You are on page 1of 7

T H E S YS T E M IC MI C R O B I O M E

REVIEW appendage that contains the hair follicle and its


associated sebaceous gland. Sebaceous glands
Microbiota and maintenance of skin barrier function excrete a waxy, oily substance called sebum
that emolliates the skin and selects for micro-
Tamia A. Harris-Tryon1 and Elizabeth A. Grice2 bial species that metabolize the nutrients it
contains. The pilosebaceous unit is also hypoxic,
Human skin forms a protective barrier against the external environment and is our first line of defense providing an ideal environment for the facul-
against toxic, solar, and pathogenic insults. Our skin also defines our outward appearance, protects tative anaerobe Cutibacterium acnes, which
our internal tissues and organs, acts as a sensory interface, and prevents dehydration. Crucial to the dominates this niche. Other signature bacte-
skin’s barrier function is the colonizing microbiota, which provides protection against pathogens, ria of the skin surface include the coagulase-
tunes immune responses, and fortifies the epithelium. Here we highlight recent advances in our negative staphylococci (CoNS) species, such as
understanding of how the microbiota mediates multiple facets of skin barrier function. We discuss recent Staphylococcus epidermidis, which are equipped
insights into pathological host–microbiota interactions and implications for disorders of the skin and to adhere and persist on human skin and tol-
distant organs. Finally, we examine how microbiota-based mechanisms can be targeted to prevent erate the conditions. Skin sites with higher
or manage skin disorders and impaired wound healing. moisture and occlusion (e.g., the groin, axilla,
and umbilicus) are enriched by Corynebacterium

H
species that can be lipid-dependent and slow-
umans live in partnership with their mi- although generally in lower numbers. The skin growing in culture (Fig. 1).
crobiota, complex communities of bacte- also houses eukaryotic organisms, but these are The skin microbiota experiences a major
ria, fungi, and viruses that inhabit the less abundant than bacteria. Malassezia species, shift at puberty when sex hormones drive mat-
body’s surfaces. These relationships have for example, predominate among fungal com- uration of the sebaceous gland and initiate se-
been forged and challenged over millions munities throughout adult human skin (Fig. 1), bum production. The introduction of lipid-rich

Downloaded from https://www.science.org on December 13, 2023


of years of coevolution. Thus, it is unsurprising and the eight-legged arachnid genus Demodex sebum drives expansion of lipophilic C. acnes
that our microbial cohabitants are major partic- resides within the hair follicle. Initial coloniza- and Malassezia spp. on the skin surface, which
ipants in shaping and maintaining essential tion of neonatal skin is suspected to occur during correlates with serum sex hormone concen-
physiological processes. As our outermost barrier delivery. In infants born vaginally, Lactobacillus, trations (5). Outside of puberty, the strains of
against the external environment, the skin is col- Prevotella, or Sneathia species are transferred to bacteria and fungi colonizing the skin remain
onized by a distinctive commensal microbiota that the skin during passage through the cervix and relatively stable within an individual over time
stimulates and educates defense and immune vagina. These species disappear by 6 weeks of (2). This stability is remarkable given the con-
responses, contributes to proper differentiation age, when the microbiota begins to develop a more tinuous disturbances imposed by lifestyle, the
and epithelialization, and even provides direct skin-like profile enriched with Staphylococcus environment, and other host-specific factors,
defense against pathogenic microorganisms. and Corynebacterium species. In infants born by including constant shedding of terminally dif-
In this Review, we examine our current cesarian section, Staphylococcus, Corynebacterium, ferentiated keratinocytes (squames) and secre-
understanding of the skin’s microbial ecology and Cutibacterium species predominate, with- tions of sweat and sebum (Fig. 1). C. acnes and
and highlight recent insights into the micro- out a preceding vaginal signature (4). the pilosebaceous unit are likely major stabi-
biota’s role in shaping and fortifying the One of the major drivers of the skin’s micro- lizers of this effect in the human skin micro-
barrier function of the skin. We also con- bial ecology is the pilosebaceous unit, a skin biome. The spatial architecture and hypoxic
sider pathological microbe–host interactions
and their role in skin disease and disruptions Lifestyle factors Systemic host factors Environmental factors
to other organ systems. Finally, we consider Skin product Antibiotics and Age Ultraviolet and other radiation
how these interactions could be leveraged to usage medications Hormones Climate
prevent or treat skin disease and impaired Social and Hygiene Genetic variation Pollution
wound healing. physical Diet
environment Immune system
Microbial ecology of the human skin Local host factors
Human skin, with its hypersaline and acidic Sweat
Water, Na, Cl, K, urea, lactate
environment and low nutrient availability, is
distinct from other mucosa and epithelia. Both Keratinized squames
Amino acids
culture-based approaches and metagenomic
Sebum
profiling strategies of increasing resolution Triglycerides, free fatty acids,
demonstrate that the human skin microbiota wax esters, squalene
comprises a restricted set of bacterial, fun-
gal, and viral inhabitants (1–3). Within the skin, Skin commensals
bacteria predominantly belong to three phyla, Ac- Corynebacterium species
tinobacteria, Firmicutes, and Proteobacteria,
Cutibacterium acnes
ILLUSTRATION: KELLIE HOLOSKI/SCIENCE

