Advances in Microbiome-Derived Solutions and Methodologies Are Founding a New Era in Skin Health and Care
Abstract
:1. Introduction
2. The Skin and Its Microbiome
3. Skin/Scalp Conditions and Microbiome
3.1. Variability across Age
3.2. Variability Due to Extrinsic Factors
3.3. Variability Associated with Skin Health and Disorders
3.3.1. Role of S. epidermidis
3.3.2. Role of C. acnes
3.3.3. Role of Other Microorganisms
4. Methods in Skin Microbiome Exploration
4.1. Perspectives for Skin Microbiome Metabolomics Analysis
4.2. Expectations from Metagenomic and Data Science
4.3. Meta-Omics and System Biology Approach
4.4. Network-Based Models’ Approach
5. Microbiome-Based Cosmetic Solutions and Technologies
“Ferments, lysates, extracts, filtrates or any combination of these ingredients that are not living but which have been obtained by means of probiotic bacteria (Bacillus, Bifidobacterium, Lactobacillus, Lactococcus, Vitreoscilla, Streptococcus thermophilus, Leuconostoc) or fungi used primarily as fermentation facilitators (Saccharomyces, Candida bombicola, Kloeckera, Hansenula-Pichia, Aspergillus)”:
“Non-viable microorganisms (inactivated/heat-killed), mostly lactic-acid forming bacteria: Enterococcus faecalis, Lactobacillus (paracasei, casei, acidophilus), Lactococcus, or Vitreoscilla filiform”.
“Metabolic products/by-products (isolated) including bacteriocin extract, ectoin, succinic acid, lactic acid, hydrolyzed yogurt protein, sodium hyaluronate, and milk proteins” [204].
5.1. Postbiotics
5.1.1. Probiotic Fractions
5.1.2. Effector Molecules/Metabolites
5.1.3. Modulation of the Skin Microbiome
5.1.4. Cross Talk with the Immune Response
6. Future Implications/Outlook
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Blaser, M.J.; Cardon, Z.G.; Cho, M.K.; Dangl, J.L.; Donohue, T.J.; Green, J.L.; Knight, R.; Maxon, M.E.; Northen, T.R.; Pollard, K.S.; et al. Toward a Predictive Understanding of Earth’s Microbiomes to Address 21st Century Challenges. mBio 2016, 7, e00714-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brody, H. The gut microbiome. Nature 2020, 577, S5. [Google Scholar] [CrossRef]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef] [PubMed]
- Grice, E.A. The skin microbiome: Potential for novel diagnostic and therapeutic approaches to cutaneous disease. Semin. Cutan. Med. Surg. 2014, 33, 98–103. [Google Scholar] [CrossRef] [Green Version]
- Gallo, R.L. Human Skin Is the Largest Epithelial Surface for Interaction with Microbes. J. Investig. Dermatol. 2017, 137, 1213–1214. [Google Scholar] [CrossRef] [Green Version]
- Grice, E.A.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oh, J.; Byrd, A.L.; Park, M.; Kong, H.H.; Segre, J.A.; NISC Comparative Sequencing Program. Temporal Stability of the Human Skin Microbiome. Cell 2016, 165, 854–866. [Google Scholar] [CrossRef] [Green Version]
- Grice, E.A.; Segre, J.A. The skin microbiome. Nat. Rev. Microbiol. 2011, 9, 244–253. [Google Scholar] [CrossRef]
- Byrd, A.L.; Belkaid, Y.; Segre, J.A. The human skin microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef]
- Findley, K.; Oh, J.; Yang, J.; Conlan, S.; Deming, C.; Meyer, J.A.; Schoenfeld, D.; Nomicos, E.; Park, M.; Kong, H.H.; et al. Topographic diversity of fungal and bacterial communities in human skin. Nature 2013, 498, 367–370. [Google Scholar] [CrossRef]
- Cogen, A.L.; Nizet, V.; Gallo, R.L. Skin microbiota: A source of disease or defence? Br. J. Dermatol. 2008, 158, 442–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.E.; Tsao, H. The skin microbiome: Current perspectives and future challenges. J. Am. Acad. Dermatol. 2013, 69, 143–155.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ohnemus, U.; Kohrmeyer, K.; Houdek, P.; Rohde, H.; Wladykowski, E.; Vidal, S.; Horstkotte, M.A.; Aepfelbacher, M.; Kirschner, N.; Behne, M.J.; et al. Regulation of Epidermal Tight-Junctions (TJ) during Infection with Exfoliative Toxin-Negative Staphylococcus Strains. J. Investig. Dermatol. 2008, 128, 906–916. [Google Scholar] [CrossRef] [Green Version]
- Kuo, I.-H.; Carpenter-Mendini, A.; Yoshida, T.; McGirt, L.Y.; Ivanov, A.I.; Barnes, K.C.; Gallo, R.L.; Borkowski, A.W.; Yamasaki, K.; Leung, D.Y.; et al. Activation of Epidermal Toll-Like Receptor 2 Enhances Tight Junction Function: Implications for Atopic Dermatitis and Skin Barrier Repair. J. Investig. Dermatol. 2013, 133, 988–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Linehan, J.L.; Harrison, O.R.; Han, S.-J.; Byrd, A.L.; Vujkovic-Cvijin, I.; Villarino, A.V.; Sen, S.K.; Shaik, J.; Smelkinson, M.; Tamoutounour, S.; et al. Non-classical Immunity Controls Microbiota Impact on Skin Immunity and Tissue Repair. Cell 2018, 172, 784–796.e18. [Google Scholar] [CrossRef] [Green Version]
- Cogen, A.L.; Yamasaki, K.; Muto, J.; Sanchez, K.M.; Crotty Alexander, L.; Tanios, J.; Lai, Y.; Kim, J.E.; Nizet, V.; Gallo, R.L. Staphylococcus epidermidis antimicrobial delta-toxin (phenol-soluble modulin-gamma) cooperates with host antimicrobial peptides to kill group A Streptococcus. PLoS ONE 2010, 5, e8557. [Google Scholar] [CrossRef]
- Lai, Y.; Cogen, A.L.; Radek, K.A.; Park, H.J.; Macleod, D.T.; Leichtle, A.; Ryan, A.F.; di Nardo, A.; Gallo, R.L. Activation of TLR2 by a small molecule produced by Staphylococcus epidermidis increases antimicrobial defense against bacterial skin in-fections. J. Investig. Dermatol. 2010, 130, 2211–2221. [Google Scholar]
- Lai, Y.; Di Nardo, A.; Nakatsuji, T.; Leichtle, A.; Yang, Y.; Cogen, A.L.; Wu, Z.-R.; Hooper, L.V.; Schmidt, R.R.; Von Aulock, S.; et al. Commensal bacteria regulate Toll-like receptor 3–dependent inflammation after skin injury. Nat. Med. 2009, 15, 1377–1382. [Google Scholar] [CrossRef]
- 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]
- Pastar, I.; O’Neill, K.; Padula, L.; Head, C.R.; Burgess, J.L.; Chen, V.; Garcia, D.; Stojadinovic, O.; Hower, S.; Plano, G.V.; et al. Staphylococcus epidermidis Boosts Innate Immune Response by Activation of Gamma Delta T Cells and Induction of Perforin-2 in Human Skin. Front. Immunol. 2020, 11, 2253. [Google Scholar] [CrossRef]
- Scharschmidt, T.C.; Vasquez, K.S.; Truong, H.-A.; Gearty, S.V.; Pauli, M.L.; Nosbaum, A.; Gratz, I.K.; Otto, M.; Moon, J.J.; Liese, J.; et al. A Wave of Regulatory T Cells into Neonatal Skin Mediates Tolerance to Commensal Microbes. Immunity 2015, 43, 1011–1021. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Christensen, G.J.M.; Brüggemann, H. Bacterial skin commensals and their role as host guardians. Benef. Microbes 2014, 5, 201–215. [Google Scholar] [CrossRef] [PubMed]
- Claesen, J.; Spagnolo, J.B.; Ramos, S.F.; Kurita, K.L.; Byrd, A.L.; Aksenov, A.A.; Melnik, A.V.; Wong, W.R.; Wang, S.; Hernandez, R.D.; et al. A Cutibacterium acnes antibiotic modulates human skin microbiota composition in hair follicles. Sci. Transl. Med. 2020, 12. [Google Scholar] [CrossRef] [PubMed]
- Kannan, R.; Ng, M.J.M. Cutaneous lesions and vitamin B12 deficiency. Can. Fam. Physician 2008, 54, 529–532. [Google Scholar] [PubMed]
- Kang, D.; Shi, B.; Erfe, M.C.; Craft, N.; Li, H. Vitamin B12 modulates the transcriptome of the skin microbiota in acne path-ogenesis. Sci. Transl. Med. 2015, 7, 293ra103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mahabadi, N.; Bhusal, A.; Banks, S.W. Riboflavin Deficiency; StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
- Williams, J.D.; Jacobson, E.L.; Kim, H.; Kim, M.; Jacobson, M.K. Folate in Skin Cancer Prevention. Subcell. Biochem. 2011, 56, 181–197. [Google Scholar] [CrossRef] [Green Version]
- Hasoun, L.Z.; Bailey, S.W.; Outlaw, K.K.; Ayling, J.E. Effect of serum folate status on total folate and 5-methyltetrahydrofolate in human skin. Am. J. Clin. Nutr. 2013, 98, 42–48. [Google Scholar] [CrossRef] [Green Version]
- Rembe, J.D.; Fromm-Dornieden, C.; Stuermer, E.K. Effects of Vitamin B Complex and Vitamin C on Human Skin Cells: Is the Perceived Effect Measurable? Adv. Skin Wound Care 2018, 31, 225–233. [Google Scholar] [CrossRef]
- Ridaura, V.K.; Bouladoux, N.; Claesen, J.; Chen, Y.E.; Byrd, A.L.; Constantinides, M.G.; Merrill, E.D.; Tamoutounour, S.; Fischbach, M.A.; Belkaid, Y. Contextual control of skin immunity and inflammation by Corynebacterium. J. Exp. Med. 2018, 215, 785–799. [Google Scholar] [CrossRef] [Green Version]
