You are on page 1of 7

Available online at www.sciencedirect.

com

ScienceDirect

Mechanisms for control of skin immune function by the


microbiome
Teruaki Nakatsuji, Joyce Y Cheng and Richard L Gallo

The skin represents the largest area for direct contact between typically provide semi-specific and highly potent defense
microbes and host immunocytes and is a site for constant against a broad range of microbes by disrupting mem-
communication between the host and this diverse and essential branes. Thus, control of the expression and post-tran-
microbial community. Coagulase-negative staphylococci are scriptional activation of these peptides and proteins is a
an abundant bacterial genus on the human skin and are primary innate host defense mechanism for directly sup-
regulated through various mechanisms that include the pressing microbial growth [1,2]. AMP expression
epidermal barrier environment and innate and adaptive immune increases when the skin detects danger signals or inflam-
systems within the epidermis and dermis. In turn, some species matory cues in response to wounding or infection, and this
and strains of these bacteria produce beneficial products that is mediated by both classical and unique signaling path-
augment host immunity by exerting specifically targeted ways that include pattern recognition receptors, cyto-
antimicrobial, anti-inflammatory, or anti-neoplastic activity kines, vitamins, hormones, and neuropeptides [1,3–6].
while also promoting broad innate and adaptive immune Activation of AMP activity is essential for control of
responses. The use of selected skin commensals as a bacterial growth on the skin in case of epithelial barrier
therapeutic has shown promise in recent human clinical trials. damage.
This emerging concept of bacteriotherapy is defining
mechanisms of action and validating the dependence on the Despite intrinsic expression of AMPs by diverse cells in the
microbiome for maintenance of immune homeostasis. skin, an abundant and diverse population of microbial
organisms colonizes the epithelial surfaces of mammals.
Address This community of bacteria, fungi, protozoa, and viruses
Department of Dermatology, University of California, San Diego, United has been called the ‘microbiome’. Emerging evidence over
States
the last 15 years has suggested that the microbiome con-
Corresponding author: Gallo, Richard L (rgallo@health.ucsd.edu) tributes crucially not only to human health and a well-
functioning immune system but also to the pathogenesis of
diverse diseases including atopic dermatitis (AD), inflam-
Current Opinion in Immunology 2021, 72:324–330
matory bowel disease, obesity, and cancer [7,8]. These
This review comes from a themed issue on Allergy and associations have driven the hypothesis that our body is
hypersensitivity
in continuous communication across epithelial barriers
Edited by Toshiaki Kawakami and Ulrich Blank with microbial ecosystems that influence both local and
For a complete overview see the Issue and the Editorial systemic homeostasis within our bodies. The following will
Available online 16th September 2021 review evidence that supports this hypothesis.
https://doi.org/10.1016/j.coi.2021.09.001
It is a common misconception that human skin is a flat
0952-7915/ã 2021 The Authors. Published by Elsevier Ltd. This is an
surface and impermeable barrier for microbes. In reality,
open access article under the CC BY license (http://creativecommons.
org/licenses/by/4.0/). it contains complex anatomical structures, including
grooves, hair follicles, sebaceous glands, and eccrine
sweat glands with specific physiological functions to
maintain a dynamic environmental interface. Follicular
glands provide a rich source of triglycerides and fatty acids
for microbial growth. Indeed, a larger quantity of
microbes reside within hair follicles and eccrine glands
Human skin provides a unique milieu for
than on the epidermal surface of human skin [9]. When
host–microbe interactions
the various anatomical structures of the skin are taken
Mammalian skin possesses intrinsic defenses against
together, the total surface area of this interface is vast,
infection by microbial pathogens. Both skin resident cells
comparable to or larger than that of the intestine, but
and commensal bacteria participate in a variety of protec-
without a thick mucous layer separating microbes from
tive innate and adaptive immune mechanisms. Antimi-
the epithelial surface [10]. This physical structure and
crobial peptides (AMPs) form an important part of this
concentration of microbes within protected follicles
defense. They are produced by various cells in the
implicates the skin as a major site for potential host–
epidermis and dermis, including keratinocytes, sebo-
microbe interactions. In particular, follicular epithelial
cytes, and immunocytes such as neutrophils, mast cells,
surfaces represent an important interface for immune
monocytes, T-cells, and dermal adipocytes [1]. AMPs

