmental factors may have contributed to this, as they may adversely affect the microbial diversity and metabolism of the skin, which might in turn lower the organ’s immune tolerance [42][43]. Disease-specific examples will be provided in the following sections for three selected conditions, i.e., (1) acne inversa, (2) (muco-)cutaneous candidiasis, and (3) psoriasis. Psoriasis and acne inversa are not directly associated with skin infections by pathogenic microorganisms. However, psoriasis as one of the most prevalent skin conditions is being studied intensively in regard to microbiome analysis. Researchers aim to find microbial species which affect this disease, both positively, and negatively. The role between the skin microbiome and acne inversa, on the other hand, has not been established yet. In contrast to psoriasis, research on acne inversa is still in the early stages. Researchers would like to point out the current state of research on these two diseases. In contrast to non-infectious skin diseases, researchers also aim to elucidate the impact of the skin microbiome during the infectious disease candidiasis.

3. The Role of Microbiota in Selected Dermatological Diseases

3.1. Hidradenitis Suppurativa (Acne Inversa)

Hidradenitis suppurativa (HS), also known as acne inversa, is a chronic inflammatory skin disorder [45][46][47]. It is clinically characterized by deep-seated fistulae, nodules, abscesses, sinus tracts, and scars in the axillae, inguinal folds, perianal and perineal regions, buttocks, and infra/inter-mammary folds. HS is a painful and discomforting skin condition, which has an adverse impact on the psycho-social health of affected patients. Indeed, research has elucidated that people suffering from HS develop depression more frequently and are twice more likely to be unemployed [48][49]. The morbidity associated with HS is significantly higher than observed in other inflammatory skin diseases [49][50]. HS severity can be measured by different staging systems, e.g., the Hurley classification, Sartorius scoring, and HS severity index [45]. With regard to epidemiology, HS has a relatively high prevalence in young females of African descent [45][51]. The etiology of HS has been linked to several contributing factors such as (i) cutaneous microbiome dysbiosis, (ii) genetics, (iii) lifestyle specificities and obesity, (iv) hormonal dysbalance, and (v) immune system modifications. Here, researchers focus mainly on the influence of the skin microbiota on disease severity.
A variety of studies demonstrated that skin microbiota play a key role in the etiopathogenesis of HS [52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67]. Using a bacteriological culture-based approach, Brook et al. described a polymicrobial nature of HS lesions. S. aureus, Streptococcus pyogenes, and Pseudomonas aeruginosa were the most prevalent aerobic bacteria, and Peptostreptococcus spp., Prevotella spp., microaerophilic streptococci, Fusobacterium spp., and Bacteroides spp. were the most common anaerobes [45][46][47][48][49][50][51][52][53][54][55][56][57]. Lapin et al. confirmed such a bacterial diversity in HS lesions and described S. aureus and coagulase-negative staphylococci as the most abundant aerobic bacteria species, while Peptostreptococcus spp. and Cutibacterium acnes were the most common anaerobes [68]. However, S. aureus was not frequently isolated from acute lesions, which raises the question of how the microbiota composition might shift during acute flares of HS and in different disease activity stages. A study carried out by Guet-Revillet et al. employed a combination of microbiological culture and metagenomic techniques to address this issue. According to the bacterial culture method, two microbiological profiles were apparent (A and B). In profile A (Hurley stage 1), S. lugdunensis was predominant, while in profile B (Hurley stages 2 and 3), a variety of anaerobic bacteria species were isolated. Metagenomic sequencing after PCR amplification of 16S rRNA yielded similar results, with staphylococci predominating in profile A and different anaerobic species (mainly Prevotella spp., Porphyromonas spp., Anaerococcus spp., and Mobiluncus spp.) being more characteristic of more advanced disease stages of the disease [53]. More recent studies carried out by Naik et al. showed a decrease in the abundance of Cutibacterium spp. in a study cohort which suffered from HS and was instructed to avoid any type of detergents, cosmetics, oral and topical antibiotics, and bathing before skin swabs were taken [68]. This result was consistent with other studies, which reported a decrease of Cutibacterium spp. and an increase of Prevotella spp. and Porphyromonas spp. as being potentially linked to disease progression and severity [69].

3.2. (Muco-)Cutaneous Candidiasis

Candidiasis or thrush is a fungal infection caused by members of the genus Candida [70]. Only a few members of the genus are pathogenic to humans. Out of the over 150 species of Candida, Candida albicans is considered the most pathogenic. Yet, other species such as C. glabrata, C. tropicalis, C. krusei, C. kefyr, C. guilliermondii, C. dubliniensis, and, more recently, C. auris, may also cause human disease, and some of these pathogens are considered as potential public health concerns because of their relatively high propensity to cause outbreaks and to become resistant to commonly used anti-fungal agents [71][72][73][74][75][76][77][78][79][80][81][82][83][84][85][86]. C. albicans is most commonly found in the vagina of about 30 percent of women [87]. Other non-C. albicans species are also found in the reproductive tract of about 10–30 percent of women [87][88][89][90][91][92][93]. Candida species are opportunistic pathogens that cause infections ranging from superficial oral thrush and vaginitis to systemic, potentially fatal infections (candidemia) [70][71][94]. Mucocutaneous candidiasis can be classified into non-genital (oropharyngeal and invasive infections) and genitourinary diseases [76]. Here, researchers will focus on the common genitourinary diseases caused by Candida species, which share many characteristics with cutaneous candidiasis. Candidiasis occurs in men, women, and children alike. The most prevalent genitourinary diseases are vulvovaginal candidiasis in women, balanitis, balanoposthitis in men, and candiduria in both sexes and children [76]. Vulvovaginal candidiasis (VVC) is a common infection of the lower female reproductive tract [76][77]. VVC is characterized by itching, burning and painful urination, redness, curd-like and foul-smelling vaginal discharge, and painful sexual intercourse [76][77]. About 75 percent of women develop VVC at least once during their lifetime [76][77][78]. VVC can be recurrent and it is medically termed recurrent vulvovaginal candidiasis (RVVC), if it occurs more than four times per year [76][77]. Approximately 8 percent of women globally suffer from RVVC [76][77]. Several factors influence the occurrence of VVC and RVVC such as the use of antibiotics, sexual activity and HIV infection, high estrogen-containing oral contraceptives, pregnancy, the use of sodium cotransporter 2 (SGLT2) inhibitors, and uncontrolled diabetes mellitus [76][80]. Compared to oropharyngeal candidiasis, VVC occurs frequently also in immunocompetent and otherwise healthy women [76][77][78][79][80][81][82]. RVVC requires the long-term use of antifungal drugs to prevent it from relapsing [76][77][83]. One study carried out by Ceccarani et al. in 2019 showed a significant reduction of Lactobacillus spp. in infected vaginal ecosystems of 18 females suffering from VVC compared to 21 healthy individuals [84]. These findings suggest a protective role of Lactobacillus spp., such as L. crispatus, to prevent overgrowth of Candida spp. However, some studies also suggest an abundance of Lactobacillus spp. with a greater incidence of Candida colonization [87][88][95]. Eastment et al. conducted a study in 2021 to analyze the microbiota composition of VVC patients in Kenya and the United States. Microbiota composition was acquired using amplicon sequencing. The study demonstrated that a higher relative abundance of Megasphaera species and Mageeibacillus indolicus was associated with a lower risk of yeast detection in the vaginal area. Moreover, higher abundances of Bifidobacterium bifidum, several Streptococcus spp., and Aerococcus christensenii were correlated with yeast detection, suggesting a possible interaction of those species, thereby increasing the colonization of yeast species in the vaginal area [85]. Other studies suggest the co-occurrence of Candida with other bacterial vaginosis (BV)-associated bacteria [95][96][97][98][99].