with associated bacteriophages further modu-


lating bacterial community dynamics and vir- Coagulase-negative
ulence. Eukaryotic viruses are also present, Staphylococcus species

Malassezia species
1
Department of Dermatology, University of Texas Southwestern
Medical Center, Dallas, TX, USA. 2Departments of Dermatology Fig. 1. Factors that influence the microbial colonization of skin. Local, systemic, environmental, and
and Microbiology, Perelman School of Medicine, University lifestyle factors together contribute to colonization and stability of the skin microbiome. Pictured are
of Pennsylvania, Philadelphia, PA, USA.
Email: tamia.harris-tryon@utsouthwestern.edu (T.A.H.-T.); common human skin commensals. Hydrophilic and lipophilic microbes are indicated with a water droplet
egrice@pennmedicine.upenn.edu (E.A.G.) (blue) and oil droplet (yellow), respectively.

Harris-Tryon et al., Science 376, 940–945 (2022) 27 May 2022 1 of 6


conditions of the pilosebaceous unit Chemical barrier
impose a bottleneck on C. acnes, In addition to the physical distance
allowing early colonizing strains of from the environment provided by
C. acnes to predominate a given fol- the corneocytes, keratinocytes, and
licle with limited competition (6). skin lipids, the acidic skin surface
Although pulse disturbance exper- creates a chemical environment that
iments with topical antiseptics dis- restricts bacterial colonization. Both
place sensitive lower-abundance taxa, Microbial C. acnes and Corynebacterium spp.
barrier
C. acnes remains during commu- secrete lipases that hydrolyze free
nity recovery (7). By contrast, lon- fatty acids from triglycerides in se-
ger disruptions to the microbiota bum (16, 17). Free fatty acids further
through the use of systemic anti- augment skin immunity by directly
biotics impose long-lasting effects inhibiting bacteria and by stimulat-
at the community level and drive Lipase ing the expression of human b-defensin
selection for antibiotic-resistant 2 (hBD-2) (18). C. acnes also binds di-
staphylococci. Systemic antibiot- rectly to free fatty acids, suggesting
ics also increase gene mobilization that the availability of free fatty acids
Physical Chemical
among the microbiome, which is barrier barrier facilitates the colonization of C. acnes.
indicative of a stress response (8).
Innate immune barrier
Microbes fortify multiple facets The microbiota is intimately asso-
Immune
of the skin barrier barrier ciated with the skin epithelium, and