Current Opinion in Immunology 2021, 72:324–330 www.sciencedirect.com


The skin microbiome as an element of immune system Nakatsuji, Cheng and Gallo 325

system interaction with microbes. Sebaceous and eccrine skin immune system (Figure 1). For example, S. epider-
glands also actively regulate the growth of resident bac- midis produces a uniquely structured lipoteichoic acid
teria through production of AMPs and antimicrobial lipids that blocks overactivation of pro-inflammatory signaling
to prevent unwanted bacterial overgrowth [11,12]. These triggered by damage-associated molecular pattern recog-
facts suggest that mammalian skin is uniquely suited to nition after tissue damage [20,21]. In this relationship,
harbor, control, and protect a microbial ecosystem. the host benefits from controlled wound repair, while the
commensals are protected from excessive acute inflam-
Elements of the microbial community residing on the mation. A lipopeptide produced by S. epidermidis defends
human skin surface routinely penetrate the epidermal the host from pathogen infection by enhancing AMP
barrier and enter dermal compartments [9], suggesting production in human keratinocytes and mast cells, boost-
that bacteria in the dermis directly interact with various ing the innate immune system [22–24].
dermal cell types and influence their phenotypes. Skin
penetration by bacteria is dependent upon proteolytic Colonization with S. epidermidis has been shown to
mechanisms and is independent from translocation by enhance CD8+ skin-resident T cell functions via activa-
CD11c+ phagocytic immune cells [13]. Indeed, the com- tion of IL-1 signaling [25]. This in turn promotes upre-
position and richness of the microbial community in the gulation of AMP production in keratinocytes, limiting
dermal compartment differ from those in the superficial pathogen invasion [26]. S. epidermidis-specific CD8+ T-
epidermal layer [14], indicating that only selected bacte- cells express genes associated with immunoregulatory
ria can translocate across the epithelial barrier. Bacterial and tissue repair signatures [27]. Furthermore, it has been
penetration is highly regulated by epidermal barrier ele- demonstrated in mice that early life exposure to ribofla-
ments, including the AMP cathelicidin and filaggrin vin-synthesizing commensals, including S. epidermidis,
barrier protein [13]. Skin-resident innate lymphoid cells promotes development of skin-residing mucosal-associ-
(ILCs) isolated from the epidermis, dermis, or subcutis ated invariant T cells in adult life [28]. These events in
demonstrate layer-specific transcriptomic profiles [15], development of specific types of T-cells are integral to
indicating they receive unique signals from the microbial controlling tissue repair and homeostasis.
community in different anatomical layers of the skin.
Therefore, the epithelial barrier regulates host–microbe S. epidermidis also facilitates wound healing independent
interactions not only by selecting for bacterial survival but of T-cell-mediated mechanisms through the activation of
also by controlling the movement of commensal bacteria. neutrophils to express the chemokine CXCL10 [29].
CXCL10 forms a complex with DNA from commensals,
The microbiome is a master manipulator of which activates type I interferon-producing plasmacytoid
cutaneous immunity dendritic cells and accelerates wound closure. Further-
The mechanisms by which certain microbes evade detec- more, selected strains of S. epidermidis produce 6-N-
tion by the cutaneous immune system during skin colo- hydroxyaminopurine, a unique nucleobase analog, with
nization are of great interest. Regulatory T cells are the capacity to inhibit DNA replication in cancer cells.
essential for establishing and maintaining immune Topical application of the S. epidermidis strain protects
homeostasis in epithelial tissues. Skin colonization by mouse skin from neoplasia induced by mutagen sensiti-
Staphylococcus epidermidis during the neonatal period zation and ultraviolet B exposure [30].
may establish immune tolerance through accumulation
of commensal-specific regulatory T cells after second Given the body of evidence suggesting that the commu-
exposure to the same species in later life [16]. In contrast, nity of symbiotic bacteria influences and is necessary to
exposure to a-toxin produced by S. aureus prevents this maintain health, the hologenome theory was introduced
immune tolerance to commensals and activates the alar- to better explain host–microbiome homeostasis [31,32].
min pathway instead, resulting in breakdown of inflam- In this theory, a holobiont (a host and all of its associated
matory homeostasis and enhancing immune response to symbiotic microbes) should be considered as a single unit
commensals [17]. Moreover, short-chain free fatty acids in the process of evolution, even though the genomes of
(SCFAs), fermentation products of sebum by skin com- host and symbionts have separate origins.
mensal bacteria Corynebacterium acnes, can also actively
control immune tolerance to commensals through inhibi- The microbiome provides additional layers of
tion of histone deacetylase activity [18,19]. Thus, com- antimicrobial barrier on human skin
mensals likely influence immune tolerance through mul- In general, bacterial populations inhabiting the same
tiple mechanisms to maintain inflammatory homeostasis. ecological niche compete with each other for nutrients
and other resources using strategies including production
Although many microbes on the skin are potential oppor- of antibiotics, secretion of digestive enzymes, and quo-
tunitic pathogens or do not influence the host immune rum sensing. Recent studies have demonstrated that this
system, some selected species or strains of bacteria appear ecological concept also applies within human-associated
to have a mutualistic and beneficial relationship with the microbial communities such as the gut and skin

www.sciencedirect.com Current Opinion in Immunology 2021, 72:324–330


326 Allergy and hypersensitivity

Figure 1

Current Opinion in Immunology

Beneficial actions of coagulase-negative staphylococci in the skin microbiome.