- Sowada, J.; Schmalenberger, A.; Ebner, I.; Luch, A.; Tralau, T. Degradation of benzo[a]pyrene by bacterial isolates from human skin. FEMS Microbiol. Ecol. 2014, 88, 129–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patra, V.; Byrne, S.N.; Wolf, P. The Skin Microbiome: Is It Affected by UV-induced Immune Suppression? Front. Microbiol. 2016, 7, 1235. [Google Scholar] [CrossRef] [Green Version]
- Flores, G.E.; Caporaso, J.G.; Henley, J.B.; Rideout, J.R.; Domogala, D.; Chase, J.; Leff, J.W.; Vázquez-Baeza, Y.; Gonzalez, A.; Knight, R.; et al. Temporal variability is a personalized feature of the human microbiome. Genome Biol. 2014, 15, 531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skowron, K.; Bauza-Kaszewska, J.; Kraszewska, Z.; Wiktorczyk-Kapischke, N.; Grudlewska-Buda, K.; Kwiecińska-Piróg, J.; Wałecka-Zacharska, E.; Radtke, L.; Gospodarek-Komkowska, E. Human Skin Microbiome: Impact of Intrinsic and Extrinsic Factors on Skin Microbiota. Microorganisms 2021, 9, 543. [Google Scholar] [CrossRef]
- Oh, J.; Byrd, A.L.; Deming, C.; Conlan, S.; Kong, H.H.; Segre, J.A.; NISC Comparative Sequencing Program. Biogeography and individuality shape function in the human skin metagenome. Nature 2014, 514, 59–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sowada, J.; Lemoine, L.; Schön, K.; Hutzler, C.; Luch, A.; Tralau, T. Toxification of polycyclic aromatic hydrocarbons by commensal bacteria from human skin. Arch. Toxicol. 2017, 91, 2331–2341. [Google Scholar] [CrossRef] [Green Version]
- Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [Green Version]
- Zhu, T.; Liu, X.; Kong, F.-Q.; Duan, Y.-Y.; Yee, A.L.; Kim, M.; Galzote, C.; Gilbert, J.A.; Quan, Z.-X. Age and Mothers: Potent Influences of Children’s Skin Microbiota. J. Investig. Dermatol. 2019, 139, 2497–2505.e6. [Google Scholar] [CrossRef]
- Tollanes, M.C.; Moster, D.; Daltveit, A.K.; Irgens, L.M. Cesarean section and risk of severe childhood asthma: A popula-tion-based cohort study. J. Pediatr. 2008, 153, 112–116. [Google Scholar] [CrossRef]
- Renz-Polster, H.; David, M.R.; Buist, A.S.; Vollmer, W.M.; O’Connor, E.A.; Frazier, E.A.; Wall, M.A. Caesarean section delivery and the risk of allergic disorders in childhood. Clin. Exp. Allergy 2005, 35, 1466–1472. [Google Scholar] [CrossRef]
- Kennedy, E.A.; Connolly, J.; Hourihane, J.O.; Fallon, P.G.; McLean, W.H.I.; Murray, D.; Jo, J.-H.; Segre, J.A.; Kong, H.H.; Irvine, A.D. Skin microbiome before development of atopic dermatitis: Early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year. J. Allergy Clin. Immunol. 2016, 139, 166–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loo, E.X.L.; Sim, J.Z.T.; Loy, S.L.; Goh, A.; Chan, Y.H.; Tan, K.H.; Yap, F.; Gluckman, P.D.; Godfrey, K.M.; van Bever, H.; et al. Associations between caesarean delivery and allergic outcomes: Results from the GUSTO study. Ann. Allergy Asthma Immunol. 2017, 118, 636–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chu, D.M.; Ma, J.; Prince, A.L.; Antony, K.M.; Seferovic, M.D.; Aagaard, K.M. Maturation of the infant microbiome com-munity structure and function across multiple body sites and in relation to mode of delivery. Nat. Med. 2017, 23, 314–326. [Google Scholar] [CrossRef] [Green Version]
- Capone, K.A.; Dowd, S.E.; Stamatas, G.N.; Nikolovski, J. Diversity of the Human Skin Microbiome Early in Life. J. Investig. Dermatol. 2011, 131, 2026–2032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dréno, B.; Pécastaings, S.; Corvec, S.; Veraldi, S.; Khammari, A.; Roques, C. Cutibacterium acnes (Propionibacterium acnes) and acne vulgaris: A brief look at the latest updates. J. Eur. Acad. Dermatol. Venereol. 2018, 32 (Suppl. 2), 5–14. [Google Scholar] [CrossRef] [Green Version]
- Mourelatos, K.; Eady, E.A.; Cunliffe, W.J.; Clark, S.M.; Cove, J.H. Temporal changes in sebum excretion and propionibacterial colonization in preadolescent children with and without acne. Br. J. Dermatol. 2007, 156, 22–31. [Google Scholar] [CrossRef] [PubMed]
- Shi, B.; Bangayan, N.J.; Curd, E.; Taylor, P.A.; Gallo, R.L.; Leung, D.Y.M.; Li, H. The skin microbiome is different in pediatric versus adult atopic dermatitis. J. Allergy Clin. Immunol. 2016, 138, 1233–1236. [Google Scholar] [CrossRef] [Green Version]
- Jo, J.-H.; Deming, C.; Kennedy, E.A.; Conlan, S.; Polley, E.C.; Ng, W.-I.; Segre, J.A.; Kong, H.H.; NISC Comparative Sequencing Program. Diverse Human Skin Fungal Communities in Children Converge in Adulthood. J. Investig. Dermatol. 2016, 136, 2356–2363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jugé, R.; Rouaud-Tinguely, P.; Breugnot, J.; Servaes, K.; Grimaldi, C.; Roth, M.-P.; Coppin, H.; Closs, B. Shift in skin microbiota of Western European women across aging. J. Appl. Microbiol. 2018, 125, 907–916. [Google Scholar] [CrossRef]
- Zhai, W.; Huang, Y.; Zhang, X.; Fei, W.; Chang, Y.; Cheng, S.; Zhou, Y.; Gao, J.; Tang, X.; Zhang, X.; et al. Profile of the skin microbiota in a healthy Chinese population. J. Dermatol. 2018, 45, 1289–1300. [Google Scholar] [CrossRef]
- Kim, H.-J.; Kim, J.J.; Myeong, N.R.; Kim, T.; Kim, D.; An, S.; Kim, H.; Park, T.; Jang, S.I.; Yeon, J.H.; et al. Segregation of age-related skin microbiome characteristics by functionality. Sci. Rep. 2019, 9, 16748. [Google Scholar] [CrossRef] [Green Version]
- Wu, L.; Zeng, T.; Deligios, M.; Milanesi, L.; Langille, M.G.I.; Zinellu, A.; Rubino, S.; Carru, C.; Kelvin, D.J. Age-Related Variation of Bacterial and Fungal Communities in Different Body Habitats across the Young, Elderly, and Centenarians in Sardinia. mSphere 2020, 5. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Bai, X.; Peng, T.; Yi, X.; Luo, L.; Yang, J.; Liu, J.; Wang, Y.; He, T.; Wang, X.; et al. New Insights into the Skin Microbial Communities and Skin Aging. Front. Microbiol. 2020, 11, 565549. [Google Scholar] [CrossRef]
- Leyden, J.J.; McGiley, K.J.; Mills, O.H.; Kligman, A.M. Age-Related Changes in The Resident Bacterial Flora Of The Human Face. J. Investig. Dermatol. 1975, 65, 379–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed]
- Leung, M.H.Y.; Tong, X.; Bastien, P.; Guinot, F.; Tenenhaus, A.; Appenzeller, B.M.R.; Betts, R.J.; Mezzache, S.; Li, J.; Bourokba, N.; et al. Changes of the human skin microbiota upon chronic exposure to polycyclic aromatic hydrocarbon pollutants. Microbiome 2020, 8, 100. [Google Scholar] [CrossRef]
- Rosenthal, M.; Goldberg, D.; Aiello, A.; Larson, E.; Foxman, B. Skin microbiota: Microbial community structure and its potential association with health and disease. Infect. Genet. Evol. 2011, 11, 839–848. [Google Scholar] [CrossRef] [Green Version]
- Dimitriu, P.A.; Iker, B.; Malik, K.; Leung, H.; Mohn, W.W.; Hillebrand, G.G. New Insights into the Intrinsic and Extrinsic Factors That Shape the Human Skin Microbiome. mBio 2019, 10, e00839-19. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.-J.; Kim, H.; Kim, J.J.; Myeong, N.R.; Kim, T.; Park, T.; Kim, E.; Choi, J.-Y.; Lee, J.; An, S.; et al. Fragile skin microbiomes in megacities are assembled by a predominantly niche-based process. Sci. Adv. 2018, 4, e1701581. [Google Scholar] [CrossRef] [Green Version]
- Clemente, J.C.; Pehrsson, E.C.; Blaser, M.J.; Sandhu, K.; Gao, Z.; Wang, B.; Magris, M.; Hidalgo, G.; Contreras, M.; Noya-Alarcón, Ó.; et al. The microbiome of uncontacted Amerindians. Sci. Adv. 2015, 1, e1500183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Araviiskaia, E.; Berardesca, E.; Bieber, T.; Gontijo, G.; Viera, M.S.; Marrot, L.; Chuberre, B.; Dreno, B. The impact of airborne pollution on skin. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1496–1505. [Google Scholar] [CrossRef]
- Faergemann, J.; Larkö, O. The effect of UV-light on human skin microorganisms. Acta Derm. Venereol. 1987, 67, 69–72. [Google Scholar] [PubMed]
- 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. 2018, 28, 136–141. [Google Scholar] [CrossRef]
- Krutmann, J.; Bouloc, A.; Sore, G.; Bernard, B.A.; Passeron, T. The skin aging exposome. J. Dermatol. Sci. 2017, 85, 152–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.J.; Jeong, S.E.; Lee, S.; Kim, S.; Han, H.; Jeon, C.O. Effects of cosmetics on the skin microbiome of facial cheeks with different hydration levels. Microbiol. Open 2017, 7, e00557. [Google Scholar] [CrossRef]