Downloaded from https://www.science.org on December 13, 2023


The skin is a formidable structure the host and microbe have the ca-
composed of a stratified, cornified pacity for cross-talk. Microbes can
epithelium of keratinocytes, which Fig. 2. The skin microbiota mediates multiple levels of barrier function. stimulate a range of innate immune
undergo terminal differentiation. Skin microbes form the first barrier against the environment through various responses that often depend on the
These physical structures are fur- mechanisms of colonization resistance, including resource exclusion, direct metabolic and inflammatory con-
ther fortified by chemical and im- inhibition, and/or interference. The skin microbiota also contributes to the texts. For example, filamentous and
munological features that enhance differentiation and epithelialization of the physical skin barrier. Microbes boost yeast forms of Candida albicans stim-
the barrier. The skin microbiota af- the chemical barrier of the skin by producing lipases that digest sebum ulate distinct immune responses in
fects all aspects of the skin barrier, triglycerides to free fatty acids, which amplify the acidity of skin and restrict the skin (19). Similarly, the T cell re-
while also directly interacting with colonization by transient and pathogenic species. Finally, microbes stimulate sponse to S. epidermidis in skin re-
commensal and pathogenic mi- innate and adaptive immune defenses such as release of antimicrobial peptides, quires the expression of specific
crobes encountered at the surface induction of neonatal tolerance, and development of protective immunity. glycans on the bacterial surface that
(Fig. 2). We next discuss how mi- interact with C-type lectins on host
crobes interact with the skin bar- innate immune cells (20). Oxygen
rier’s microbial, chemical, and innate and limited to CoNS species in the skin microbiota. availability can also affect host–microbial inter-
adaptive immune components. C. acnes competes to maintain its niche in the actions at the skin surface. The microaerophilic
human pilosebaceous unit, with specific strains bacterium C. acnes ferments the glycerol back-
Microbial barrier producing a thiopeptide antibiotic, cutimycin, bone of triglyceride and generates short-chain
The skin microbiota itself is a barrier against that limits S. aureus colonization (13). How these fatty acids (SCFAs). In turn, SCFAs inhibit his-
invasion, colonization, and infection by for- individual interactions coalesce in a commu- tone deacetylases (HDACs), which can act as
eign and pathogenic microbes. Living in poly- nity setting and how this affects the community epigenetic regulators of the immune system (21).
microbial communities, skin microbes vie for structure and function remain unclear. Unlike in the gastrointestinal tract, where SCFAs
resources and have evolved mechanisms to have anti-inflammatory effects on gut immunity,
directly antagonize their rivals. Multiple CoNS Physical barrier SCFAs have proinflammatory effects in the skin.
species, such as Staphylococcus hominis, produce Keratinocytes undergo a program of tightly By means of keratinocytes, C. acnes–derived
antibiotics with unique chemistry and potent regulated terminal differentiation to form the SCFAs inhibit HDAC8 and HDAC9 and stimu-
inhibitory activity against the major skin stratum corneum, a process that also can be late inflammation through TLR signaling (22).
pathogen Staphylococcus aureus (9). Other mediated by the microbiota. Self-renewing basal In the sebaceous gland, SCFAs derived from
species such as Staphylococcus capitis antag- keratinocytes exit the cell cycle and acquire the C. acnes fermentation augment inflammation
onize S. aureus through interference with the machinery (e.g., intermediate filaments and through the activation of the free fatty acid
accessory gene regulator (agr) quorum sensing lipid granules) that together form the “bricks receptor (23). Thus, a microbe’s metabolic and
pathways, which are required for S. aureus and mortar” of the permeability barrier. This inflammatory context can result in distinct
virulence (10, 11). Notably, many of these barrier also directly interfaces with the micro- types of immune responses.
ILLUSTRATION: KELLIE HOLOSKI/SCIENCE