Gram positive bacteria are the dominant organisms that colonize human skin and Coagulase negative Staphylococci are the major class of these
microbes that can establish themselves in the commensal skin community. Of these diverse species, some have a mutualistic beneficial
interactions with the immune system through a variety of strain-specific mechanisms. 6-HAP, 6-N-hydroxyaminopurine; LTA, lipoteichoic acid;
DAMP, damage-associated molecular pattern; CoNS, coagulase-negative staphylococcus; AMP, antimicrobial peptide; MAIT cells, mucosal-
associated invariant T cells; pDC, plasmacytoid dendritic cell.

microbiome [33,34]. In cooperation with the host immune the epithelial barrier and promote inflammation [39]. The
system, these antagonistic interactions determine the presence of S. aureus strains with functional agr quorum
composition of a functional antimicrobial barrier. sensing systems is associated with AD disease; in contrast,
S. aureus strains with loss-of-function mutations in the agr
Coagulase-negative staphylococcus (CoNS) is an abun- gene are more prevalent in non-AD subjects [40], impli-
dant culturable member of the human skin microbiome cating S. aureus quorum sensing in the pathogenesis of
[35]. Bacteriocins, a class of prokaryotic antimicrobials AD. Various CoNS strains within the human skin micro-
produced by some strains of CoNS, serve as an important biome produce a unique cyclic peptide known as the
antimicrobial barrier on the skin surface that comple- autoinducing peptide (AIP), which blocks quorum sens-
ments the expression of AMPs by the host. For example, ing system in a wide range of S. aureus strains [41].
lantibiotics and phenol-soluble modulins (PSMs) pro- Recent observations suggest that certain strains of S.
duced by CoNS limit survival of pathogenic bacteria epidermidis abundantly produce cysteine proteases under
on the skin surface [34,36,37]. Importantly, these pro- the control of quorum sensing, which exacerbates skin
karyotic AMPs are found abundantly on normal skin and inflammation in AD, while AIP produced by a S. hominis
can synergize with host AMPs including cathelicidin and strain can inhibit quorum sensing by S. epidermidis [42]. S.
b-defensin-2 against pathogenic bacteria, which further aureus and S. epidermidis protease production under con-
enhances antimicrobial defense [34,36–38]. Prokaryotic trol of the agr system have also been recently shown to
AMPs selectively exhibit bactericidal activity against skin closely associate with exacerbations of disease in the
pathogens, such as S. aureus (S. aureus), Group A Strepto- monogenetic human disorder Netherton’s syndrome,
coccus, and Escherichia coli, but are not typically active and this too can be inhibited by AIPs from CoNS resident
against commensal organisms such as S. epidermidis. This on human skin [43]. Thus, interspecies antagonism by
selectivity may be important to maintaining healthy and AIPs produced by beneficial commensal strains can
diverse microbial ecosystems. reduce the detrimental effects of pathogenic bacteria.

Quorum sensing is a form of communication that allows Targeting dysbiosis in atopic dermatitis offers
bacterial cells to share information about population novel therapeutic approaches
density and regulate gene expression accordingly. S. Because the cutaneous immune system actively regulates
aureus relies on quorum sensing to secrete selective toxins the microbial community, dysregulation of AMP expres-
and proteases such as PSMs and d-hemolysin that damage sion on the skin can lead to an imbalance in the microbial

Current Opinion in Immunology 2021, 72:324–330 www.sciencedirect.com


The skin microbiome as an element of immune system Nakatsuji, Cheng and Gallo 327