- Urban, J.; Fergus, D.J.; Savage, A.M.; Ehlers, M.; Menninger, H.L.; Dunn, R.R.; Horvath, J.E. The effect of habitual and ex-perimental antiperspirant and deodorant product use on the armpit microbiome. PeerJ 2016, 4, e1605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Two, A.M.; Nakatsuji, T.; Kotol, P.F.; Arvanitidou, E.; Du-Thumm, L.; Hata, T.R.; Gallo, R.L. The Cutaneous Microbiome and Aspects of Skin Antimicrobial Defense System Resist Acute Treatment with Topical Skin Cleansers. J. Investig. Dermatol. 2016, 136, 1950–1954. [Google Scholar] [CrossRef] [Green Version]
- Bouslimani, A.; da Silva, R.; Kosciolek, T.; Janssen, S.; Callewaert, C.; Amir, A.; Dorrestein, K.; Melnik, A.V.; Zaramela, L.S.; Kim, J.-N.; et al. The impact of skin care products on skin chemistry and microbiome dynamics. BMC Biol. 2019, 17, 47. [Google Scholar] [CrossRef] [PubMed]
- Pinto, D.; Ciardiello, T.; Franzoni, M.; Pasini, F.; Giuliani, G.; Rinaldi, F. Effect of commonly used cosmetic preservatives on skin resident microflora dynamics. Sci. Rep. 2021, 11, 8695. [Google Scholar] [CrossRef]
- Murphy, B.; Hoptroff, M.; Arnold, D.; Eccles, R.; Campbell-Lee, S. In-vivo impact of common cosmetic preservative systems in full formulation on the skin microbiome. PLoS ONE 2021, 16, e0254172. [Google Scholar] [CrossRef]
- Hong, K.-B.; Hong, Y.H.; Jung, E.Y.; Jo, K.; Suh, H.J. Changes in the Diversity of Human Skin Microbiota to Cosmetic Serum Containing Prebiotics: Results from a Randomized Controlled Trial. J. Pers. Med. 2020, 10, 91. [Google Scholar] [CrossRef] [PubMed]
- Misra, N.; Clavaud, C.; Guinot, F.; Bourokba, N.; Nouveau, S.; Mezzache, S.; Palazzi, P.; Appenzeller, B.M.R.; Tenenhaus, A.; Leung, M.H.Y.; et al. Multi-omics analysis to decipher the molecular link between chronic exposure to pollution and human skin dysfunction. Sci. Rep. 2021, 11, 18302. [Google Scholar] [CrossRef] [PubMed]
- Ganju, P.; Nagpal, S.; Mohammed, M.H.; Kumar, P.N.; Pandey, R.; Natarajan, V.T.; Mande, S.S.; Gokhale, R.S. Microbial community profiling shows dysbiosis in the lesional skin of Vitiligo subjects. Sci. Rep. 2016, 6, srep18761. [Google Scholar] [CrossRef] [Green Version]
- Nakatsuji, T.; Gallo, R.L. The role of the skin microbiome in atopic dermatitis. Ann. Allergy, Asthma Immunol. 2018, 122, 263–269. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kong, H.H.; Oh, J.; Deming, C.; Conlan, S.; Grice, E.A.; Beatson, M.A.; Nomicos, E.; Polley, E.C.; Komarow, H.D.; Murray, P.R.; et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012, 22, 850–859. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar]
- Gaitanis, G.; Magiatis, P.; Hantschke, M.; Bassukas, I.D.; Velegraki, A. The Malassezia genus in skin and systemic diseases. Clin. Microbiol. Rev. 2012, 25, 106–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theelen, B.; Cafarchia, C.; Gaitanis, G.; Bassukas, I.D.; Boekhout, T.; Dawson, T.L., Jr. Malassezia ecology, pathophysiology, and treatment. Med. Mycol. 2018, 56 (Suppl. 1), S10–S25. [Google Scholar] [CrossRef] [Green Version]
- Nowicka, D.; Nawrot, U. Contribution of Malassezia spp. to the development of atopic dermatitis. Mycoses 2019, 62, 588–596. [Google Scholar] [CrossRef] [PubMed]
- Amaya, M.; Tajima, M.; Okubo, Y.; Sugita, T.; Nishikawa, A.; Tsuboi, R. Molecular analysis of Malassezia microflora in the lesional skin of psoriasis patients. J. Dermatol. 2007, 34, 619–624. [Google Scholar] [CrossRef]
- Stehlikova, Z.; Kostovcik, M.; Kostovcikova, K.; Kverka, M.; Juzlova, K.; Rob, F.; Hercogova, J.; Bohac, P.; Pinto, Y.; Uzan, A.; et al. Dysbiosis of Skin Microbiota in Psoriatic Patients: Co-occurrence of Fungal and Bacterial Communities. Front. Microbiol. 2019, 10, 438. [Google Scholar] [CrossRef]
- Claudel, J.P.; Auffret, N.; Leccia, M.T.; Poli, F.; Corvec, S.; Dreno, B. Staphylococcus epidermidis: A Potential New Player in the Physiopathology of Acne? Dermatology 2019, 235, 287–294. [Google Scholar] [CrossRef]
- Bek-Thomsen, M.; Lomholt, H.B.; Kilian, M. Acne is Not Associated with Yet-Uncultured Bacteria. J. Clin. Microbiol. 2008, 46, 3355–3360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fitz-Gibbon, S.; Tomida, S.; Chiu, B.H.; Nguyen, L.; Du, C.; Liu, M.; Elashoff, D.; Erfe, M.C.; Loncaric, A.; Kim, J.; et al. Pro-pionibacterium acnes strain populations in the human skin microbiome associated with acne. J. Investig. Dermatol. 2013, 133, 2152–2160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akaza, N.; Akamatsu, H.; Numata, S.; Yamada, S.; Yagami, A.; Nakata, S.; Matsunaga, K. Microorganisms inhabiting fol-licular contents of facial acne are not only Propionibacterium but also Malassezia spp. J. Dermatol. 2016, 43, 906–911. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Li, H. Acne, the Skin Microbiome, and Antibiotic Treatment. Am. J. Clin. Dermatol. 2019, 20, 335–344. [Google Scholar] [CrossRef] [PubMed]
- Yamasaki, K.; Gallo, R.L. The molecular pathology of rosacea. J. Dermatol. Sci. 2009, 55, 77–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murillo, N.; Aubert, J.; Raoult, D. Microbiota of Demodex mites from rosacea patients and controls. Microb. Pathog. 2014, 71–72, 37–40. [Google Scholar] [CrossRef]
- Clavaud, C.; Jourdain, R.; Bar-Hen, A.; Tichit, M.; Bouchier, C.; Pouradier, F.; el Rawadi, C.; Guillot, J.; Menard-Szczebara, F.; Breton, L.; et al. Dandruff is associated with disequilibrium in the proportion of the major bacterial and fungal populations colonizing the scalp. PLoS ONE 2013, 8, e58203. [Google Scholar] [CrossRef]
- An, Q.; Sun, M.; Qi, R.-Q.; Zhang, L.; Zhai, J.-L.; Hong, Y.-X.; Song, B.; Chen, H.-D.; Gao, X.-H. High Staphylococcus epidermidis Colonization and Impaired Permeability Barrier in Facial Seborrheic Dermatitis. Chin. Med J. 2017, 130, 1662–1669. [Google Scholar] [CrossRef]
- Wikramanayake, T.C.; Borda, L.J.; Miteva, M.; Paus, R. Seborrheic dermatitis—Looking beyondMalassezia. Exp. Dermatol. 2019, 28, 991–1001. [Google Scholar] [CrossRef] [Green Version]
- Massiot, P.; Clavaud, C.; Thomas, M.; Ott, A.; Guéniche, A.; Panhard, S.; Muller, B.; Michelin, C.; Kerob, D.; Bouloc, A.; et al. Continuous clinical improvement of mild-to-moderate seborrheic dermatitis and rebalancing of the scalp microbiome using a selenium disulfide–based shampoo after an initial treatment with ketoconazole. J. Cosmet. Dermatol. 2021. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Grice, E.A.; Segre, J.A. The Human Microbiome: Our Second Genome. Annu. Rev. Genom. Hum. Genet. 2012, 13, 151–170. [Google Scholar] [CrossRef] [Green Version]
- Byrd, A.L.; Deming, C.; Cassidy, S.K.B.; Harrison, O.J.; Ng, W.-I.; Conlan, S.; Belkaid, Y.; Segre, J.A.; Kong, H.H.; Program, N.C.S. Staphylococcus aureusandStaphylococcus epidermidisstrain diversity underlying pediatric atopic dermatitis. Sci. Transl. Med. 2017, 9, eaal4651. [Google Scholar] [CrossRef] [Green Version]
- Kong, H.H.; Segre, J.A. Skin Microbiome: Looking Back to Move Forward. J. Investig. Dermatol. 2012, 132, 933–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moran, M.C.; Cahill, M.P.; Brewer, M.G.; Yoshida, T.; Knowlden, S.; Perez-Nazario, N.; Schlievert, P.M.; Beck, L.A. Staphy-lococcal Virulence Factors on the Skin of Atopic Dermatitis Patients. mSphere 2019, 4, e00616-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamazaki, Y.; Nakamura, Y.; Núñez, G. Role of the microbiota in skin immunity and atopic dermatitis. Allergol. Int. 2017, 66, 539–544. [Google Scholar] [CrossRef] [PubMed]
- Bukowski, M.; Wladyka, B.; Dubin, G. Exfoliative Toxins of Staphylococcus aureus. Toxins 2010, 2, 1148–1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iwase, T.; Uehara, Y.; Shinji, H.; Tajima, A.; Seo, H.; Takada, K.; Agata, T.; Mizunoe, Y. Staphylococcus epidermidis Esp inhibits Staphylococcus aureus biofilm formation and nasal colonization. Nature 2010, 465, 346–349. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Cau, L.; Williams, M.R.; Butcher, A.M.; Nakatsuji, T.; Kavanaugh, J.S.; Cheng, J.Y.; Shafiq, F.; Higbee, K.; Hata, T.R.; Horswill, A.R.; et al. Staphylococcus epidermidis protease EcpA can be a deleterious component of the skin microbiome in atopic der-matitis. J. Allergy Clin. Immunol. 2021, 147, 955–966.e16. [Google Scholar] [CrossRef]