antagonistic mechanisms synergize with host biota, resident or transient, and is subject to Skin microbes further bolster skin immunity
antimicrobial responses. For example, lugdu- microbial regulation. The microbiota is required by stimulating the production of host-derived
nin, a peptide antibiotic produced by Staphy- for normal skin barrier structure and function in antimicrobial peptides and proteins (AMPs),
lococcus lugdunensis, induces keratinocytes to mice and promotes differentiation and epithelial which act as natural antibiotics. The expression
produce the antimicrobial peptide LL-37 and integrity through signaling of the keratinocyte of the AMP LL-37, a fragment of the protein
neutrophil chemoattractant CXCL8 through aryl hydrocarbon receptor (AHR) (14). Skin bacte- cathelicidin, increases in response to activation
the Toll-like receptor–myeloid differentiation ria also secrete sphingomyelinases that process of TLR signaling initiated by microbial signals
primary response protein 88 (TLR–MyD88) lamellar lipids into ceramides (15), a critical (24). In addition to the cathelicidin family of
pathway (12). Competitive mechanisms are not component of the stratum corneum. AMPs, the skin also generates members of the

Harris-Tryon et al., Science 376, 940–945 (2022) 27 May 2022 2 of 6


T H E S YS T E M IC MI C R O B I O M E

b-defensin family with bactericidal action against However, this interaction is context-dependent, biota (41). Thus, microbial metabolites produced
Escherichia coli and S. aureus strains. The as a high-fat diet instead promotes cutaneous by skin commensals are depleted during dis-
sebaceous gland responds to Gram-negative inflammation (17). Thus, the inflammatory ease states, which may maintain and exacer-
lipopolysaccharide by generating the small milieu present at the time of microbial expo- bate inflammation and barrier disruption.
proline-rich proteins SPRR1 and SPRR2, which sure affects the immune response within the A dysregulated or dysfunctional immune sys-
directly disrupt negatively charged bacterial skin. Taken together, these findings highlight tem also has impacts on the skin microbiota,
membranes (25). The skin also produces nu- the key role that microbes play in the recruitment which can further exacerbate disease. S. aureus
merous cationic intrinsically disordered pro- and stimulation of immune cells in the skin. and S. epidermidis, for example, are more abun-
teins with broad antimicrobial activity (26). dant and cause greater amounts of skin damage
These AMPs act in concert to provide the skin Pathological microbial–host interactions in Netherton syndrome patients, who have a
with a range of antimicrobial defenses against and skin disorders genetic defect in the skin protease inhibitor
the microbes encountered in the environment. From an ecological standpoint, microbial com- lymphoepithelial Kazal-type–related protease
The skin microbiota also helps coordinate in- munities are inevitably destined to change in inhibitor 1 (LEKTI-1) (42). Skin infections and
nate immune responses during wound repair. structure and function when their niche is dis- shifts in skin colonization can also occur in hu-
Similar to observations made in the lung and gut, rupted. As such, an altered skin microbiome is mans with primary immunodeficiency disor-
the commensal microbiota in the skin elicits a more often the rule than the exception in skin ders. For example, in patients with dedicator of
type I interferon (IFN) response during this pro- disease. Shifts in resource availability and, in cytokinesis protein 8 (DOCK8) deficiency, the
cess (27). In response to microbial stimuli, neutro- some cases, complete devastation of their habi- cutaneous virome is enriched with a diversity
phils express CXCL10, which recruits activated tat are factors that drive the depletion of normal of eukaryotic viruses, including human papil-
plasmacytoid dendritic cells (pDCs) to sites of skin residents in favor of opportunists. Owing loma viruses (43). Genetic deletion studies in
injury. pDCs generate type I IFNs, which acceler- to the tight interconnectivity of the microbiota murine models further highlight the key role
ate wound repair through stimulation of fibro- with its host, it is difficult to distinguish be- that the immune system plays in restricting the