community known as ‘dysbiosis’. In many cases, dysbiosis capitis, which produce bacteriocins that selectively kill S.
can trigger the development of skin disorders through the aureus, are much rarer on AD skin than on healthy skin
detrimental effects of overgrown pathogens as well as [34]. A lower frequency of commensal CoNS strains on
commensals. In addition, dysbiosis disrupts symbiotic the skin has been observed to correlate with S. aureus
interactions with beneficial bacteria within the micro- colonization and contributes to the overgrowth of detri-
biome, disequilibrating cutaneous homeostasis. Com- mental pathogens [34].
bined, the abnormal skin immune environment of inflam-
matory skin diseases promotes dysbiosis that is Normalization of the skin microbiome offers a promising
manifested as both an increase in pathogenic microbes therapeutic avenue for restoring homeostasis in the man-
and a decrease in beneficial organisms. This dysbiosis in agement of inflammatory skin diseases such as AD. One
turn drives further exacerbation of the host inflammatory approach to this has been by targeting defective host
response with resulting establishment of chronic disease immune systems that cause the skin dysbiosis. Because of
(Figure 2). the action of 1,25-hydroxyvitamin D to induce the human
AMP gene CAMP, oral vitamin D supplementation has
In patients with AD, expression of AMPs such as cathe- been evaluated as a method to increase skin cathelicidin
licidin and b-defensins on the skin is decreased by over- production and improve disease severity in AD patients
expression of Th2 cytokines, including IL-4, IL-13 and [47,48], though further studies are needed to determine
TSLP, in comparison with other inflammatory conditions, whether this increase functionally protects against skin
such as psoriasis and cutaneous wounds [36,37]. Although infections. Cross-sectional observational studies have
AMP production in AD skin is greater than that in non- noted a correlation between serum 25-hydroxyvitamin
inflamed healthy skin, it is not enough to suppress growth D deficiency, increased skin colonization by S. aureus, and
of S. aureus. With its relative paucity of AMPs, AD skin is increased disease severity in AD patients [49,50]. Another
more susceptible to colonization and infection by patho- approach has been to consider an old remedy for AD and
gens like S. aureus and herpes virus [44,45]. In addition, other skin diseases, topical coal tar. Activation of the aryl
AMP dysregulation in AD weakens selective pressure on hydrocarbon receptor by coal tar is postulated to induce
skin microbiota, altering the microbial composition of AD expression of keratinocyte-derived AMPs. Topical coal
skin compared with normal skin [46]. Strains of commen- tar treatment has been shown to decrease staphylococcus
sal CoNS such as S. epidermidis, S. hominis, S. warneri, or S. abundance in AD patients compared with vehicle and

Figure 2

Current Opinion in Immunology

Dysbiosis fuels the chronic disease cycle.


Barrier dysfunction on the skin surface may promote dysbiosis with overgrowth of pathogenic bacterial species and/or a decrease in beneficial
commensal strains. The dysbiosis in the skin microbiome can expand defects in the physical barrier and stimulate inflammation that then further
promotes inflammation and additional injury (red arrows). Bacteriotherapy using rationally designed or selected beneficial commensals could
directly and indirectly restore beneficial interactions between host and microbes (green lines). Solid green lines represent relationships supported
by human clinical studies and broken lines represent relationships validated only in preclinical investigations. Such bacteriotherapy could be
expected to be useful for a wide range of disease states associated with dysbiosis.