- Williams, M.R.; Cau, L.; Wang, Y.; Kaul, D.; Sanford, J.A.; Zaramela, L.S.; Khalil, S.; Butcher, A.M.; Zengler, K.; Horswill, A.R.; et al. Interplay of Staphylococcal and Host Proteases Promotes Skin Barrier Disruption in Netherton Syndrome. Cell Rep. 2020, 30, 2923–2933.e7. [Google Scholar] [CrossRef] [Green Version]
- Lee, Y.B.; Byun, E.J.; Kim, H.S. Potential Role of the Microbiome in Acne: A Comprehensive Review. J. Clin. Med. 2019, 8, 987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saint-Leger, D.; Bague, A.; Lefebvre, E.; Cohen, E.; Chivot, M. A possible role for squalene in the pathogenesis of acne. II. In vivo study of squalene oxides in skin surface and intra-comedonal lipids of acne patients. Br. J. Dermatol. 1986, 114, 543–552. [Google Scholar] [CrossRef] [PubMed]
- Saint-Leger, D.; Bague, A.; Cohen, E.; Lchivot, M. A possible role for squalene in the pathogenesis of acne. I. In vitro study of squalene oxidation. Br. J. Dermatol. 1986, 114, 535–542. [Google Scholar] [CrossRef] [PubMed]
- Jarrousse, V.; Castex-Rizzi, N.; Khammari, A.; Charveron, M.; Dréno, B. Modulation of integrins and filaggrin expression by Propionibacterium acnes extracts on keratinocytes. Arch. Dermatol. Res. 2007, 299, 441–447. [Google Scholar] [CrossRef]
- Isard, O.; Knol, A.C.; Aries, M.F.; Nguyen, J.M.; Khammari, A.; Castex-Rizzi, N.; Dreno, B. Propionibacterium acnes activates the IGF-1/IGF-1R system in the epidermis and induces keratinocyte proliferation. J. Investig. Dermatol. 2011, 131, 59–66. [Google Scholar] [CrossRef] [Green Version]
- Iinuma, K.; Sato, T.; Akimoto, N.; Noguchi, N.; Sasatsu, M.; Nishijima, S.; Kurokawa, I.; Ito, A. Involvement of Propioni-bacterium acnes in the augmentation of lipogenesis in hamster sebaceous glands in vivo and in vitro. J. Investig. Dermatol. 2009, 129, 2113–2119. [Google Scholar] [CrossRef] [Green Version]
- Graham, G.M.; Farrar, M.D.; Cruse-Sawyer, J.E.; Holland, K.T.; Ingham, E. Proinflammatory cytokine production by human keratinocytes stimulated with Propionibacterium acnes and P. acnes GroEL. Br. J. Dermatol. 2004, 150, 421–428. [Google Scholar] [CrossRef] [PubMed]
- Jugeau, S.; Tenaud, I.; Knol, A.C.; Jarrousse, V.; Quereux, G.; Khammari, A.; Dreno, B. Induction of toll-like receptors by Propionibacterium acnes. Br. J. Dermatol. 2005, 153, 1105–1113. [Google Scholar] [CrossRef]
- Johnson, T.; Kang, D.; Barnard, E.; Li, H. Strain-Level Differences in Porphyrin Production and Regulation in Propionibacte-rium acnes Elucidate Disease Associations. mSphere 2016, 1, e00023-15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barnard, E.; Johnson, T.; Ngo, T.; Arora, U.; Leuterio, G.; McDowell, A.; Li, H. Porphyrin Production and Regulation in Cu-taneous Propionibacteria. mSphere 2020, 5, e00793-19. [Google Scholar] [CrossRef] [Green Version]
- Vlachos, C.; Schulte, B.M.; Magiatis, P.; Adema, G.J.; Gaitanis, G. Malassezia-derived indoles activate the aryl hydrocarbon receptor and inhibit Toll-like receptor-induced maturation in monocyte-derived dendritic cells. Br. J. Dermatol. 2012, 167, 496–505. [Google Scholar] [CrossRef] [PubMed]
- Barnard, E.; Shi, B.; Kang, D.; Craft, N.; Li, H. The balance of metagenomic elements shapes the skin microbiome in acne and health. Sci. Rep. 2016, 6, 39491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, Y.J.; Lee, H.K. The Role of Skin and Orogenital Microbiota in Protective Immunity and Chronic Immune-Mediated Inflammatory Disease. Front. Immunol. 2017, 8, 1955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, S.-H.; Yu, H.-Y.; Chang, Y.-C.; Hui, R.C.-Y.; Huang, Y.-C.; Huang, Y.-H. Host characteristics and dynamics of Staphylococcus aureus colonization in patients with moderate-to-severe psoriasis before and after treatment: A prospective cohort study. J. Am. Acad. Dermatol. 2018, 81, 605–607. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Li, J.; Zhu, W.; Kuang, Y.; Liu, T.; Zhang, W.; Chen, X.; Peng, C. Skin and Gut Microbiome in Psoriasis: Gaining Insight into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front. Microbiol. 2020, 11, 3201. [Google Scholar] [CrossRef]
- Polak, K.; Jobbágy, A.; Muszyński, T.; Wojciechowska, K.; Frątczak, A.; Bánvölgyi, A.; Bergler-Czop, B.; Kiss, N. Microbiome Modulation as a Therapeutic Approach in Chronic Skin Diseases. Biomedicines 2021, 9, 1436. [Google Scholar] [CrossRef]
- Dreno, B.; Martin, R.; Khammari, A.; Moyal, D.; Henley, J.B.; Seite, S. Skin Microbiome and Acne Vulgaris: Staphylococcus, a New Actor in Acne. Ski. J. Cutan. Med. 2017, 1, s54. [Google Scholar] [CrossRef] [Green Version]
- Fournière, M.; Latire, T.; Souak, D.; Feuilloley, M.G.J.; Bedoux, G. Staphylococcus epidermidis and Cutibacterium acnes: Two Major Sentinels of Skin Microbiota and the Influence of Cosmetics. Microorganisms 2020, 8, 1752. [Google Scholar] [CrossRef] [PubMed]
- Grice, E.A.; Kong, H.H.; Renaud, G.; Young, A.C.; 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]
- Lauber, C.L.; Zhou, N.; Gordon, J.I.; Knight, R.; Fierer, N. Effect of storage conditions on the assessment of bacterial community structure in soil and human-associated samples. FEMS Microbiol. Lett. 2010, 307, 80–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chng, K.R.; Tay, A.S.L.; Li, C.; Ng, A.H.Q.; Wang, J.; Suri, B.K.; Matta, S.A.; McGovern, N.; Janela, B.; Wong, X.F.C.C.; et al. Whole metagenome profiling reveals skin microbiome-dependent susceptibility to atopic dermatitis flare. Nat. Microbiol. 2016, 1, 16106. [Google Scholar] [CrossRef]
- Salter, S.J.; Cox, M.J.; Turek, E.M.; Calus, S.T.; Cookson, W.O.; Moffatt, M.F.; Turner, P.; Parkhill, J.; Loman, N.J.; Walker, A.W. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 2014, 12, 87. [Google Scholar] [CrossRef] [Green Version]
- Meisel, J.S.; 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. [Google Scholar] [CrossRef]
- Gao, Z.; Perez, G.P.; 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]
- Kong, H.H.; Andersson, B.; Clavel, T.; Common, J.; Jackson, S.; Olson, N.; Segre, J.A.; Traidl-Hoffmann, C. Performing Skin Microbiome Research: A Method to the Madness. J. Investig. Dermatol. 2017, 137, 561–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, J.Z.; Sergeant, M.J.; Lee, O.Y.; Minnikin, D.E.; Besra, G.S.; Pap, I.; Spigelman, M.; Donoghue, H.D.; Pallen, M.J. Meta-genomic analysis of tuberculosis in a mummy. N. Engl. J. Med. 2013, 369, 289–290. [Google Scholar] [CrossRef]
- Pallen, M.J. Diagnostic metagenomics: Potential applications to bacterial, viral and parasitic infections. Parasitology 2014, 141, 1856–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lloyd-Price, J.; Abu-Ali, G.; Huttenhower, C. The healthy human microbiome. Genome Med. 2016, 8, 51. [Google Scholar] [CrossRef] [Green Version]
- Bell, E.C.; Ravis, W.R.; Lloyd, K.B.; Stokes, T.J. Effects of St. John’s wort supplementation on ibuprofen pharmacokinetics. Ann. Pharmacother. 2007, 41, 229–234. [Google Scholar] [CrossRef]
- Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The role of the microbiome in cancer development and therapy. CA Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef] [Green Version]
- Chiu, C.Y.; Miller, S.A. Clinical metagenomics. Nat. Rev. Genet. 2019, 20, 341–355. [Google Scholar] [CrossRef] [PubMed]
- Johnson, C.H.; Ivanisevic, J.; Siuzdak, G. Metabolomics: Beyond biomarkers and towards mechanisms. Nat. Rev. Mol. Cell Biol. 2016, 17, 451–459. [Google Scholar] [CrossRef] [Green Version]
- Chen, H.; Nwe, P.-K.; Yang, Y.; Rosen, C.E.; Bielecka, A.A.; Kuchroo, M.; Cline, G.W.; Kruse, A.C.; Ring, A.M.; Crawford, J.M.; et al. A Forward Chemical Genetic Screen Reveals Gut Microbiota Metabolites That Modulate Host Physiology. Cell 2019, 177, 1217–1231.e18. [Google Scholar] [CrossRef] [PubMed]