Downloaded from https://www.science.org on December 13, 2023


blast and macrophage growth factor responses. tween “the chicken and the egg” in the absence microbiota. Mice devoid of type 2 innate lym-
Indeed, the recruitment of antigen-presenting of experimental approaches that rely on cultured phoid cells (ILC2s) have enlarged sebaceous
cells to the skin is microbiota-dependent (28). isolates in experimental model systems. Whether glands and generate greater amounts of anti-
Similarly, microbes enhance skin regeneration causative or a consequence, altered microbial microbial lipids that restrict colonization of
in wound repair and hair follicle neogenesis communities can mediate tissue damage and/or Gram-positive commensals (44). By contrast,
through a process that requires interleukin-1 inflammation across a variety of skin disorders. mice lacking T cells and epidermal expression
receptor (IL-1R)–MYD88 signaling (29). S. aureus is a frequent opportunist of the skin of the transcription factor JunB are unable to
and overwhelms the commensal microbiota in control S. aureus inflammation at the skin
Adaptive immune barrier barrier disorders such as atopic dermatitis (AD) surface and recapitulate several aspects of
The skin is home to a diverse repertoire of adapt- and skin wounds (34–36). In the setting of atopic inflammation (45).
ive immune cells, among them vast pools of disease, S. aureus can evade the host immune As shotgun metagenomics and culture-based
resident memory T cells poised to respond to response to establish chronic infection. Moreover, investigations have advanced, it is becoming
various environmental stimuli, including patho- S. aureus and some CoNS species produce pro- clear that strain-level variations of human skin
genic and commensal microbes. In early infancy, teases and other factors that further damage the commensals and pathogens also have an impact
exposure to the skin commensal S. epidermidis barrier and drive pathological inflammatory on disease pathogenesis. Specific strains of
mediates the influx of regulatory T cells (Tregs) responses (11, 37). In addition to direct damage, S. aureus correlate with disease severity and clin-
into the skin (30). This wave of Treg migration S. aureus interferes with adaptive immune re- ical outcomes in different contexts (34, 36).
occurs concurrently with hair follicle development sponses by producing alpha toxins that trigger Acneic skin is colonized with C. acnes strains that
and requires the production of chemokines gen- IL-1R–mediated inflammation and prevent the inherently produce greater amounts of the pro-
erated by the hair follicle keratinocytes (30, 31). accumulation of S. aureus–specific Tregs and inflammatory metabolite porphyrin compared
Tregs, along with many other immune cell sub- the development of tolerance to S. aureus later with C. acnes strains recovered from healthy skin
sets in the skin, ultimately reside adjacent to the in life (38). (46). Moreover, porphyrin production by C. acnes
hair follicle, with specificity to the microbial anti- In AD and other dysbiotic contexts where is under the control of vitamin B12. A vitamin B12
gens detected during this developmental window. an opportunist overtakes the ecosystem, there supplementation study in humans showed in-
In a parallel process, mucosal-associated in- is a depletion of the commensal microbes and creased porphyrin production leading to acne
variant T (MAIT) cells are acquired in infancy their mediators that previously supported the development (47). This is a potential molecu-
during a similar time-restricted developmen- skin’s barrier defenses. For example, tryptophan lar mechanism that may explain how the same
tal window. MAIT cells are absent in germ-free metabolites are reduced in AD skin. When these species of bacteria can both cause disease and
mice, and their development requires vitamin metabolites are therapeutically administered, reside as a member of the healthy skin micro-
B2 metabolites that are only produced by bacte- inflammation in mouse models of AD is at- biota. A challenge going forward will be to iden-
ria and fungi, not mammalian cells. In the thymus, tenuated by AHR (39). Coal tar, one of the oldest tify specific markers of virulence within bacterial
exposure to 5-(2-oxopropylideneamino)-6-D- therapies for AD, can activate the AHR in the strains that can inform management strategies
ribitylaminouracil, a bacterial metabolite skin to drive differentiation programs, AMP for problematic microbial burdens.
of vitamin B2 trafficked to the thymus from expression, and normalization of the micro- Another advantage of culture-independent
mucosal sites, mediates MAIT cell expansion biome (40). Dysregulation of tryptophan catab- approaches is that they have greatly facilitated
and targeting to the skin and mucosal sites olism by the microbiota may also contribute the identification of fastidious anaerobic micro-
(32, 33). Microbial cell surface molecules can to hidradenitis suppurativa (HS), a condition biota in skin disease and wounds. In chronic
also act as signals to the host. Most species of characterized clinically by festering wounds of wounds and HS, mixed communities of Gram-
Corynebacterium contain mycolic acid in their the armpits and groin whose pathogenesis is positive anaerobic bacteria can inhabit deeper
cell envelope. Mycolic acid from Corynebacterium poorly understood. HS lesions are also defi- tissues of the skin (48). Although the skin com-
species can promote gd T cell accumulation in cient in AHR activation, which coincides with a mensal C. acnes may be found in some abun-
an IL-23–dependent manner under steady state. depletion of tryptophan-metabolizing micro- dance in these mixed communities, other players,