www.sciencedirect.com Current Opinion in Immunology 2021, 72:324–330


328 Allergy and hypersensitivity

shifted the microbial composition toward that of healthy tumors (Figure 1). Translation of these additional find-
controls [51]. Modern, targeted biologic therapy has also ings to treat a wide range of human skin disorders is
shown promise. Excess production of Th2 cytokines is a currently underway.
key element in the pathophysiology of AD and sup-
presses host AMP functions to control S. aureus survival. Conclusion
Dupilumab, a fully human antibody against IL-4 receptor The preponderance of current experimental and clinical
a, inhibits both IL-4 and IL-13 signaling and should studies have shown that selected species or strains within
theoretically enable more normal AMP function. This the skin microbial community can work together with the
hypothesis was supported in a randomized, double-blind, cutaneous immune system to maintain local and systemic
placebo-controlled trial, that showed sixteen weeks of defense against pathogenic bacteria. The skin micro-
treatment with dupilumab increased microbial diversity biome is therefore correctly considered an important
and decreased S. aureus abundance [52]. component of the human holobiont system. Dysbiosis
in the colonized microbial community can promote
Future perspectives on development of inflammatory disease that further disrupts immune
bacteriotherapies using beneficial homeostasis and may lead to the development of skin
commensals disorders. Pharmaceutical antibiotics are widely used to
The relative deficiency of CoNS with antimicrobial activ- temporarily suppress overgrowth of pathogenic bacteria,
ity on AD skin, combined with their multiple antagonistic but the non-selective action of these antibiotics also kills
defense mechanisms against pathogenic S. aureus, present beneficial microbes and should be reserved for infections,
novel therapeutic opportunities for the use of protective an invasive stage beyond bacterial colonization. More
CoNS in the management of S. aureus colonization in AD selected, rational, and targeted use of bacteriotherapy
(Figure 2). As discussed previously, multiple beneficial for the control and regulation of immune functions is a
functions from the skin commensal microbial community promising next step in human therapeutics.
could be used therapeutically. Recently, our group con-
ducted the first multi-center, double-blind, vehicle-con- Conflict of interest statement
trolled, phase 1 clinical trial of bacteriotherapy in adult R.L.G. is a co-founder, scientific advisor, consultant and
patients with AD using a rationally selected strain of S. has equity in MatriSys Biosciences and is a consultant,
hominis (ShA9) that was isolated from a healthy individual receives income and has equity in Sente. T.N. and R.L.G
[53]. This strain is present in healthy populations, but are co-inventors of technology described in this report
not on individuals with AD [34]. ShA9 was chosen that has been disclosed to the University of California San
because it produces two lantibiotics and an AIP that Diego.
selectively target S. aureus and deleterious strains of S.
epidermidis. Bacteriotherapy with ShA9 did not cause
Acknowledgements
serious adverse events, successfully inhibited S. aureus RLG is supported by N.I.H. grantsR37AI052453,R01AR074302,
survival, and blocked quorum sensing by S. aureus on the andR01AI153185. TN and RLG are supported by NIH grants
lesional skin of AD patients. These multiple protective R01AR076082 andU01AI52038.
effects were associated with improvement in clinical AD
symptoms [53]. In other work, an open-label bacter- References and recommended reading
iotherapy trial of Roseomonas mucosa, a Gram-negative Papers of particular interest, published within the period of review,
have been highlighted as:
bacteria with anti-S. aureus activity, decreased S. aureus
colonization, and improved severity of AD compared to  of outstanding interest
baseline, though vehicle effect was not determined
1. Zhang LJ, Gallo RL: Antimicrobial peptides. Curr Biol 2016, 26:
[54,55]. In addition, topical application of a lysate of R14-R19.
Vitreoscilla filiformis has shown therapeutic efficacy in 2. Schroeder BO, Wu Z, Nuding S, Groscurth S, Marcinowski M,
treating AD in humans [56]. The therapeutic mechanism Beisner J, Buchner J, Schaller M, Stange EF, Wehkamp J:
may be through the induction of regulatory T-cells [57], Reduction of disulphide bonds unmasks potent antimicrobial
activity of human beta-defensin 1. Nature 2011, 469:419-423.
though functions of this cell type in AD are still contro-
versial. Overall, large-scale, longer-term, blinded, and 3. Zhang LJ, Sen GL, Ward NL, Johnston A, Chun K, Chen Y,
Adase C, Sanford JA, Gao N, Chensee M et al.: Antimicrobial
randomized trials are needed to fully understand the peptide LL37 and MAVS signaling drive interferon-beta
therapeutic potential of bacteriotherapy with beneficial production by epidermal keratinocytes during skin injury.
Immunity 2016, 45:119-130.
commensal microbe strains.
4. Dokoshi T, Zhang LJ, Li F, Nakatsuji T, Butcher A, Yoshida H,
Shimoda M, Okada Y, Gallo RL: Hyaluronan degradation by
In pre-clinical trials using various murine models, it has cemip regulates host defense against Staphylococcus aureus
been demonstrated that the topical application of S. skin infection. Cell Rep 2020, 30:61-68 e64.
epidermidis results a wide variety of beneficial actions to 5. Svensson D, Nebel D, Nilsson BO: Vitamin D3 modulates the
innate immune response through regulation of the hCAP-18/
the host, including anti-inflammatory activity, promotion LL-37 gene expression and cytokine production. Inflamm Res
of wound healing, and suppression of UV-B-induced skin 2016, 65:25-32.

Current Opinion in Immunology 2021, 72:324–330 www.sciencedirect.com


The skin microbiome as an element of immune system Nakatsuji, Cheng and Gallo 329