- Zierer, J.; Jackson, M.A.; Kastenmüller, G.; Mangino, M.; Long, T.; Telenti, A.; Mohney, R.P.; Small, K.S.; Bell, J.T.; Steves, C.J.; et al. The fecal metabolome as a functional readout of the gut microbiome. Nat. Genet. 2018, 50, 790–795. [Google Scholar] [CrossRef]
- Jeffery, I.B.; Das, A.; O’Herlihy, E.; Coughlan, S.; Cisek, K.; Moore, M.; Bradley, F.; Carty, T.; Pradhan, M.; Dwibedi, C.; et al. Differences in Fecal Microbiomes and Metabolomes of People With vs Without Irritable Bowel Syndrome and Bile Acid Mal-absorption. Gastroenterology 2020, 158, 1016–1028.e8. [Google Scholar] [CrossRef]
- Dumas, M.-E.; Barton, R.H.; Toye, A.; Cloarec, O.; Blancher, C.; Rothwell, A.; Fearnside, J.; Tatoud, R.; Blanc, V.; Lindon, J.C.; et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc. Natl. Acad. Sci. USA 2006, 103, 12511–12516. [Google Scholar] [CrossRef] [Green Version]
- Metwaly, A.; Dunkel, A.; Waldschmitt, N.; Raj, A.C.D.; Lagkouvardos, I.; Corraliza, A.M.; Mayorgas, A.; Martinez-Medina, M.; Reiter, S.; Schloter, M.; et al. Integrated microbiota and metabolite profiles link Crohn’s disease to sulfur metabolism. Nat. Commun. 2020, 11, 4322. [Google Scholar] [CrossRef] [PubMed]
- Zhou, W.; Sailani, M.R.; Contrepois, K.; Zhou, Y.; Ahadi, S.; Leopold, S.R.; Zhang, M.J.; Rao, V.; Avina, M.; Mishra, T.; et al. Longitudinal multi-omics of host–microbe dynamics in prediabetes. Nature 2019, 569, 663–671. [Google Scholar] [CrossRef] [PubMed]
- Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2018, 4, 293–305. [Google Scholar] [CrossRef] [PubMed]
- Sadowski, T.; Klose, C.; Gerl, M.J.; Wójcik-Maciejewicz, A.; Herzog, R.; Simons, K.; Reich, A.; Surma, M.A. Large-scale human skin lipidomics by quantitative, high-throughput shotgun mass spectrometry. Sci. Rep. 2017, 7, srep43761. [Google Scholar] [CrossRef] [Green Version]
- Kuehne, A.; Hildebrand, J.; Soehle, J.; Wenck, H.; Terstegen, L.; Gallinat, S.; Knott, A.; Winnefeld, M.; Zamboni, N. An integrative metabolomics and transcriptomics study to identify metabolic alterations in aged skin of humans in vivo. BMC Genom. 2017, 18, 169. [Google Scholar] [CrossRef] [Green Version]
- Bouslimani, A.; Porto, C.; Rath, C.M.; Wang, M.; Guo, Y.; Gonzalez, A.; Berg-Lyon, D.; Ackermann, G.; Christensen, G.J.M.; Nakatsuji, T.; et al. Molecular cartography of the human skin surface in 3D. Proc. Natl. Acad. Sci. USA 2015, 112, E2120–E2129. [Google Scholar] [CrossRef] [Green Version]
- Watrous, J.; Roach, P.; Alexandrov, T.; Heath, B.S.; Yang, J.Y.; Kersten, R.D.; van der Voort, M.; Pogliano, K.; Gross, H.; Raaijmakers, J.M.; et al. Mass spectral molecular networking of living microbial colonies. Proc. Natl. Acad. Sci. USA 2012, 109, E1743–E1752. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.; Carver, J.J.; Phelan, V.V.; Sanchez, L.M.; Garg, N.; Peng, Y.; Nguyen, D.D.; Watrous, J.; Kapono, C.A.; Luzzatto-Knaan, T.; et al. Sharing and community curation of mass spectrometry data with Global Natural Products Social Molecular Networking. Nat. Biotechnol. 2016, 34, 828–837. [Google Scholar] [CrossRef] [Green Version]
- Leao, T.F.; Clark, C.M.; Bauermeister, A.; Elijah, E.O.; Gentry, E.C.; Husband, M.; Oliveira, M.F.; Bandeira, N.; Wang, M.; Dorrestein, P.C. Quick-start infrastructure for untargeted metabolomics analysis in GNPS. Nat. Metab. 2021, 3, 880–882. [Google Scholar] [CrossRef]
- Jarmusch, A.K.; Vrbanac, A.; Momper, J.D.; Ma, J.D.; Alhaja, M.; Liyanage, M.; Knight, R.; Dorrestein, P.C.; Tsunoda, S.M. Enhanced Characterization of Drug Metabolism and the Influence of the Intestinal Microbiome: A Pharmacokinetic, Microbiome, and Untargeted Metabolomics Study. Clin. Transl. Sci. 2020, 13, 972–984. [Google Scholar] [CrossRef] [Green Version]
- Garg, N.; Wang, M.; Hyde, E.; da Silva, R.R.; Melnik, A.V.; Protsyuk, I.; Bouslimani, A.; Lim, Y.W.; Wong, R.; Humphrey, G.; et al. Three-Dimensional Microbiome and Metabolome Cartography of a Diseased Human Lung. Cell Host Microbe 2017, 22, 705–716.e4. [Google Scholar] [CrossRef]
- Ding, L.; Bar-Shalom, R.; Aharonovich, D.; Kurisawa, N.; Patial, G.; Li, S.; He, S.; Yan, X.; Iwasaki, A.; Suenaga, K.; et al. Metabolomic Characterization of a cf. Neolyngbya Cyanobacterium from the South China Sea Reveals Wenchangamide A, a Lipopeptide with In Vitro Apoptotic Potential in Colon Cancer Cells. Mar. Drugs 2021, 19, 397. [Google Scholar] [CrossRef] [PubMed]
- Luzzatto-Knaan, T.; Garg, N.; Wang, M.; Glukhov, E.; Peng, Y.; Ackermann, G.; Amir, A.; Duggan, B.M.; Ryazanov, S.; Gerwick, L.; et al. Digitizing mass spectrometry data to explore the chemical diversity and distribution of marine cyanobacteria and algae. eLife 2017, 6, e24214. [Google Scholar] [CrossRef] [PubMed]
- Petras, D.; Minich, J.J.; Cancelada, L.B.; Torres, R.R.; Kunselman, E.; Wang, M.; White, M.E.; Allen, E.E.; Prather, K.A.; Aluwihare, L.I.; et al. Non-targeted tandem mass spectrometry enables the visualization of organic matter chemotype shifts in coastal seawater. Chemosphere 2021, 271, 129450. [Google Scholar] [CrossRef] [PubMed]
- Kapono, C.A.; Morton, J.T.; Bouslimani, A.; Melnik, A.V.; Orlinsky, K.; Knaan, T.L.; Garg, N.; Vázquez-Baeza, Y.; Protsyuk, I.; Janssen, S.; et al. Creating a 3D microbial and chemical snapshot of a human habitat. Sci. Rep. 2018, 8, 3669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McCall, L.-I.; Callewaert, C.; Zhu, Q.; Song, S.J.; Bouslimani, A.; Minich, J.J.; Ernst, M.; Ruiz-Calderon, J.F.; Cavallin, H.; Pereira, H.S.; et al. Home chemical and microbial transitions across urbanization. Nat. Microbiol. 2019, 5, 108–115. [Google Scholar] [CrossRef]
- Jarmusch, A.K.; Elijah, E.O.; Vargas, F.; Bouslimani, A.; da Silva, R.R.; Ernst, M.; Wang, M.; Del Rosario, K.K.; Dorrestein, P.C.; Tsunoda, S.M. Initial Development toward Non-Invasive Drug Monitoring via Untargeted Mass Spectrometric Analysis of Human Skin. Anal. Chem. 2019, 91, 8062–8069. [Google Scholar] [CrossRef] [PubMed]
- Bouslimani, A.; Melnik, A.V.; Xu, Z.; Amir, A.; da Silva, R.R.; Wang, M.; Bandeira, N.; Alexandrov, T.; Knight, R.; Dorrestein, P.C. Lifestyle chemistries from phones for individual profiling. Proc. Natl. Acad. Sci. USA 2016, 113, E7645–E7654. [Google Scholar] [CrossRef] [Green Version]
- Jarmusch, A.K.; Wang, M.; Aceves, C.M.; Advani, R.S.; Aguirre, S.; Aksenov, A.A.; Aleti, G.; Aron, A.T.; Bauermeister, A.; Bolleddu, S.; et al. ReDU: A framework to find and reanalyze public mass spectrometry data. Nat. Methods 2020, 17, 901–904. [Google Scholar] [CrossRef]
- Nothias, L.-F.; Petras, D.; Schmid, R.; Dührkop, K.; Rainer, J.; Sarvepalli, A.; Protsyuk, I.; Ernst, M.; Tsugawa, H.; Fleischauer, M.; et al. Feature-based molecular networking in the GNPS analysis environment. Nat. Methods 2020, 17, 905–908. [Google Scholar] [CrossRef]
- Tripathi, A.; Vázquez-Baeza, Y.; Gauglitz, J.M.; Wang, M.; Dührkop, K.; Nothias-Esposito, M.; Acharya, D.D.; Ernst, M.; van der Hooft, J.J.J.; Zhu, Q.; et al. Chemically informed analyses of metabolomics mass spectrometry data with Qemistree. Nat. Chem. Biol. 2020, 17, 146–151. [Google Scholar] [CrossRef] [PubMed]
- Da Silva, R.R.; Wang, M.; Nothias, L.-F.; Van Der Hooft, J.J.J.; Caraballo-Rodríguez, A.M.; Fox, E.; Balunas, M.J.; Klassen, J.L.; Lopes, N.P.; Dorrestein, P.C. Propagating annotations of molecular networks using in silico fragmentation. PLOS Comput. Biol. 2018, 14, e1006089. [Google Scholar] [CrossRef] [PubMed]
- Sugimoto, Y.; Camacho, F.R.; Wang, S.; Chankhamjon, P.; Odabas, A.; Biswas, A.; Jeffrey, P.D.; Donia, M.S. A metagenomic strategy for harnessing the chemical repertoire of the human microbiome. Science 2019, 366, eaax9176. [Google Scholar] [CrossRef]
- Blin, K.; Shaw, S.; Steinke, K.; Villebro, R.; Ziemert, N.; Lee, S.Y.; Medema, M.H.; Weber, T. antiSMASH 5.0: Updates to the secondary metabolite genome mining pipeline. Nucleic Acids Res. 2019, 47, W81–W87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khaldi, N.; Seifuddin, F.T.; Turner, G.; Haft, D.; Nierman, W.C.; Wolfe, K.H.; Fedorova, N.D. SMURF: Genomic mapping of fungal secondary metabolite clusters. Fungal Genet. Biol. 2010, 47, 736–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skinnider, M.A.; Merwin, N.J.; Johnston, C.W.; Magarvey, N.A. PRISM 3: Expanded prediction of natural product chemical structures from microbial genomes. Nucleic Acids Res. 2017, 45, W49–W54. [Google Scholar] [CrossRef] [Green Version]