An important limitation of these studies is that Lactobacillus spp. were not identified at the species level [84][87][88][95]. Different Lactobacillus spp. can either be beneficial for the host and protect from an infection with Candida spp., or increase the risk of such a colonization. Therefore, identification to the species level is crucial when regarding the impact of Lactobacillus spp. on candidiasis. For example, studies hypothesized that the presence of L. iners in the female reproductive tract creates a suitable environment for the co-occurrence of C. albicans, especially when compared to L. crispatus. Tortelli et al. proved this hypothesis by examining the vaginal microbiome of women. They observed that women whose reproductive tract were colonized by L. iners had an increased incidence of the co-occurrence of Candida spp. in comparison to women with diverse vaginal microbiomes and those colonized by L. crispatus. It was speculated that L. iners produces more lactic acid, which increases the pH of the vagina and significantly supports the colonization and co-occurrence of Candida spp. [93]. However, a main limitation of this was that the entire vaginal microbiome may not have been captured because of the use of amplicon sequencing only to detect Lactobacillus species. [93].
There is a high possibility that other anaerobic bacteria present in the female reproductive tract may play crucial roles in the etiopathogenesis of VVC and RVVC. The researchers also did not consider the various stages of the menstrual cycles of the women sampled. It would be interesting to understand the role of the menstrual cycle in the initiation and progression of VVC and if the vaginal microbiota composition changes during the different stages of the menstrual cycle. A study using modern shotgun genome sequencing would shed more light on these interesting research questions. With regard to Candida balanitis, a lot is still unknown about the influence of the host-microbiome on disease occurrence. Studies carried out by Meng et al. showed that sexual behavior (the use of condoms), circumcision, lifestyle, and personal hygiene play an important role in disease incidence. They observed that men who use condoms are more likely to have Staphylococcus spp. as the prevalent bacteria implicated in disease severity, while men without sexual activities are more likely to harbour Prevotella spp. [100]. However, considerably less studies have been carried out on Candida balanitis than vaginitis.

3.3. Psoriasis

Psoriasis (PS) is a chronic inflammatory skin disease thought to have a genetic and immunological link [101]. About 2 percent of the world’s population is affected by PS. Its prevalence varies according to geographical locations [101][102]. Studies have also shown that Caucasians and Scandinavians are more likely to be affected by PS than Africans and Asians [102][103][104][105]. The etiology of PS remains vividly debated; however, some studies have shown a correlation between the skin microbiota dysbiosis and the occurrence of PS [101][106]. Several studies have established a relationship between the colonization of the psoriatic skin lesions and S. aureus. The researchers observed that approximately 60 percent of people affected by psoriasis have their skin and nares colonized by S. aureus, as compared to 3 to 30 percent in healthy people, thereby pointing out the possibility that S. aureus may exacerbate PS flares [106][107][108]. Viruses, such as the human papillomaviruses (HPV) have also been implicated to play a role in the occurrence of PS [106]. Fry and Baker proposed that HPV infection of the keratinocyte is supported by epidermal proliferation, and that epidermal hyperproliferation, which is a characteristic of PS, results in an opportunistic infection [106]. Arguably, HPV might be one assumed autoantigen recognized by the CD4+ and CD8+ T cells in psoriatic lesions [106][109]. Malassezia spp. have also been hypothesized as contributors in the development of this disease. Malassezia is a common inhabitant of the scalp [106]. It was observed that orally administered antifungal drugs resulted in a significant decrease in the abundance of the yeast on the scalp and an improvement of the disease condition [110][111]. To further examine this finding, Lober et al. carried out patch testing of sonicated, heat-killed Malassezia spp. cells on patients with inactive PS. It was observed that skin lesions clinically and histologically similar to PS were formed [110]. To further demonstrate the relationship between the skin microbiota and PS flares, Alekseyenko et al. performed a study between 2008 and 2011, which included 75 patients with psoriasis and 124 healthy controls. Skin swabs were obtained from all participants. Diseased individuals were swabbed at the affected and unaffected body sites. DNA was extracted and the bacterial composition analyzed by amplicon sequencing. The researchers found a significant increase in the abundance of Corynebacterium, Propionibacterium, Staphylococcus, and Streptococcus in psoriatic plaques [55]. In contrast to the previous results, Gao et al. revealed that members of the phylum Firmicutes were overrepresented in PS lesions, while the members of the phylum Actinobacteria and Cutibacterium spp. were the least abundant [57]. Likewise, Quan et al. analyzed the microbiota of healthy individuals and affected and unaffected skin of PS patients. They observed an abundance of Corynebacterium sp. and a decrease in the abundance of Cutibacterium in PS lesions [112]. Drago et al. identified a dysbiosis of the microbiome of one Italian PS patient in 2016. They observed that a significant increase in the abundance of Proteobacteria and a decrease in the abundance of Streptococcaceae, Rhodobacteraceae, Campylobacteraceae, Moraxellaceae, and Firmicutes was associated with PS plagues [113]. Contrary to the result of the experiment published by Lober et al., Paulino revealed that the occurrence and distribution of Malassezia spp. were not host-specific and its population on the skin remained stable over time. Additionally, there was no significant difference in the abundance of Malassezia in both diseased and healthy skin areas of PS patients. However, only two individuals, one male and one female located in North America were included in the study [114]. In order to estimate a significance of these findings, more participants should be included in the study. Different climate zones should also be considered, as the environment (e.g., temperature range, humidity, and air pollution) plays a major role in the skin microbial composition [115].