6. Aberg KM, Radek KA, Choi EH, Kim DK, Demerjian M, Hupe M, 22. Lai Y, Cogen AL, Radek KA, Park HJ, Macleod DT, Leichtle A,
Kerbleski J, Gallo RL, Ganz T, Mauro T et al.: Psychological Ryan AF, Di Nardo A, Gallo RL: Activation of TLR2 by a small
stress downregulates epidermal antimicrobial peptide molecule produced by Staphylococcus epidermidis increases
expression and increases severity of cutaneous infections in antimicrobial defense against bacterial skin infections. J Invest
mice. J Clin Invest 2007, 117:3339-3349. Dermatol 2010, 130:2211-2221.
7. Nakatsuji T, Gallo RL: The role of the skin microbiome in atopic 23. Li D, Lei H, Li Z, Li H, Wang Y, Lai Y: A novel lipopeptide from skin
dermatitis. Ann Allergy Asthma Immunol 2019, 122:263-269. commensal activates TLR2/CD36-p38 MAPK signaling to
increase antibacterial defense against bacterial infection.
8. Cani PD, Jordan BF: Gut microbiota-mediated inflammation in PLoS One 2013, 8:e58288.
obesity: a link with gastrointestinal cancer. Nat Rev
Gastroenterol Hepatol 2018, 15:671-682. 24. Igawa S, Di Nardo A: Skin microbiome and mast cells. Transl Res
2017, 184:68-76.
9. Nakatsuji T, Chiang HI, Jiang SB, Nagarajan H, Zengler K,
 Gallo RL: The microbiome extends to subepidermal 25. Naik S, Bouladoux N, Wilhelm C, Molloy MJ, Salcedo R,
compartments of normal skin. Nat Commun 2013, 4:1431 Kastenmuller W, Deming C, Quinones M, Koo L, Conlan S et al.:
Demonstration that the microbiome of the skin surface penetrates into Compartmentalized control of skin immunity by resident
follicles and below the basement membrane. commensals. Science 2012, 337:1115-1119.
10. Gallo RL: Human skin is the largest epithelial surface for 26. Naik S, Bouladoux N, Linehan JL, Han SJ, Harrison OJ, Wilhelm C,
interaction with microbes. J Invest Dermatol 2017, 137:1213- Conlan S, Himmelfarb S, Byrd AL, Deming C et al.: Commensal-
1214. dendritic-cell interaction specifies a unique protective skin
immune signature. Nature 2015, 520:104-108.
11. Nakatsuji T, Kao MC, Zhang L, Zouboulis CC, Gallo RL, Huang CM:
Sebum free fatty acids enhance the innate immune defense of 27. Linehan JL, Harrison OJ, Han SJ, Byrd AL, Vujkovic-Cvijin I,
human sebocytes by upregulating beta-defensin-2 Villarino AV, Sen SK, Shaik J, Smelkinson M, Tamoutounour S
expression. J Invest Dermatol 2010, 130:985-994. et al.: Non-classical immunity controls microbiota impact on
12. Fischer CL, Blanchette DR, Brogden KA, Dawson DV, Drake DR, skin immunity and tissue repair. Cell 2018, 172:784-796 e718.
Hill JR, Wertz PW: The roles of cutaneous lipids in host defense. 28. Constantinides MG, Link VM, Tamoutounour S, Wong AC, Perez-
Biochim Biophys Acta 2014, 1841:319-322.  Chaparro PJ, Han SJ, Chen YE, Li K, Farhat S, Weckel A et al.:
13. Nakatsuji T, Chen TH, Two AM, Chun KA, Narala S, Geha RS, MAIT cells are imprinted by the microbiota in early life and
Hata TR, Gallo RL: Staphylococcus aureus exploits epidermal promote tissue repair. Science 2019, 366
barrier defects in atopic dermatitis to trigger cytokine The authors demonstrat that exposure to riboflavin-synthesizing com-
expression. J Invest Dermatol 2016, 136:2192-2200. mensal microbes in early life are required for development of functional
mucosal-associated invariant T cells. This paper is presents another
14. Bay L, Barnes CJ, Fritz BG, Thorsen J, Restrup MEM, mechanism by which commensals can direct immune cell development.
Rasmussen L, Sorensen JK, Hesselvig AB, Odgaard A, Hansen AJ
et al.: Universal dermal microbiome in human skin. mBio 2020, 29. Di Domizio J, Belkhodja C, Chenuet P, Fries A, Murray T,
11. Mondejar PM, Demaria O, Conrad C, Homey B, Werner S et al.:
The commensal skin microbiota triggers type I IFN-dependent
15. Kobayashi T, Voisin B, Kim DY, Kennedy EA, Jo JH, Shih HY, innate repair responses in injured skin. Nat Immunol 2020,
Truong A, Doebel T, Sakamoto K, Cui CY et al.: Homeostatic 21:1034-1045.
control of sebaceous glands by innate lymphoid cells
regulates commensal bacteria equilibrium. Cell 2019, 176:982- 30. Nakatsuji T, Chen TH, Butcher AM, Trzoss LL, Nam SJ,
997 e916. Shirakawa KT, Zhou W, Oh J, Otto M, Fenical W et al.: A
commensal strain of Staphylococcus epidermidis protects
16. Scharschmidt TC, Vasquez KS, Pauli ML, Leitner EG, Chu K, against skin neoplasia. Sci Adv 2018, 4:eaao4502.
Truong HA, Lowe MM, Sanchez Rodriguez R, Ali N, Laszik ZG
et al.: Commensal microbes and hair follicle morphogenesis 31. Rosenberg E, Zilber-Rosenberg I: The hologenome concept of
coordinately drive treg migration into neonatal skin. Cell Host evolution after 10 years. Microbiome 2018, 6:78.
Microbe 2017, 21:467-477 e465.
32. Bordenstein SR, Theis KR: Host biology in light of the
17. Leech JM, Dhariwala MO, Lowe MM, Chu K, Merana GR, microbiome: ten principles of holobionts and hologenomes.
 Cornuot C, Weckel A, Ma JM, Leitner EG, Gonzalez JR et al.: PLoS Biol 2015, 13:e1002226.
Toxin-triggered interleukin-1 receptor signaling enables
early-life discrimination of pathogenic versus commensal skin 33. Garcia-Gutierrez E, Mayer MJ, Cotter PD, Narbad A: Gut
bacteria. Cell Host Microbe 2019, 26:795-809 e795 microbiota as a source of novel antimicrobials. Gut Microbes
The authors demonstrate that neonatal colonization by Staphylococcus 2019, 10:1-21.
epidermidis promotes immune tollerance to commensals through accu-
mulation of commensal-specific regulatory T cells. The study describes a 34. Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, Shafiq F,
mechanism by which skin discriminates between commensal and patho- Kotol PF, Bouslimani A, Melnik AV et al.: Antimicrobials from
genic microbes. human skin commensal bacteria protect against
Staphylococcus aureus and are deficient in atopic dermatitis.
18. Sanford JA, Zhang LJ, Williams MR, Gangoiti JA, Huang CM, Sci Transl Med 2017, 9.
Gallo RL: Inhibition of HDAC8 and HDAC9 by microbial short-
chain fatty acids breaks immune tolerance of the epidermis to 35. Roth RR, James WD: Microbial ecology of the skin. Annu Rev
TLR ligands. Sci Immunol 2016, 1. Microbiol 1988, 42:441-464.