- Cao, L.; Gurevich, A.; Alexander, K.L.; Naman, C.B.; Leão, T.; Glukhov, E.; Luzzatto-Knaan, T.; Vargas, F.; Quinn, R.; Bouslimani, A.; et al. MetaMiner: A Scalable Peptidogenomics Approach for Discovery of Ribosomal Peptide Natural Products with Blind Modifications from Microbial Communities. Cell Syst. 2019, 9, 600–608.e4. [Google Scholar] [CrossRef]
- Behsaz, B.; Bode, E.; Gurevich, A.; Shi, Y.N.; Grundmann, F.; Acharya, D.; Caraballo-Rodriguez, A.M.; Bouslimani, A.; Pa-nitchpakdi, M.; Linck, A.; et al. Integrating genomics and metabolomics for scalable non-ribosomal peptide discovery. Nat. Commun. 2021, 12, 3225. [Google Scholar] [CrossRef]
- Mohimani, H.; Liu, W.-T.; Kersten, R.D.; Moore, B.S.; Dorrestein, P.C.; Pevzner, P.A. NRPquest: Coupling Mass Spectrometry and Genome Mining for Nonribosomal Peptide Discovery. J. Nat. Prod. 2014, 77, 1902–1909. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Guo, J.; Dai, S.; Ouyang, Y.; Wu, H.; Sun, W.; Wang, G. Exploring and exploiting microbial diversity through meta-genomics for natural product drug discovery. Curr. Top. Med. Chem. 2009, 9, 1525–1535. [Google Scholar] [CrossRef]
- Chavira, A.; Belda-Ferre, P.; Kosciolek, T.; Ali, F.; Dorrestein, P.C.; Knight, R. The Microbiome and Its Potential for Pharma-cology. Handb. Exp. Pharmacol. 2019, 260, 301–326. [Google Scholar]
- Culligan, E.P.; Sleator, R.D.; Marchesi, J.R.; Hill, C. Metagenomics and novel gene discovery: Promise and potential for novel therapeutics. Virulence 2014, 5, 399–412. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Liu, Y.; Du, Z.; Zhang, L.; Chen, J.; Shen, Z.; Liu, Q.; Qin, J.; Lv, H.; Wang, H.; et al. Skin microbiota analysis-inspired development of novel anti-infectives. Microbiome 2020, 8, 85. [Google Scholar] [CrossRef]
- Mathieu, A.; Vogel, T.M.; Simonet, P. The future of skin metagenomics. Res. Microbiol. 2014, 165, 69–76. [Google Scholar] [CrossRef] [PubMed]
- Van den Bogert, B.; Boekhorst, J.; Pirovano, W.; May, A. On the Role of Bioinformatics and Data Science in Industrial Microbiome Applications. Front. Genet. 2019, 10, 721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.; Li, L.; Butcher, J.; Stintzi, A.; Figeys, D. Advancing functional and translational microbiome research using meta-omics approaches. Microbiome 2019, 7, 154. [Google Scholar] [CrossRef]
- Heinken, A.; Thiele, I. Systems biology of host-microbe metabolomics. Wiley Interdiscip. Rev. Syst. Biol. Med. 2015, 7, 195–219. [Google Scholar] [CrossRef]
- Nielsen, J. Systems Biology of Metabolism: A Driver for Developing Personalized and Precision Medicine. Cell Metab. 2017, 25, 572–579. [Google Scholar] [CrossRef] [Green Version]
- Magnusdottir, S.; Thiele, I. Modeling metabolism of the human gut microbiome. Curr. Opin. Biotechnol. 2018, 51, 90–96. [Google Scholar] [CrossRef]
- Cakir, T.; Panagiotou, G.; Uddin, R.; Durmus, S. Novel Approaches for Systems Biology of Metabolism-Oriented Patho-gen-Human Interactions: A Mini-Review. Front. Cell Infect. Microbiol. 2020, 10, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernabé, B.P.; Cralle, L.; Gilbert, J.A. Systems biology of the human microbiome. Curr. Opin. Biotechnol. 2018, 51, 146–153. [Google Scholar] [CrossRef] [Green Version]
- Son, J.W.; Shoaie, S.; Lee, S. Systems Biology: A Multi-Omics Integration Approach to Metabolism and the Microbiome. Endocrinol. Metab. 2020, 35, 507–514. [Google Scholar] [CrossRef] [PubMed]
- Francke, C.; Siezen, R.J.; Teusink, B. Reconstructing the metabolic network of a bacterium from its genome. Trends Microbiol. 2005, 13, 550–558. [Google Scholar] [CrossRef] [PubMed]
- Oberhardt, M.A.; Palsson, B.O.; Papin, J.A. Applications of genome-scale metabolic reconstructions. Mol. Syst. Biol. 2009, 5, 320. [Google Scholar] [CrossRef]
- Duarte, N.C.; Becker, S.A.; Jamshidi, N.; Thiele, I.; Mo, M.L.; Vo, T.D.; Srivas, R.; Palsson, B. Global reconstruction of the human metabolic network based on genomic and bibliomic data. Proc. Natl. Acad. Sci. USA 2007, 104, 1777–1782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brunk, E.; Sahoo, S.; Zielinski, D.C.; Altunkaya, A.; Dräger, A.; Mih, N.; Gatto, F.; Nilsson, A.; Gonzalez, G.A.P.; Aurich, M.K.; et al. Recon3D enables a three-dimensional view of gene variation in human metabolism. Nat. Biotechnol. 2018, 36, 272–281. [Google Scholar] [CrossRef]
- Haraldsdóttir, H.S.; Thiele, I.; Fleming, R.M. Comparative evaluation of open source software for mapping between metabolite identifiers in metabolic network reconstructions: Application to Recon 2. J. Chemin- 2014, 6, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Magnusdottir, S.; Heinken, A.; Kutt, L.; Ravcheev, D.A.; Bauer, E.; Noronha, A.; Greenhalgh, K.; Jäger, C.; Baginska, J.; Wilmes, P.; et al. Generation of genome-scale metabolic reconstructions for 773 members of the human gut microbiota. Nat. Biotechnol. 2016, 35, 81–89. [Google Scholar] [CrossRef] [PubMed]
- Hussan, J.R.; Hunter, P.J. Our natural “makeup” reveals more than it hides: Modeling the skin and its microbiome. WIREs Mech. Dis. 2020, 13, e1497. [Google Scholar] [CrossRef]
- Thiele, I.; Palsson, B.O. A protocol for generating a high-quality genome-scale metabolic reconstruction. Nat. Protoc. 2010, 5, 93–121. [Google Scholar] [CrossRef] [Green Version]
- E Larsen, P.; Collart, F.R.; Field, D.; Meyer, F.; Keegan, K.P.; Henry, C.S.; McGrath, J.; Quinn, J.; A Gilbert, J. Predicted Relative Metabolomic Turnover (PRMT): Determining metabolic turnover from a coastal marine metagenomic dataset. Microb. Informatics Exp. 2011, 1, 4. [Google Scholar] [CrossRef] [PubMed]
- Noecker, C.; Eng, A.; Srinivasan, S.; Theriot, C.M.; Young, V.B.; Jansson, J.K.; Fredricks, D.N.; Borenstein, E. Metabolic Mod-el-Based Integration of Microbiome Taxonomic and Metabolomic Profiles Elucidates Mechanistic Links between Ecological and Metabolic Variation. mSystems 2016, 1, e00013-15. [Google Scholar] [CrossRef] [Green Version]
- Chang, Y.-L.; Rossetti, M.; Vlamakis, H.; Casero, D.; Sunga, G.; Harre, N.; Miller, S.; Humphries, R.; Stappenbeck, T.; Simpson, K.W.; et al. A screen of Crohn’s disease-associated microbial metabolites identifies ascorbate as a novel metabolic inhibitor of activated human T cells. Mucosal Immunol. 2018, 12, 457–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mallick, H.; Franzosa, E.A.; Mclver, L.J.; Banerjee, S.; Sirota-Madi, A.; Kostic, A.D.; Clish, C.B.; Vlamakis, H.; Xavier, R.J.; Huttenhower, C. Predictive metabolomic profiling of microbial communities using amplicon or metagenomic sequences. Nat. Commun. 2019, 10, 3136. [Google Scholar] [CrossRef] [Green Version]
- Yin, X.; Altman, T.; Rutherford, E.; West, K.A.; Wu, Y.; Choi, J.; Beck, P.L.; Kaplan, G.G.; Dabbagh, K.; DeSantis, T.Z.; et al. A Comparative Evaluation of Tools to Predict Metabolite Profiles From Microbiome Sequencing Data. Front. Microbiol. 2020, 11, 3132. [Google Scholar] [CrossRef] [PubMed]
- Zmora, N.; Soffer, E.; Elinav, E. Transforming medicine with the microbiome. Sci. Transl. Med. 2019, 11, eaaw1815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jobin, C. Precision medicine using microbiota. Science 2018, 359, 32–34. [Google Scholar] [CrossRef]
- Mills, S.; Stanton, C.; Lane, J.A.; Smith, G.J.; Ross, R.P. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019, 11, 923. [Google Scholar] [CrossRef] [Green Version]
- Guarner, F.; Schaafsma, G.J. Probiotics. Int. J. Food Microbiol. 1998, 39, 237–238. [Google Scholar] [CrossRef]
- Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
- Wieërs, G.; Belkhir, L.; Enaud, R.; Leclercq, S.; De Foy, J.-M.P.; Dequenne, I.; De Timary, P.; Cani, P.D. How Probiotics Affect the Microbiota. Front. Cell. Infect. Microbiol. 2020, 9, 454. [Google Scholar] [CrossRef] [Green Version]
- Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef] [PubMed]
- Report of the ICCR: ICCR Microbiome and Cosmetics—Survey of Products Ingredients Terminologies and Regulatory Approaches. 2021. Available online: https://www.iccr-cosmetics.org/topics-documents/14-microbiome (accessed on 25 November 2021).