Another study from Italy employed shotgun metagenomic sequencing to analyze the skin microbiome of PS patients. Skin lesions were compared to non-lesional skin areas. The findings suggested an increase in Staphylococcus spp. in lesioned skin. On average, the observed alpha diversity, a measure to analyze the biodiversity of microbiomes, was significantly lower in diseased compared to unaffected areas [115]. Similar results were shown concerning the gut microbiome of PS patients. Indeed, an interesting study by Todberg and colleagues analyzed very recently whether PS also has an impact on the gut microbiome. They performed shotgun metagenomic sequencing on fecal samples of 53 untreated patients with plaque PS and 52 healthy controls. Overall, participants suffering from plaque PS showed a significantly reduced alpha diversity of their gut microbiomes compared to healthy participants. They further confirmed the higher abundance of Blautia spp. in diseased participants, whereas Faecalibacterium spp. displayed a lower abundance [116]. The limitations of the aforementioned studies include small sample sets, sometimes a lack of precise details on the actual study design, and the more commonly used amplicon sequencing instead of untargeted sequencing methods.

3.4. Correlation of the Skin Microbiome and Other Dermatological Conditions

Apart from the three selected skin conditions, i.e., (i) hidradenitis suppurativa (acne inversa), (ii) (muco-)cutaneous candidiasis, and (iii) psoriasis, there are many other skin conditions, for which the role of the microbiome has been described regarding disease initiation and progression. Atopic dermatitis (AD), for example, has been extensively studied. AD is a chronic inflammatory disease, affecting 1–3% of adults and 5–20% of children worldwide [117][118][119]. Especially an increase of S. aureus has been correlated with lesional skin areas of AD patients and a more severe form of the disease [120]. In contrast, S. epidermidis has been associated with milder cases of AD [8][121][122]. Overall, a lower alpha diversity has also been shown for AD patients in contrast to healthy skin [118][123][124]. Specific bacterial genera that decreased in lesional skin of AD patients are Streptococcus, Cutibacterium, and Corynebacterium [8][125]. A shift to a more diverse microbiome has previously been achieved after UV-B exposure [126]. However, probiotic baths and lotions, as well as a specific inhibition of S. aureus might also become adjunctive therapy options.
Skin cancer, such as malignant melanoma and basal cell carcinoma, has also been shown to be affected by the cutaneous microbiome [127][128][129]. One striking study carried out by Mekadim et al. in 2022 detected an increase in Fusobacterium, Trueperella, Staphylococcus, Streptococcus, and Bacteroides in melanoma tissue, compared to healthy skin. The researchers were also able to show a two-fold decrease in alpha diversity of melanoma tissue in contrast to healthy skin [126].
Next to skin cancer, certain autoimmune diseases also play a crucial role in dermatology, e.g., systemic sclerosis and bullous pemphigoid. The skin microbiome has been described for both dermatological conditions [130][131]. Johnson et al. have analyzed the skin microbiome of systemic sclerosis patients in 2019. They described an increase in gram-negative taxa, such as Burkholderia, Citrobacter, and Vibrio, whereas lipophilic taxa seem to be increased [131]. More research is needed to evaluate existing microbiome data and gain new insights into the complex interplay of microbes and the host.

References

  1. Sanford, J.A.; Gallo, R.L. Functions of the skin microbiota in health and disease. Semin. Immunol. 2013, 25, 370–377.
  2. Roth, R.R.; James, W.D. Microbial ecology of the skin. Annu. Rev. Microbiol. 1988, 42, 441–464.
  3. Byrd, A.; Belkaid, Y.; Segre, J. The human skin microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155.
  4. Scharschmidt, T.C.; Fischbach, M.A. What lives on our skin: Ecology, genomics and therapeutic opportunities of the skin microbiome. Drug Discov. Today Dis. Mech. 2013, 10, e83–e89.
  5. Grice, E.A. The intersection of microbiome and host at the skin interface: Genomic- and metagenomic-based insights. Genome Res. 2015, 25, 1514–1520.
  6. Belkaid, Y.; Segre, J.A. Dialogue between skin microbiota and immunity. Science 2014, 346, 954–959.
  7. Zhou, H.; Shi, L.; Ren, Y.; Tan, X.; Liu, W.; Liu, Z. Applications of Human Skin Microbiota in the Cutaneous Disorders for Ecology-Based Therapy. Front. Cell. Infect. Microbiol. 2010, 10, 609.
  8. Kong, H.; Oh, J.; Deming, C.; Conlan, S.; Grice, E.A.; Beatson, M.A.; Nomicos, E.; Polley, E.C.; Komarow, H.D.; NISC Comparative Sequence Program; et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012, 20, 850–859.
  9. Paulino, L.; Tseng, C.H.; Strober, B.E.; Blaser, M.J. Molecular analysis of fungal microbiota in samples from healthy human skin and psoriatic lesions. J. Clin. Microbiol. 2006, 44, 2933–2941.
  10. Scholz, C.F.P.; Kilian, M. The natural history of cutaneous propionibacteria, and reclassification of selected species within the genus Propionibacterium to the proposed novel genera Acidipropionibacterium gen. nov Cutibacterium gen. nov. and Pseudopropionibacterium gen. nov. Int. J. Syst. Evol. Microbiol. 2016, 66, 4422–4432.
  11. Grice, E.; Kong, H.H.; Conlan, S.; Deming, C.B.; Davis, J.; Young, A.C.; NISC Comparative Sequencing Program; Bouffard, G.G.; Blakesley, R.W.; Murray, P.R.; et al. Topographical and temporal diversity of the human skin microbiome. Science 2009, 324, 1190–1192.
  12. Oh, J.; Byrd, A.L.; Deming, C.; Conlan, S.; NISC Comparative Sequencing Program; Kong, H.H.; Segre, J.A. Biogeography and individuality shape function in the human skin metagenome. Nature 2014, 514, 59–64.
  13. Findley, K.; Oh, J.; Yang, J.; Conlan, S.; Deming, C.; Meyer, J.A.; Schoenfeld, D.; Nomicos, E.; Park, M.; NIH Intramural Sequencing Center Comparative Sequencing Program; et al. Topographic diversity of fungal and bacterial communities in human skin. Nature 2015, 498, 367–370.