19. Sanford JA, O’Neill AM, Zouboulis CC, Gallo RL: Short-chain fatty 36. Cogen AL, Yamasaki K, Sanchez KM, Dorschner RA, Lai Y,
acids from Cutibacterium acnes activate both a canonical and MacLeod DT, Torpey JW, Otto M, Nizet V, Kim JE et al.: Selective
epigenetic inflammatory response in human sebocytes. J antimicrobial action is provided by phenol-soluble modulins
Immunol 2019, 202:1767-1776. derived from Staphylococcus epidermidis, a normal resident
of the skin. J Invest Dermatol 2010, 130:192-200.
20. Lai Y, Di Nardo A, Nakatsuji T, Leichtle A, Yang Y, Cogen AL,
 Wu ZR, Hooper LV, Schmidt RR, von Aulock S et al.: Commensal 37. Cogen AL, Yamasaki K, Muto J, Sanchez KM, Crotty Alexander L,
bacteria regulate toll-like receptor 3-dependent inflammation Tanios J, Lai Y, Kim JE, Nizet V, Gallo RL: Staphylococcus
after skin injury. Nat Med 2009, 15:1377-1382 epidermidis antimicrobial delta-toxin (phenol-soluble
First detailed molecular description of a mechanism by which the micro- modulin-gamma) cooperates with host antimicrobial peptides
biome can exert anti-inflammatory effects. to kill group A Streptococcus. PLoS One 2010, 5:e8557.
21. Bernard JJ, Cowing-Zitron C, Nakatsuji T, Muehleisen B, Muto J, 38. O’Neill AM, Nakatsuji T, Hayachi A, Williams MR, Mills RH,
Borkowski AW, Martinez L, Greidinger EL, Yu BD, Gallo RL: Gonzalez DJ, Gallo RL: Identification of a human skin
Ultraviolet radiation damages self noncoding RNA and is commensal bacterium that selectively kills Cutibacterium
detected by TLR3. Nat Med 2012, 18:1286-1290. acnes. J Invest Dermatol 2020, 140:1619-1628 e1612.