- 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] [PubMed]
- Gueniche, A.; Knaudt, B.; Schuck, E.; Volz, T.; Bastien, P.; Martin, R.; Rocken, M.; Breton, L.; Biedermann, T. Effects of non-pathogenic gram-negative bacterium Vitreoscilla filiformis lysate on atopic dermatitis: A prospective, randomized, double-blind, placebo-controlled clinical study. Br. J. Dermatol. 2008, 159, 1357–1363. [Google Scholar] [CrossRef] [PubMed]
- Peguet-Navarro, J.; Dezutter-Dambuyant, C.; Buetler, T.; LeClaire, J.; Smola, H.; Blum, S.; Bastien, P.; Breton, L.; Gueniche, A. Supplementation with oral probiotic bacteria protects human cutaneous immune homeostasis after UV exposure-double blind, randomized, placebo controlled clinical trial. Eur. J. Dermatol. 2008, 18. [Google Scholar] [CrossRef]
- Gueniche, A.; Philippe, D.; Bastien, P.; Blum, S.; Buyukpamukcu, E.; Castiel-Higounenc, I. Probiotics for photoprotection. Dermatoendocrinology 2009, 1, 275–279. [Google Scholar] [CrossRef] [Green Version]
- Guéniche, A.; Bastien, P.; Ovigne, J.M.; Kermici, M.; Courchay, G.; Chevalier, V.; Breton, L.; Castiel-Higounenc, I. Bifidobacterium longum lysate, a new ingredient for reactive skin. Exp. Dermatol. 2009, 19, e1–e8. [Google Scholar] [CrossRef]
- Gueniche, A.; Benyacoub, J.; Philippe, D.; Bastien, P.; Kusy, N.; Breton, L.; Blum, S.; Castiel-Higounenc, I. Lactobacillus par-acasei CNCM I-2116 (ST11) inhibits substance P-induced skin inflammation and accelerates skin barrier function recovery in vitro. Eur. J. Dermatol. 2010, 20, 731–737. [Google Scholar]
- Li, L.; Han, Z.; Niu, X.; Zhang, G.; Jia, Y.; Zhang, S.; He, C. Probiotic Supplementation for Prevention of Atopic Dermatitis in Infants and Children: A Systematic Review and Meta-analysis. Am. J. Clin. Dermatol. 2018, 20, 367–377. [Google Scholar] [CrossRef]
- Reygagne, P.; Bastien, P.; Couavoux, M.P.; Philippe, D.; Renouf, M.; Castiel-Higounenc, I.; Gueniche, A. The positive benefit of Lactobacillus paracasei NCC2461 ST11 in healthy volunteers with moderate to severe dandruff. Benef. Microbes 2017, 8, 671–680. [Google Scholar] [CrossRef]
- Benyacoub, J.; Bosco, N.; Blanchard, C.; Demont, A.; Philippe, D.; Castiel-Higounenc, I.; Gueniche, A. Immune modulation property of Lactobacillus paracasei NCC2461 (ST11) strain and impact on skin defences. Benef. Microbes 2014, 5, 129–136. [Google Scholar] [CrossRef] [PubMed]
- Gueniche, A.; Philippe, D.; Bastien, P.; Reuteler, G.; Blum, S.; Castiel-Higounenc, I.; Breton, L.; Benyacoub, J. Randomised double-blind placebo-controlled study of the effect of Lactobacillus paracasei NCC 2461 on skin reactivity. Benef. Microbes 2014, 5, 137–145. [Google Scholar] [CrossRef]
- Bouilly-Gauthier, D.; Jeannes, C.; Maubert, Y.; Duteil, L.; Queille-Roussel, C.; Piccardi, N.; Montastier, C.; Manissier, P.; Pierard, G.; Ortonne, J.P. Clinical evidence of benefits of a dietary supplement containing probiotic and carotenoids on ultravio-let-induced skin damage. Br. J. Dermatol. 2010, 163, 536–543. [Google Scholar] [CrossRef] [PubMed]
- Kober, M.-M.; Bowe, W.P. The effect of probiotics on immune regulation, acne, and photoaging. Int. J. Women’s Dermatol. 2015, 1, 85–89. [Google Scholar] [CrossRef] [Green Version]
- Han, N.; Jia, L.; Guo, L.; Su, Y.; Luo, Z.; Du, J.; Mei, S.; Liu, Y. Balanced oral pathogenic bacteria and probiotics promoted wound healing via maintaining mesenchymal stem cell homeostasis. Stem Cell Res. Ther. 2020, 11, 61. [Google Scholar] [CrossRef]
- Kunz, A.N.; Noel, J.M.; Fairchok, M.P. Two Cases of Lactobacillus Bacteremia During Probiotic Treatment of Short Gut Syndrome. J. Pediatr. Gastroenterol. Nutr. 2004, 38, 457–458. [Google Scholar] [CrossRef]
- Yelin, I.; Flett, K.B.; Merakou, C.; Mehrotra, P.; Stam, J.; Snesrud, E.; Hinkle, M.; Lesho, E.; McGann, P.; McAdam, A.J.; et al. Genomic and epidemiological evidence of bacterial transmission from probiotic capsule to blood in ICU patients. Nat. Med. 2019, 25, 1728–1732. [Google Scholar] [CrossRef]
- Galdeano, C.M.; Perdigon, G. Role of viability of probiotic strains in their persistence in the gut and in mucosal immune stimulation. J. Appl. Microbiol. 2004, 97, 673–681. [Google Scholar] [CrossRef]
- Sawada, J.; Morita, H.; Tanaka, A.; Salminen, S.; He, F.; Matsuda, H. Ingestion of heat-treated Lactobacillus rhamnosus GG prevents development of atopic dermatitis in NC/Nga mice. Clin. Exp. Allergy 2006, 37, 296–303. [Google Scholar] [CrossRef]
- França, K. Topical Probiotics in Dermatological Therapy and Skincare: A Concise Review. Dermatol. Ther. 2020, 11, 71–77. [Google Scholar] [CrossRef] [PubMed]
- Al-Ghazzewi, F.H.; Tester, R.F. Impact of prebiotics and probiotics on skin health. Benef. Microbes 2014, 5, 99–107. [Google Scholar] [CrossRef] [PubMed]
- Lopes, E.G.; Moreira, D.A.; Gullón, P.; Gullón, B.; Cardelle-Cobas, A.; Tavaria, F.K. Topical application of probiotics in skin: Adhesion, antimicrobial and antibiofilmin vitroassays. J. Appl. Microbiol. 2017, 122, 450–461. [Google Scholar] [CrossRef]
- Lim, H.Y.; Jeong, D.; Park, S.H.; Shin, K.K.; Hong, Y.H.; Kim, E.; Yu, Y.-G.; Kim, T.-R.; Kim, H.; Lee, J.; et al. Antiwrinkle and Antimelanogenesis Effects of Tyndallized Lactobacillus acidophilus KCCM12625P. Int. J. Mol. Sci. 2020, 21, 1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brandi, J.; Cheri, S.; Manfredi, M.; Di Carlo, C.; Vanella, V.V.; Federici, F.; Bombiero, E.; Bazaj, A.; Rizzi, E.; Manna, L.; et al. Exploring the wound healing, anti-inflammatory, anti-pathogenic and proteomic effects of lactic acid bacteria on keratinocytes. Sci. Rep. 2020, 10, 11572. [Google Scholar] [CrossRef] [PubMed]
- Jung, Y.-O.; Jeong, H.; Cho, Y.; Lee, E.-O.; Jang, H.-W.; Kim, J.; Nam, K.T.; Lim, K.-M. Lysates of a Probiotic, Lactobacillus rhamnosus, Can Improve Skin Barrier Function in a Reconstructed Human Epidermis Model. Int. J. Mol. Sci. 2019, 20, 4289. [Google Scholar] [CrossRef] [Green Version]
- Dimarzio, L.; Cinque, B.; Cupelli, F.; De Simone, C.; Cifone, M.G.; Giuliani, M. Increase of Skin-Ceramide Levels in Aged Subjects following a Short-Term Topical Application of Bacterial Sphingomyelinase from Streptococcus Thermophilus. Int. J. Immunopathol. Pharmacol. 2008, 21, 137–143. [Google Scholar] [CrossRef]
- Volz, T.; Skabytska, Y.; Guenova, E.; Chen, K.M.; Frick, J.S.; Kirschning, C.J.; Kaesler, S.; Rocken, M.; Biedermann, T. Non-pathogenic 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]
- Ding, T.; Schloss, P.D. Dynamics and associations of microbial community types across the human body. Nature 2014, 509, 357–360. [Google Scholar] [CrossRef]
- Gallo, R.L.; Hooper, L.V. Epithelial antimicrobial defence of the skin and intestine. Nat. Rev. Immunol. 2012, 12, 503–516. [Google Scholar] [CrossRef] [Green Version]
- Nodake, Y.; Matsumoto, S.; Miura, R.; Honda, H.; Ishibashi, G.; Matsumoto, S.; Dekio, I.; Sakakibara, R. Pilot study on novel skin care method by augmentation with Staphylococcus epidermidis, an autologous skin microbe—A blinded randomized clinical trial. J. Dermatol. Sci. 2015, 79, 119–126. [Google Scholar] [CrossRef]
- Hols, P.; Ledesma-García, L.; Gabant, P.; Mignolet, J. Mobilization of Microbiota Commensals and Their Bacteriocins for Therapeutics. Trends Microbiol. 2019, 27, 690–702. [Google Scholar] [CrossRef] [Green Version]
- Cogen, A.L.; Yamasaki, K.; Sanchez, K.M.; Dorschner, R.A.; Lai, Y.; MacLeod, D.T.; Torpey, J.W.; Otto, M.; Nizet, V.; Kim, J.E.; et al. Selective Antimicrobial Action Is Provided by Phenol-Soluble Modulins Derived from Staphylococcus epidermidis, a Normal Resident of the Skin. J. Investig. Dermatol. 2010, 130, 192–200. [Google Scholar] [CrossRef] [Green Version]
- Drake, D.R.; Brogden, K.A.; Dawson, D.V.; Wertz, P.W. Thematic Review Series: Skin Lipids. Antimicrobial lipids at the skin surface. J. Lipid Res. 2008, 49, 4–11. [Google Scholar] [CrossRef] [Green Version]
- Thiboutot, D.M. Inflammasome Activation by Propionibacterium acnes: The Story of IL-1 in Acne Continues to Unfold. J. Investig. Dermatol. 2014, 134, 595–597. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Wang, W.; Wu, Y.; Ma, X.; Zhou, W.; Lai, Y. Lipopeptide 78 from Staphylococcus epidermidis Activates beta-Catenin to Inhibit Skin Inflammation. J. Immunol. 2019, 202, 1219–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bitschar, K.; Staudenmaier, L.; Klink, L.; Focken, J.; Sauer, B.; Fehrenbacher, B.; Herster, F.; Bittner, Z.; Bleul, L.; Schaller, M.; et al. Staphylococcus aureus Skin Colonization Is Enhanced by the Interaction of Neutrophil Extracellular Traps with Keratinocytes. J. Investig. Dermatol. 2019, 140, 1054–1065.e4. [Google Scholar] [CrossRef] [PubMed]
- Paetzold, B.; Willis, J.R.; Pereira de Lima, J.; Knodlseder, N.; Bruggemann, H.; Quist, S.R.; Gabaldon, T.; Guell, M. Skin microbiome modulation induced by probiotic solutions. Microbiome 2019, 7, 95. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.E.; Fischbach, M.A.; Belkaid, Y. Skin microbiota-host interactions. Nature 2018, 553, 427–436. [Google Scholar] [CrossRef]