  14. Kong, H.H.; Segre, J.A. Skin microbiome: Looking back to move forward. J. Investig. Dermatol. 2012, 132, 933–939.
  15. Woese, C.; Fox, G.E. Phylogenetic structure of the prokaryotic domain: The primary kingdoms. Proc. Natl Acad. Sci. USA 1977, 74, 5088–5090.
  16. Schoch, C.L.; Seifert, K.A.; Huhndorf, S.; Robert, V.; Spouge, J.L.; Levesque, C.A.; Chen, W.; Fungal Barcoding Consortium, & Fungal Barcoding Consortium Author List. Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for fungi. Proc. Natl. Acad. Sci. USA 2012, 109, 6241–6246.
  17. Jo, J.; Kennedy, E.A.; Kong, H.H. Research techniques made simple: Bacterial 16S ribosomal RNA gene sequencing in cutaneous research. J. Investig. Dermatol. 2016, 136, e23–e27.
  18. Goodman, A.L.; Kallstrom, G.; Faith, J.J.; Reyes, A.; Moore, A.; Dantas, G.; Gordon, J.I. Extensive personal human gut microbiota culture collections characterized and manipulated in gnotobiotic mice. Proc. Natl. Acad. Sci. USA 2011, 108, 6252–6257.
  19. Weyrich, L.; Dixit, S.; Farrer, A.G.; Cooper, A.J.; Cooper, A.J. The skin microbiome: Associations between altered microbial communities and disease. Australas. J. Dermatol. 2015, 54, 268–274.
  20. Grogan, M.; Bartow-McKenney, C.; Flowers, L.; Knight, S.A.B.; Uberoi, A.; Grice, E.A. Research techniques made simple: Profiling the skin microbiota. J. Investig. Dermatol. 2019, 139, 747.e741–752.e741.
  21. Meisel, J.; Hannigan, G.D.; Tyldsley, A.S.; SanMiguel, A.J.; Hodkinson, B.P.; Zheng, Q.; Grice, E.A. Skin microbiome surveys are strongly influenced by experimental design. J. Investig. Dermatol. 2016, 136, 947–956.
  22. Zeeuwen, P.; Boekhorst, J.; Ederveen, T.; Kleerebezem, M.; Schalkwijk, J.; van Hijum, S.; Timmerman, H.M. Reply to Meisel et al. J. Investig. Dermatol. 2017, 137, 961–962.
  23. Castelino, M.; Eyre, S.; Moat, J.; Fox, G.; Martin, P.; Ho, P.; Upton, M.; Barton, A. Optimisation of methods for bacterial skin microbiome investigation: Primer selection and comparison of the 454 versus MiSeq platform. Nat. Rev. Microbiol. 2017, 17, 23.
  24. Schloss, P.; Westcott, S.L.; Ryabin, T.; Hall, J.R.; Hartmann, M.; Hollister, E.B.; Lesniewski, R.A.; Oakley, B.B.; Parks, D.H.; Robinson, C.J.; et al. Introducing mothur: Open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 2009, 75, 7537–7541.
  25. Caporaso, J.G.; Kuczynski, J.; Stombaugh, J.; Bittinger, K.; Bushman, F.D.; Costello, E.K.; Fierer, N.; Peña, A.G.; Goodrich, J.K.; Gordon, J.I.; et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Method. 2010, 7, 335–336.
  26. Yuan, S.; Cohen, D.B.; Ravel, J.; Abdo, Z.; Forney, L.J. Evaluation of methods for the extraction and purification of DNA from the human microbiome. PLoS ONE 2012, 7, e33865.
  27. Tomida, S.; Nguyen, L.; Chiu, B.H.; Liu, J.; Sodergren, E.; Weinstock, G.M.; Li, H. Pan-genome and comparative genome analyses of propionibacterium acnes reveal its genomic diversity in the healthy and diseased human skin microbiome. mBio 2013, 4, e000030-13.
  28. Costello, E.; 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.
  29. Nguyen, A.; Athena, M.S. The Dynamics of the Skin’s Immune System. Int. J. Mol. Sci. 2019, 20, 1811.
  30. Hsu, Y.; Li, L.; Fuchs, E. Emerging interactions between skin stem cells and their niches. Nat. Med. 2014, 20, 847–856.
  31. Ovaere, P.; Lippens, S.; Vandenabeele, P.; Declercq, W. The emerging roles of serine protease cascades in the epidermis. Trends Biochem. Sci. 2009, 34, 453–463.
  32. Shirshin, E.A.; Gurfinkel, Y.I.; Priezzhev, A.V.; Fadeev, V.V.; Lademann, J.; Darvin, M.E. Two-photon autofluorescence lifetime imaging of human skin papillary dermis in vivo: Assessment of blood capillaries and structural proteins localization. Sci. Rep. 2017, 7, 1171.
  33. Woodley, D. Distinct Fibroblasts in the Papillary and Reticular Dermis: Implications for Wound Healing. Dermatol. Clin. 2017, 35, 95–100.
  34. Driskell, R.R.; Jahoda, C.A.; Chuong, C.M.; Watt, F.M.; Horsley, V. Defining dermal adipose tissue. Exp. Dermatol. 2014, 23, 629–631.
  35. Rodrigues, M.; Kosaric, N.; Bonham, C.A.; Gurtner, G.C. Wound Healing: A Cellular Perspective. Physiol. Rev. 2019, 99, 665–706.
  36. Gurtner, G.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound Healing: A Cellular Perspective. Nature 2008, 453, 314–321.
  37. Cildir, G.; Akincilar, S.C.; Tergaonkar, V. Chronic adipose tissue inflammation: All immune cells on the stage. Trends Mol. Med. 2013, 19, 487–500.
  38. Lunjani, N.; Ahearn-Ford, S.; Dube, F.S.; Hlela, C.; O’Mahony, L. Mechanisms of microbe-immune system dialogue within the skin. Genes Immun. 2021, 22, 276–288.
  39. Lebeer, S.; Oerlemans, E.; Claes, I.; Wuyts, S.; Henkens, T.; Spacova, I.; van den Broek, M.; Tuyaerts, I.; Wittouck, S.; De Boeck, I.; et al. Topical cream with live lactobacilli modulates the skin microbiome and reduces acne symptoms. BioRxiv 2018.
  40. 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.