www.sciencedirect.com Current Opinion in Immunology 2021, 72:324–330


330 Allergy and hypersensitivity

39. Schilcher K, Horswill AR: Staphylococcal biofilm development: 49. Gilaberte Y, Sanmartin R, Aspiroz C, Hernandez-Martin A,
structure, regulation, and treatment strategies. Microbiol Mol Benito D, Sanz-Puertolas P, Alonso M, Torrelo A, Torres C:
Biol Rev 2020, 84. Correlation between serum 25-hydroxyvitamin D and
virulence genes of Staphylococcus aureus isolates colonizing
40. Nakamura Y, Takahashi H, Takaya A, Inoue Y, Katayama Y, children with atopic dermatitis. Pediatr Dermatol 2015, 32:506-
Kusuya Y, Shoji T, Takada S, Nakagawa S, Oguma R et al.: 513.
Staphylococcus agr virulence is critical for epidermal
colonization and associates with atopic dermatitis 50. Ibrahim HM, El-Taieb MA, Hassan MH, Mohamed AAE, Kotop EA,
development. Sci Transl Med 2020, 12. Abd-Ellah OH, Hegazy EM: Relations between vitamin D3, total
and specific IgE for house dust mites in atopic dermatitis
41. Williams MR, Costa SK, Zaramela LS, Khalil S, Todd DA, patients. Sci Rep 2020, 10:21000.
 Winter HL, Sanford JA, O’Neill AM, Liggins MC, Nakatsuji T et al.:
Quorum sensing between bacterial species on the skin 51. Smits JPH, Ederveen THA, Rikken G, van den Brink NJM, van
protects against epidermal injury in atopic dermatitis. Sci Vlijmen-Willems I, Boekhorst J, Kamsteeg M, Schalkwijk J, van
Transl Med 2019, 11 Hijum S, Zeeuwen P et al.: Targeting the cutaneous microbiota
The authors show an important role for quorum-sensing in Staphylococ- in atopic dermatitis by coal tar via AHR-dependent induction
cus aureus in the pathogenesis of skin disease This study helps to of antimicrobial peptides. J Invest Dermatol 2020, 140:415-424
understand how communication between bacteria helps to maintain e410.
the epithelial barrier.
52. Callewaert C, Nakatsuji T, Knight R, Kosciolek T, Vrbanac A,
42. Cau L, Williams MR, Butcher AM, Nakatsuji T, Kavanaugh JS, Kotol P, Ardeleanu M, Hultsch T, Guttman-Yassky E,
Cheng JY, Shafiq F, Higbee K, Hata TR, Horswill AR et al.: Bissonnette R et al.: IL-4Ralpha blockade by dupilumab
Staphylococcus epidermidis protease EcpA can be a decreases Staphylococcus aureus colonization and increases
deleterious component of the skin microbiome in atopic microbial diversity in atopic dermatitis. J Invest Dermatol 2020,
dermatitis. J Allergy Clin Immunol 2021, 147:955-966.e16. 140:191-202 e197.
53. Nakatsuji T, Hata T, Tong Y, Cheng JY, Shafiq F, Butcher AM,
43. Williams MR, Cau L, Wang Y, Kaul D, Sanford JA, Zaramela LS,
 Salem SS, Brinton SL, Rudman-Spergel AK, Johnson K et al.:
Khalil S, Butcher AM, Zengler K, Horswill AR et al.: Interplay of
Development of a human skin commensal microbe for
Staphylococcal and host proteases promotes skin barrier
bacteriotherapy of atopic dermatitis and use in a phase
disruption in netherton syndrome. Cell Rep 2020, 30:2923-2933
1 randomized clinical trial. Nat Med 2021, 27:700-709 Published
e2927.
on 22 Februaly 2021
44. Hata TR, Kotol P, Boguniewicz M, Taylor P, Paik A, Jackson M, The authors address the mechanisms and clinical potential of a human
Nguyen M, Kabigting F, Miller J, Gerber M et al.: History of skin commensal strain of coagulase-negative staphylococcus that pro-
eczema herpeticum is associated with the inability to induce duces lantibiotics and inhibits quorum sensing of S. aureus on mice and
human beta-defensin (HBD)-2, HBD-3 and cathelicidin in the humans. This is the first double-blinded, placebo-controlled, randomized
skin of patients with atopic dermatitis. Br J Dermatol 2010, and multi-institutional human clinical study of bacteriotherapy for a skin
163:659-661. disorder.
54. Myles IA, Earland NJ, Anderson ED, Moore IN, Kieh MD,
45. Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T,
Williams KW, Saleem A, Fontecilla NM, Welch PA, Darnell DA et al.:
Gallo RL, Leung DY: Endogenous antimicrobial peptides and
First-in-human topical microbiome transplantation with
skin infections in atopic dermatitis. N Engl J Med 2002,
Roseomonas mucosa for atopic dermatitis. JCI Insight 2018, 3.
347:1151-1160.
55. Myles IA, Castillo CR, Barbian KD, Kanakabandi K, Virtaneva K,
46. Fyhrquist N, Muirhead G, Prast-Nielsen S, Jeanmougin M, Olah P, Fitzmeyer E, Paneru M, Otaizo-Carrasquero F, Myers TG,
Skoog T, Jules-Clement G, Feld M, Barrientos-Somarribas M, Markowitz TE et al.: Therapeutic responses to Roseomonas
Sinkko H et al.: Microbe-host interplay in atopic dermatitis and mucosa in atopic dermatitis may involve lipid-mediated TNF-
psoriasis. Nat Commun 2019, 10:4703. related epithelial repair. Sci Transl Med 2020, 12.
47. Hata TR, Kotol P, Jackson M, Nguyen M, Paik A, Udall D, 56. Gueniche A, Hennino A, Goujon C, Dahel K, Bastien P, Martin R,
Kanada K, Yamasaki K, Alexandrescu D, Gallo RL: Administration Jourdain R, Breton L: Improvement of atopic dermatitis skin
of oral vitamin D induces cathelicidin production in atopic symptoms by Vitreoscilla filiformis bacterial extract. Eur J
individuals. J Allergy Clin Immunol 2008, 122:829-831. Dermatol 2006, 16:380-384.
48. Albenali LH, Danby S, Moustafa M, Brown K, Chittock J, 57. Volz T, Skabytska Y, Guenova E, Chen KM, Frick JS,
Shackley F, Cork MJ: Vitamin D and antimicrobial peptide levels Kirschning CJ, Kaesler S, Rocken M, Biedermann T:
in patients with atopic dermatitis and atopic dermatitis Nonpathogenic bacteria alleviating atopic dermatitis
complicated by eczema herpeticum: a pilot study. J Allergy Clin inflammation induce IL-10-producing dendritic cells and
Immunol 2016, 138:1715-1719 e1714. regulatory Tr1 cells. J Invest Dermatol 2014, 134:96-104.

Current Opinion in Immunology 2021, 72:324–330 www.sciencedirect.com

You might also like