- Shu, M.; Wang, Y.; Yu, J.; Kuo, S.; Coda, A.; Jiang, Y.; Gallo, R.L.; Huang, C.-M. Fermentation of Propionibacterium acnes, a Commensal Bacterium in the Human Skin Microbiome, as Skin Probiotics against Methicillin-Resistant Staphylococcus aureus. PLoS ONE 2013, 8, e55380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Youn, S.H.; Choi, C.W.; Choi, J.W.; Youn, S.W. The skin surface pH and its different influence on the development of acne lesion according to gender and age. Ski. Res. Technol. 2012, 19, 131–136. [Google Scholar] [CrossRef]
- Allhorn, M.; Arve-Butler, S.; Brüggemann, H.; Lood, R. A novel enzyme with antioxidant capacity produced by the ubiquitous skin colonizer Propionibacterium acnes. Sci. Rep. 2016, 6, 36412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dreno, B.; Bagatin, E.; Blume-Peytavi, U.; Rocha, M.; Gollnick, H. Female type of adult acne: Physiological and psychological considerations and management. J. der Dtsch. Dermatol. Ges. 2018, 16, 1185–1194. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Nakamura, K.; O’Neill, A.M.; Williams, M.R.; Cau, L.; Nakatsuji, T.; Horswill, A.R.; Gallo, R.L. Short chain fatty acids produced by Cutibacterium acnes inhibit biofilm formation by Staphylococcus epidermidis. Sci. Rep. 2020, 10, 21237. [Google Scholar] [CrossRef] [PubMed]
- Christensen, G.J.M.; Scholz, C.F.P.; Enghild, J.; Rohde, H.; Kilian, M.; Thürmer, A.; Brzuszkiewicz, E.; Lomholt, H.B.; Brüggemann, H. Antagonism between Staphylococcus epidermidis and Propionibacterium acnes and its genomic basis. BMC Genom. 2016, 17, 152. [Google Scholar] [CrossRef] [Green Version]
- Belkaid, Y.; Tamoutounour, S. The influence of skin microorganisms on cutaneous immunity. Nat. Rev. Immunol. 2016, 16, 353–366. [Google Scholar] [CrossRef]
- 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. Nat. Cell Biol. 2015, 520, 104–108. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Mascarenhas, N.; Eckmann, L.; Miyamoto, Y.; Sun, X.; Kawakami, T.; Di Nardo, A. Skin microbiome promotes mast cell maturation by triggering stem cell factor production in keratinocytes. J. Allergy Clin. Immunol. 2016, 139, 1205–1216.e6. [Google Scholar] [CrossRef] [Green Version]
- SanMiguel, A.; Grice, E.A. Interactions between host factors and the skin microbiome. Cell Mol. Life Sci. 2014, 72, 1499–1515. [Google Scholar] [CrossRef] [Green Version]
- Williams, M.R.; Gallo, R.L. The Role of the Skin Microbiome in Atopic Dermatitis. Curr. Allergy Asthma Rep. 2015, 15, 65. [Google Scholar] [CrossRef]
- Sanford, J.A.; Zhang, L.-J.; Williams, M.R.; Gangoiti, J.A.; Huang, C.-M.; Gallo, R.L. Inhibition of HDAC8 and HDAC9 by microbial short-chain fatty acids breaks immune tolerance of the epidermis to TLR ligands. Sci. Immunol. 2016, 1, eaah4609. [Google Scholar] [CrossRef] [PubMed]
- Brandwein, M.; Bentwich, Z.; Steinberg, D. Endogenous Antimicrobial Peptide Expression in Response to Bacterial Epidermal Colonization. Front. Immunol. 2017, 8, 1637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.E.; Kim, J.-M.; Jeong, S.K.; Jeon, J.E.; Yoon, H.-J.; Jeong, M.-K.; Lee, S.H. Protease-activated receptor-2 mediates the expression of inflammatory cytokines, antimicrobial peptides, and matrix metalloproteinases in keratinocytes in response to Propionibacterium acnes. Arch. Dermatol. Res. 2010, 302, 745–756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Donnarumma, G.; Perfetto, B.; Paoletti, I.; Oliviero, G.; Clavaud, C.; Del Bufalo, A.; Guéniche, A.; Jourdain, R.; Tufano, M.A.; Breton, L. Analysis of the response of human keratinocytes to Malassezia globosa and restricta strains. Arch. Dermatol. Res. 2014, 306, 763–768. [Google Scholar] [CrossRef]
- Ishibashi, Y.; Sugita, T.; Nishikawa, A. Cytokine secretion profile of human keratinocytes exposed to Malassezia yeasts. FEMS Immunol. Med. Microbiol. 2006, 48, 400–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, J.; Luo, Y.; Zhu, Z.; Zhou, Y.; Sun, L.; Gao, J.; Sun, J.; Wang, G.; Yao, X.; Li, W. A tryptophan metabolite of the skin mi-crobiota attenuates inflammation in patients with atopic dermatitis through the aryl hydrocarbon receptor. J. Allergy Clin. Immunol. 2019, 143, 2108–2119.e12. [Google Scholar] [CrossRef] [Green Version]
- Kjer-Nielsen, L.; Patel, O.; Corbett, A.J.; Le Nours, J.; Meehan, B.; Liu, L.; Bhati, M.; Chen, Z.; Kostenko, L.; Reantragoon, R.; et al. MR1 presents microbial vitamin B metabolites to MAIT cells. Nature 2012, 491, 717–723. [Google Scholar] [CrossRef]
- Sams-Dodd, J.; Sams-Dodd, F. Time to Abandon Antimicrobial Approaches in Wound Healing: A Paradigm Shift. Wounds 2018, 30, 345–352. [Google Scholar]
- Eming, S.A.; Martin, P.; Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Transl. Med. 2014, 6, 265sr6. [Google Scholar] [CrossRef] [Green Version]
- Tomic-Canic, M.; Burgess, J.L.; O’Neill, K.E.; Strbo, N.; Pastar, I. Skin Microbiota and its Interplay with Wound Healing. Am. J. Clin. Dermatol. 2020, 21 (Suppl. 1), 36–43. [Google Scholar] [CrossRef]
- Zeeuwen, P.L.; Boekhorst, J.; van den Bogaard, E.H.; de Koning, H.D.; van de Kerkhof, P.M.; Saulnier, D.M.; van Swam, I.I.; van Hijum, S.A.; Kleerebezem, M.; Schalkwijk, J.; et al. Microbiome dynamics of human epidermis following skin barrier disruption. Genome Biol. 2012, 13, R101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heath, W.R.; Carbone, F.R. The skin-resident and migratory immune system in steady state and memory: Innate lymphocytes, dendritic cells and T cells. Nat. Immunol. 2013, 14, 978–985. [Google Scholar] [CrossRef] [PubMed]
- ISO. ISO 17516:2014 Cosmetics—Microbiology—Microbiological Limits; ISO: Geneva, Switzerland, 2014. [Google Scholar]
Skin Condition | Microbiome Shift | Reference |
---|---|---|
Atopic Dermatitis (AD) | Decreased microbial diversity Increase in S. aureus associated with disease severity and increase in S. epidermidis during flares Decrease in C. acnes, lactobacilli, and Burkholderia spp. Increase in M. sympodialis, (and secondly M. globosa, M. dermatis, M. restricta) | [75,76,77,78,79,80,81] |
Psoriasis | Increase in C. simulans, C. kroppenstedtii, Finegoldia spp., and Neisseriaceae spp. Decrease in C. acnes, lactobacilli, and Burkholderia spp. No difference in the amount of Malassezia between lesion area of psoriasis and healthy skin (measured by PCR) but there is more diversity of Malassezia species in patients with psoriasis compared to healthy individuals Increase in Brevibacterium, Kocuria palustris, Gordonia, and increase in M. restricta (back) and M. sympodialis (elbow) increased in psoriatic lesions. | [76,79,82,83] |
Acne | Increase in the proportion of C. acnes strains presenting with virulence factors Increase in S. epidermidis (secondary to C. acnes) Malassezia spp. could be involved in the development of acne | [46,84,85,86,87,88] |
Rosacea | Increased in Demodex mites on the skin. | [89,90] |
Vitiligo | Decrease in bacterial diversity | [74] |
Seborrheic dermatitis (SD) and Dandruff | Increase in S. epidermidis and decrease in C. acnes. Increase in the population of Malassezia restricta and Malassezia globosa on the scalp. Malassezia spp. metabolize and oxidize sebum-derived lipids into inflammatory compounds and produce indole derivatives (malassezin, indolocarbazole) which may impact skin inflammation through aryl hydrocarbon receptors. | [88,91,92,93,94] |
Postbiotic (Including Probiotic Fraction or Extract) | Prebiotic |
---|---|
Non-viable ingredients comprised of inactivated microorganisms and/or soluble factors (products or metabolic by-products) released by live or inactivated microorganisms, added to a cosmetic product to achieve a cosmetic benefit at the application site, either directly or via an effect on the existing microbiota. Categories: 1/Ferments, lysates, extracts, filtrates, 2/Non-viable microorganisms (inactivated/heat-killed), 3/Metabolic products/by-products (isolated) | Non-viable ingredients are added to a cosmetic product to be actively used as nutrients by the microbiota of the application site to achieve a cosmetic benefit. Examples: ingredients such as fibers, sugars, minerals, but also complex biological mixtures/extracts, etc. |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gueniche, A.; Perin, O.; Bouslimani, A.; Landemaine, L.; Misra, N.; Cupferman, S.; Aguilar, L.; Clavaud, C.; Chopra, T.; Khodr, A. Advances in Microbiome-Derived Solutions and Methodologies Are Founding a New Era in Skin Health and Care. Pathogens 2022, 11, 121. https://doi.org/10.3390/pathogens11020121
Gueniche A, Perin O, Bouslimani A, Landemaine L, Misra N, Cupferman S, Aguilar L, Clavaud C, Chopra T, Khodr A. Advances in Microbiome-Derived Solutions and Methodologies Are Founding a New Era in Skin Health and Care. Pathogens. 2022; 11(2):121. https://doi.org/10.3390/pathogens11020121
Chicago/Turabian StyleGueniche, Audrey, Olivier Perin, Amina Bouslimani, Leslie Landemaine, Namita Misra, Sylvie Cupferman, Luc Aguilar, Cécile Clavaud, Tarun Chopra, and Ahmad Khodr. 2022. "Advances in Microbiome-Derived Solutions and Methodologies Are Founding a New Era in Skin Health and Care" Pathogens 11, no. 2: 121. https://doi.org/10.3390/pathogens11020121
APA StyleGueniche, A., Perin, O., Bouslimani, A., Landemaine, L., Misra, N., Cupferman, S., Aguilar, L., Clavaud, C., Chopra, T., & Khodr, A. (2022). Advances in Microbiome-Derived Solutions and Methodologies Are Founding a New Era in Skin Health and Care. Pathogens, 11(2), 121. https://doi.org/10.3390/pathogens11020121