  41. Zipperer, A.; Konnerth, M.C.; Laux, C.; Berscheid, A.; Janek, D.; Weidenmaier, C.; Burian, M.; Schilling, N.A.; Slavetinsky, C.; Marschal, M.; et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature 2016, 535, 511–516.
  42. Alkotob, S.S.; Cannedy, C.; Harter, K.; Movassagh, H.; Paudel, B.; Prunicki, M.; Sampath, V.; Schikowski, T.; Smith, E.; Zhao, Q.; et al. Advances and novel developments in environmental influences on the development of atopic diseases. Allergy 2020, 75, 3077–3086.
  43. Krämer, U.; Sugiri, D.; Ranft, U.; Krutmann, J.; von Berg, A.; Berdel, D.; Behrendt, H.; Kuhlbusch, T.; Hochadel, M.; Wichmann, H.E.; et al. Eczema, respiratory allergies, and traffic-related air pollution in birth cohorts from small-town areas. J. Dermatol. Sci. 2009, 56, 99–105.
  44. Wang, E.; Qiang, X.; Li, J.; Zhu, S.; Wang, P. The in vitro immune-modulating properties of a sweat gland-derived antimicrobial peptide dermcidin. Shock 2016, 45, 28–32.
  45. Langan, E.; Recke, A.; Bokor-Billmann, T.; Billmann, F.; Kahle, B.K.; Zillikens, D. The Role of the Cutaneous Microbiome in Hidradenitis Suppurativa-Light at the End of the Microbiological Tunnel. Shock 2020, 21, 1205.
  46. Goldburg, S.; Strober, B.E.; Payette, M.J. Hidradenitis suppurativa: Epidemiology, clinical presentation, and pathogenesis. J. Am. Acad. Dermatol. 2020, 82, 1045–1058.
  47. Vekic, D.; Frew, J.; Cains, G.D. Hidradenitis suppurativa, a review of pathogenesis, associations and management. Australas. J. Dermatol. 2018, 59, 267–277.
  48. Marvel, J.; Vlahiotis, A.; Sainski-Nguyen, A.; Willson, T.; Kimball, A. Disease burden and cost of hidradenitis suppurativa: A retrospective examination of US administrative claims data. BMJ Open 2019, 9, 030579.
  49. Kolli, S.; Senthilnathan, A.; Cardwell, L.A.; Richardson, I.M.; Feldman, S.R.; Pichardo, R.O. Hidradenitis suppurativa has an enormous impact on patients’ lives. J. Am. Acad. Dermatol. 2020, 82, 236–238.
  50. Keary, E.A.; Hevey, D.; Tobin, A.M. qualitative analysis of psychological distress in hidradenitis suppurativa. Br. J. Dermatol. 2020, 182, 342–347.
  51. Garg, A.; Kirby, J.S.; Lavian, J.; Lin, G.; StrunK, A. Sex- and Age-Adjusted Population Analysis of Prevalence Estimates for Hidradenitis Suppurativa in the United States. JAMA Dermatol. 2017, 153, 760–764.
  52. Tulin, E. Hidradenitis suppurativa and metabolic syndrome. J. Clin. Dermatol. 2018, 36, 41–47.
  53. Guet-Revillet, H.; Coignard-Biehler, H.; Jais, J.P.; Quesne, G.; Frapy, E.; Poirée, S.; Le Guern, A.S.; Le Flèche-Matéos, A.; Hovnanian, A.; Consigny, P.H.; et al. Bacterial pathogens associated with hidradenitis suppurativa, France. Emerg. Infect. Dis. 2014, 20, 1990–1998.
  54. Highet, A.; Warren, R.E.; Weekes, A.J. Bacteriology and antibiotic treatment of perineal suppurative hidradenitis. Arch. Dermatol. 1988, 124, 1047–1051.
  55. Lapins, J.; Jarstrand, C.; Emtestam, L. Coagulase-negative staphylococci are the most common bacteria found in cultures from the deep portions of hidradenitis suppurativa lesions, as obtained by carbon dioxide laser surgery. Br. J. Dermatol. 1999, 140, 90–95.
  56. Sartorius, K.; Killasli, H.; Oprica, C.; Sullivan, A.; Lapins, J. Bacteriology of hidradenitis suppurativa exacerbations and deep tissue cultures obtained during carbondioxide laser treatment. Br. J. Dermatol. 2012, 166, 879–883.
  57. Brook, I.; Frazier, E.H. Aerobic and anaerobic microbiology of axillary hidradenitis suppurativa. J. Med. Microbiol. 1999, 48, 103–110.
  58. Alekseyenko, A.; Perez-Perez, G.I.; De Souza, A.; Strober, B.; Gao, Z.; Bihan, M.; Li, K.; Methé, B.A.; Blaser, M.J. Community differentiation of the cutaneous microbiota in psoriasis. Microbiome 2013, 1, 31.
  59. Fry, L.; Baker, B.S.; Powles, A.V.; Fahlen, A.; Engstrand, L. Is chronic plaque psoriasis triggered by microbiota in the skin? Br. J. Dermatol. 2013, 169, 47–52.
  60. Gao, Z.; Tseng, C.H.; Strober, B.E.; Pei, Z.; Blaser, M.J. Substantial alterations of the cutaneous bacterial biota in psoriatic lesions. PLoS ONE 2008, 3, e2719.
  61. Tobin, A.S. Unravelling the microbiome in psoriasis. Br. J. Dermatol. 2019, 181, 1124–1125.
  62. Ring, H.C.; Thorsen, J.; Saunte, D.M.; Lilje, B.; Bay, L.; Riis, P.T.; Larsen, N.; Andersen, L.O.; Nielsen, H.V.; Miller, I.M.; et al. The Follicular Skin Microbiome in Patients with Hidradenitis Suppurativa and Healthy Controls. JAMA Dermatol. 2017, 153, 894–905.
  63. Paller, A.; Kong, H.H.; Seed, P.; Naik, S.; Scharschmidt, T.C.; Gallo, R.L.; Luger, T.; Irvine, A.D. The microbiome in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2019, 143, 26–35.
  64. Fyhrquist, N.; Muirhead, G.; Prast-Nielsen, S.; Jeanmougin, M.; Olah, P.; Skoog, T.; Jules-Clement, G.; Feld, M.; Barrientos-Somarribas, M.; Sinkko, H.; et al. Microbe-host interplay in atopic dermatitis and psoriasis. Nat. Commun. 2019, 10, 4703.
  65. Miodovnik, M.A.; Kunstner, A.; Langan, E.A.; Zillikens, D.; Glaser, R.; Sprecher, E.; Baines, J.F.; Schmidt, E.; Ibrahim, S.M. Distinct cutaneous microbiota profile in autoimmune bullous disease patients. Exp. Dermatol. 2017, 26, 1221–1227.
  66. Scaglione, G.; Fania, L.; De Paolis, E.; De Bonis, M.; Mazzanti, C.; Di Zenzo, G.; Lechiancole, S.; Messinese, S.; Capoluongo, E. Evaluation of cutaneous, oral and intestinal microbiota in patients affected by pemphigus and bullous pem-phigoid: A pilot study. Exp. Mol. Pathol. 2019, 112, 104331.
  67. Ramasamy, S.; Barnard, E.; Dawson, T.L., Jr.; Li, H. The role of the skin microbiota in acne pathophysiology. Br. J. Dermatol. 2019, 181, 691–699.
  68. Naik, H.; Jo, J.H.; Paul, M.; Kong, H.H. Skin microbiota perturbations are distinct and disease severity-dependent in hidradenitis suppurativa. J. Investig. Dermatol. 2019, 140, 922–925.
  69. Ring, H.; Sigsgaard, V.; Thorsen, J.; Fuursted, K.; Fabricius, S.; Saunte, D.M.; Jemec, G.B. The microbiome of tunnels in hidradenitis suppurativa patients. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1775–1780.
  70. Hani, U.; Shivakumar, H.G.; Vaghela, R.; Osmani, R.A.; Shrivastava, A. Candidiasis: A fungal infection–current challenges and progress in prevention and treatment. Infectious disorders drug targets. Infect. Disord. Drug Targets 2015, 15, 42–52.
  71. Rogers, T. Antifungal chemotherapy in patients with acquired immunodeficiency syndrome. Lancet 1992, 340, 648–651.
  72. Bongomin, F.; Gago, S.; Oladele, R.O.; Denning, D.W. Global and Multi-National Prevalence of Fungal Diseases-Estimate Precision. J. Fungi 2017, 3, 57.
  73. Arendrup, M. Candida and candidemia. Susceptibility and epidemiology. Dan. Med. J. 2012, 60, B4698.
  74. Wisplinghoff, H.; Bischoff, T.; Tallent, S.M.; Seifert, H.; Wenzel, R.P.; Edmond, M.B. Nosocomial bloodstream infections in US hospitals: Analysis of 24,179 cases from a prospective nation-wide surveillance study: An official publication of the Infectious Diseases Society of America. Clin. Infect. Dis. 2004, 39, 309–317.
  75. Kullberg, B.; Arendrup, M.C. Invasive Candidiasis. N. Engl. J. Med. 2015, 373, 1445–1456.
  76. Achkar, J.; Fries, B.C. Candida infection of the genitourinary tract. J. Fungi 2020, 23, 2.
  77. Willems, H.; Ahmed, S.S.; Liu, J.; Xu, Z.; Peters, B.M. Vulvovaginal Candidiasis: A Current Understanding and Burning Questions. J. Fungi 2020, 6, 27.
  78. Sobel, J. Vaginitis. N. Engl. J. Med. 1997, 337, 1896–1903.
  79. Denning, W.; Kneale, M.; Sobel, J.D.; Rautemaa-Richardson, R. Global burden of recurrent vulvovaginal candidiasis—A systematic review. Lancet 2018, 18, 339–347.
  80. Nyirjesy, P.; Zhao, Y.; Ways, K.; Usiskin, K. Evaluation of vulvovaginal symptoms and Candida colonization in women with type 2 diabetes mellitus treated with canagliflozin, a sodium glucose co-transporter 2 inhibitor. Curr. Med. Res. Opin. 2012, 28, 1173–1178.
  81. Sobel, J.; Wiesenfeld, H.C.; Martens, M.; Danna, P.; Hooton, T.M.; Rompalo, A.; Sperling, M.; Livengood, C., III; Horowitz, B.; Von Thron, J.; et al. Global and Multi-National Prevalence of Fungal Diseases-Estimate Precision. N. Engl. J. Med. 2004, 351, 876–883.
  82. Edwards, S. Balanitis and balanoposthitis: A review. Genitourin. Med. 1996, 72, 155–159.
  83. Sobel, J.; Kauffman, C.A.; McKinsey, D.; Zervos, M.; Vazquez, J.A.; Karchmer, A.W.; Lee, J.; Thomas, C.; Panzer, H.; Dismukes, W.E. Candiduria: A randomized, double-blind study of treatment with fluconazole and placebo. The National Institute of Allergy and Infectious Diseases (NIAID) Mycoses Study Group. Clin. Infect. Dis. 2000, 30, 19–24.
  84. Ceccarani, C.; Foschi, C.; Parolin, C.; D’Antuono, A.; Gaspari, V.; Consolandi, C.; Laghi, L.; Camboni, T.; Vitali, B.; Severgnini, M.; et al. Diversity of vaginal microbiome and metabolome during genital infections. Sci. Rep. 2019, 9, 14095.
  85. Eastment, M.C.; Balkus, J.E.; Richardson, B.A.; Srinivasan, S.; Kimani, J.; Anzala, O.; Schwebke, J.; Fiedler, T.L.; Fredricks, D.N.; McClelland, R.S. Association Between Vaginal Bacterial Microbiota and Vaginal Yeast Colonization. J. Infect. Dis. 2021, 223, 914–923.
  86. Bouza, E.A.; San Juan, R.; Muñoz, P.; Voss, A.; Kluytmans, J.; Co-operative Group of the European Study Group on Nosocomial Infections. European perspective on nosocomial urinary tract infections II. Report on incidence, clinical characteristics and outcome (ESGNI-004 study). European Study Group on Nosocomial Infection. Clin. Microbiol. Infect. 2001, 7, 532–542.
  87. Beigi, R.; Meyn, L.A.; Moore, D.M.; Krohn, M.A.; Hillier, S.L. Vaginal yeast colonization in nonpregnant women: A longitudinal study. Obstet. Gynecol. 2004, 104, 926–930.
  88. Cotch, M.F.; Hillier, S.L.; Gibbs, R.S.; Eschenbach, D.A. Epidemiology and outcomes associated with moderate to heavy Candida colonization during pregnancy. Vaginal Infections and Prematurity Study Group. Am. J. Obstet. Gynecol. 1998, 178, 374–380.
  89. Brandolt, T.; Klafke, G.B.; Gonçalves, C.V.; Bitencourt, L.R.; Martinez, A.M.; Mendes, J.F.; Meireles, M.C.; Xavier, M.O. Prevalence of Candida spp. in cervical-vaginal samples and the in vitro susceptibility of isolates. Braz. J. Microbiol. 2017, 48, 145–150.
  90. Payne, M.; Ireland, D.J.; Watts, R.; Nathan, E.A.; Furfaro, L.L.; Kemp, M.W.; Keelan, J.A.; Newnham, J.P. Ureaplasma parvum genotype, combined vaginal colonisation with Candida albicans, and spontaneous preterm birth in an Australian cohort of pregnant women. BMC Pregnancy Childbirth 2016, 16, 312.
  91. Payne, M.; Cullinane, M.; Garland, S.M.; Tabrizi, S.N.; Donath, S.M.; Bennett, C.M.; Amir, L.H. Detection of Candida spp. in the vagina of a cohort of nulliparous pregnant women by culture and molecular methods: Is there an association between maternal vaginal and infant oral colonisation. Aust. N. Z. J. Obstet. Gynaecol. 2016, 56, 179–184.
  92. Fidel, P.L.; Barousse, M.; Espinosa, T.; Ficarra, M.; Sturtevant, J.; Martin, D.H.; Quayle, A.J.; Dunlap, K. An intravaginal live Candida challenge in humans leads to new hypotheses for the immunopathogenesis of vulvovaginal candidiasis. Infect. Immun. 2004, 72, 2939–2946.
  93. Tortelli, B.; Lewis, W.G.; Allsworth, J.E.; Member-Meneh, N.; Foster, L.R.; Reno, H.E.; Peipert, J.F.; Fay, J.C.; Lewis, A.L. Associations between the vaginal microbiome and Candida colonization in women of reproductive age. Am. J. Obstet. Gynecol. 2020, 222, 471.e1–471.e9.
  94. Fidel, P. Distinct protective host defenses against oral and vaginal candidiasis. Med. Mycol. 2002, 40, 359–375.
  95. McClelland, R.; Richardson, B.A.; Hassan, W.M.; Graham, S.M.; Kiarie, J.; Baeten, J.M.; Mandaliya, K.; Jaoko, W.; Ndinya-Achola, J.O.; Holmes, K.K. Prospective study of vaginal bacterial flora and other risk factors for vulvovaginal candidiasis. J. Infect. Dis. 2009, 199, 1883–1890.
  96. González-Fernández, D.; Koski, K.G.; Sinisterra, O.T.; Del Carmen Pons, E.; Murillo, E.; Scott, M.E. Interactions among urogenital, intestinal, skin, and oral infections in pregnant and lactating Panamanian Ngäbe women: A neglected public health challenge. Am. J. Trop. Med. 2015, 92, 1100–1110.
  97. Liu, M.; Xu, S.R.; He, Y.; Deng, G.H.; Sheng, H.F.; Huang, X.M.; Ouyang, C.Y.; Zhou, H.W. Diverse vaginal microbiomes in reproductive-age women with vulvovaginal candidiasis. PLoS ONE 2013, 8, e79812.
  98. Vahidnia, A.; Tuin, H.; Bliekendaal, H.; Spaargaren, J. Association of sexually transmitted infections, Candida species, gram-positive flora and perineal flora with bacterial vaginosis. New Microbiol. 2015, 38, 559–563.
  99. Rathod, S.; Klausner, J.D.; Krupp, K.; Reingold, A.L.; Madhivanan, P. Epidemiologic features of Vulvovaginal Candidiasis among reproductive-age women in India. Infectious diseases in obstetrics and gynecology. Infect. Dis. Obstet. Gynecol. 2012, 2012, 859071.
  100. Meng, L.; Jian-Xun, M.; Hang-Hang, J.; Chun-Ming, H.; Xing-Hua, G.; Li, Z. Microbiome profile in patients with adult balanoposthitis: Relationship with redundant prepuce, genital mucosa physical barrier status and inflammation. Acta. Derm. Venerol. 2021, 101, adv00466.
  101. Christophers, E. Psoriasis—Epidemiology and clinical spectrum. Infect. Immun. 2004, 26, 314–320.
  102. Rendon, A.; Schäkel, K. Psoriasis Pathogenesis and Treatment. Int. J. Mol. Sci. 2019, 20, 1475.
  103. Parisi, R.; Symmons, D.P.; Griffiths, C.E.; Ashcroft, D.M. Identification and Management of Psoriasis and Associated Comorbidity (IMPACT) project team. Global epidemiology of psoriasis: A systematic review of incidence and prevalence. J. Investig. Dermatol. 2013, 133, 377–385.
  104. Gibbs, S. Skin disease and socioeconomic conditions in rural Africa: Tanzania. Int. J. Dermatol. 1996, 35, 633–639.
  105. Rachakonda, T.; Schupp, C.W.; Armstrong, A.W. Psoriasis prevalence among adults in the United States. J. Am. Acad. Dermatol. 2014, 70, 512–516.
  106. Fry, L.; Baker, B.S. Triggering psoriasis: The role of infections and medications. J. Clin. Dermatol. 2007, 25, 606–615.
  107. Leung, D.Y.; Harbeck, R.; Bina, P.; Reiser, R.F.; Yang, E.; Norris, D.A.; Hanifin, J.M.; Sampson, H.A. Presence of IgE antibodies to staphylococcal exotoxins on the skin of patients with atopic dermatitis. Evidence for a new group of allergens. J. Clin. Investig. 1993, 92, 1374–1380.
  108. Tomi, N.; Kränke, B.; Aberer, E. Staphylococcal toxins in patients with psoriasis, atopic dermatitis, and erythroderma, and in healthy control subjects. J. Am. Acad. Dermatol. 2005, 53, 67–72.
  109. Steele, J.C.; Roberts, S.; Rookes, S.M.; Gallimore, P.H. Detection of CD4(+)- and CD8(+)-T-cell responses to human papillomavirus type 1 antigens expressed at various stages of the virus life cycle by using an enzyme-linked immunospot assay of gamma interferon release. J. Virol. 2002, 76, 6027–6036.
  110. Farr, P.M.; Krause, L.B.; Marks, J.M.; Shuster, S. Response of scalp psoriasis to oral ketoconazole. Lancet 1985, 2, 921–922.
  111. Rosenberg, E.; Belew, P.W. Improvement of psoriasis of the scalp with ketoconazole. Arch. Dermatol. 1982, 118, 370–371.
  112. Quan, C.; Chen, X.Y.; Li, X.; Xue, F.; Chen, L.H.; Liu, N.; Wang, B.; Wang, L.Q.; Wang, X.P.; Yang, H.; et al. Psoriatic lesions are characterized by higher bacterial load and imbalance between Cutibacterium and Corynebacterium. J. Am. Acad. Dermatol. 2020, 82, 955–961.
  113. Drago, L.; De Grandi, R.; Altomare, G.; Pigatto, P.; Rossi, O.; Toscano, M. Skin microbiota of first cousins affected by psoriasis and atopic dermatitis. Clin. Mol. Allergy 2016, 14, 2.
  114. Paulino, L.C.; Tseng, C.H.; Blaser, M.J. Analysis of Malassezia microbiota in healthy superficial human skin and in psoriatic lesions by multiplex real-time PCR. FEMS Yeast Res. 2008, 8, 460–471.
  115. Tett, A.; Pasolli, E.; Farina, S.; Truong, D.T.; Asnicar, F.; Zolfo, M.; Beghini, F.; Armanini, F.; Jousson, O.; De Sanctis, V.; et al. Unexplored diversity and strain-level structure of the skin microbiome associated with psoriasis. Nature 2017, 3, 14.
  116. Todberg, T.; Egeberg, A.; Zachariae, C.; Sørensen, N.; Pedersen, O.; Skov, L. Patients with psoriasis have a dysbiotic taxonomic and functional gut microbiota. Br. J. Dermatol. 2022, 187, 89–98.
  117. Ferček, I.; Lugović-Mihić, L.; Tambić-Andrašević, A.; Ćesić, D.; Grginić, A.G.; Bešlić, I.; Mravak-Stipetić, M.; Mihatov-Štefanović, I.; Buntić, A.M.; Čivljak, R. Features of the Skin Microbiota in Common Inflammatory Skin Diseases. Life 2021, 11, 962.
  118. Nutten, S. Atopic dermatitis: Global epidemiology and risk factors. Ann. Nutr. Metab. 2015, 66 (Suppl. S1), 8–16.
  119. Ahluwalia, J.; Borok, J.; Haddock, E.S.; Ms, R.S.A.; Schwartz, E.W.; Hosseini, D.; Amini, S.; Eichenfield, L.F. The microbiome in preadolescent acne: Assessment and prospective analysis of the influence of benzoyl peroxide. Pediatr. Dermatol. 2019, 36, 200–206.
  120. Auber, M.; Balica, S.; Hsu, C.Y.; Jean-Decoster, C.; Lauze, C.; Redoules, D.; Viodé, C.; Schmitt, A.M.; Serre, G.; Simon, M. Staphylococcus aureus density on lesional and nonlesional skin is strongly associated with disease severity in atopic dermatitis. J. Allergy Clin. Immunol. 2016, 137, 1272–1274.
  121. Callewaert, C.; Helffer, K.R.; Lebaron, P. Skin Microbiome and its Interplay with the Environment. Am. J. Clin. Dermatol. 2020, 21, 4–11.
  122. Seite, S.; Flores, G.E.; Henley, J.B.; Martin, R.; Zelenkova, H.; Aguilar, L.; Fierer, N. Microbiome of affected and unaffected skin of patients with atopic dermatitis before and after emollient treatment. J. Drugs Dermatol. 2014, 13, 1365–1372.
  123. Gonzalez, M.E.; Schaffer, J.V.; Orlow, S.J.; Gao, Z.; Li, H.; Alekseyenko, A.V.; Blaser, M.J. Cutaneous microbiome effects of fluticasone propionate cream and adjunctive bleach baths in childhood atopic dermatitis. J. Am. Acad. Dermatol. 2016, 75, 481–493.
  124. Baurecht, H.; Rühlemann, M.; Rodríguez, E.; Thielking, F.; Harder, I.; Erkens, A.-S.; Stölzl, D.; Ellinghaus, E.; Hotze, M.; Lieb, W.; et al. Epidermal lipid composition, barrier integrity, and eczematous inflammation are associated with skin microbiome configuration. J. Allergy Clin. Immunol. 2018, 141, 1668–1676.
  125. Shi, B.; Bangayan, N.J.; Curd, E.; Taylor, P.A.; Gallo, R.L.; Leung, D.Y.; Li, H. The skin microbiome is different in pediatric versus adult atopic dermatitis. J. Allergy Clin. Immunol. 2016, 138, 1233–1236.
  126. Lossius, A.H.; Sundnes, O.; Ingham, A.C.; Edslev, S.M.; Bjørnholt, J.V.; Lilje, B.; Bradley, M.; Asad, S.; Haraldsen, G.; Skytt-Andersen, P.; et al. Shifts in the Skin Microbiota after UVB Treatment in Adult Atopic Dermatitis. Dermatology 2022, 238, 109–120.
  127. Mekadim, C.; Skalnikova, H.K.; Cizkova, J.; Cizkova, V.; Palanova, A.; Horak, V.; Mrazek, J. Dysbiosis of skin microbiome and gut microbiome in melanoma progression. BMC Microbiol. 2022, 22, 63.
  128. Mrázek, J.; Mekadim, C.; Kučerová, P.; Švejstil, R.; Salmonová, H.; Vlasáková, J.; Tarasová, R.; Čížková, J.; Červinková, M. Melanoma-related changes in skin microbiome. Folia Microbiol. 2019, 64, 435–442.
  129. Yu, Y.; Champer, J.; Beynet, D.; Kim, J.; Friedman, A.J. The role of the cutaneous microbiome in skin cancer: Lessons learned from the gut. J. Drugs Dermatol. 2015, 14, 461–465.
  130. Belheouane, M.; Hermes, B.M.; Van Beek, N.; Benoit, S.; Bernard, P.; Drenovska, K.; Gerdes, S.; Gläser, R.; Goebeler, M.; Günther, C.; et al. Characterization of the skin microbiota in bullous pemphigoid patients and controls reveals novel microbial indicators of disease. J. Adv. Res. 2022.
  131. Johnson, M.E.; Franks, J.M.; Cai, G.; Mehta, B.K.; Wood, T.A.; Archambault, K.; Pioli, P.A.; Simms, R.W.; Orzechowski, N.; Arron, S.; et al. Microbiome dysbiosis is associated with disease duration and increased inflammatory gene expression in systemic sclerosis skin. Arthritis Res. Ther. 2019, 21, 49.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 223
Revisions: 2 times (View History)
Update Date: 29 Sep 2022
1000/1000