Harris-Tryon et al., Science 376, 940–945 (2022) 27 May 2022 3 of 6


such as Anaerococcus spp., Porphyromonas spp., skin has the capacity to control and restrict An emerging area of investigation is the in-
Finegoldia spp., Veillonella spp., and Peptostrep- commensal responses independently of other terface between skin microbiota and the neuro-
tococcus spp., are more common. In chronic mucosal sites through a sophisticated network immune axis. Bacteria can directly activate
wounds, the persistence of anaerobic commu- of immune strategies (57). More work is needed sensory neurons in the skin and cause pain
nities after debridement is associated with poor to uncover the many ways that these regulatory through the production of pore-forming toxins
wound outcomes (36, 49). The challenges of mechanisms, which contribute to sustained (62). As in interactions with other aspects of the
isolation and study of anaerobes, especially compartmentalization, malfunction in disease. host, variation at the strain level drives variable
in mixed communities, are limiting factors The gut microbiome can also affect skin in- responses, depending on the presence of spe-
in advancing our understanding of their role in flammation. For example, type 3 inflammation cialized toxins and quorum sensing systems.
skin disorders such as HS and chronic wounds. in a mouse psoriasis model is dampened in Sensory neurons in the skin are also directly
Psoriasis is another common inflammatory germ-free mice, which lack a microbiome (58). activated by the fungal pathogen C. albicans, and
skin disease. In contrast to AD and other bar- Moreover, mice that are sensitized to allergens stimulation is required for gd T cell immunity to
rier disorders, alterations to the skin micro- in the intestine through oral administration control cutaneous candidiasis through release
biota in psoriasis are more subtle, less consistent develop antigen-specific T cells in the skin after of neuropeptide CGRP (63). By contrast, the
across studies, and are more weakly associated epicutaneous challenge with the same antigen pathogen Streptococcus pyogenes, which causes
with disease (35, 50, 51). Thus, there is currently (59). In both cases, activation of the intestinal im- necrotizing fasciitis, directly activates nociceptor
limited evidence that the skin microbiota drives mune networks affects the amplitude of the in- neurons by secreting streptolysin S, which in
psoriasis pathogenesis. However, in a psoriasis flammatory signals in the skin. Thus, alterations turn promotes neuropeptide CGRP release and
mouse model, C. albicans exposure augmented in the gut microbiome may affect skin immunity, inhibits killing of S. pyogenes. In this context,
T helper 17 cell immunity with increased infil- although clear targets for therapeutic avenues to CGRP antagonism prevents necrotizing infec-
tration of and IL-17 production by ab T cells influence skin disease through modulation of the tion (64). Although these studies have focused
sensitized by Candida (52). Microbial–host in- intestinal microbes remain undefined. What has on skin pathogens in neuroimmune interac-

Downloaded from https://www.science.org on December 13, 2023


teractions at other mucosal sites are hypothe- been shown is that the dietary impacts on the gut tions, how skin commensals, at the community
sized to contribute to psoriasis as well, as microbiome, especially dietary fiber, have mean- level, contribute to our sensory perceptions
discussed in the next section. ingful effects on systemic immunity (60). Cutane- under homeostatic conditions remains under
ous innate immune responses are also linked to investigation.
Systemic roles for the skin microbiome the gut, where adequate expression of AMPs
There is increasing evidence that skin dam- that protect against bacterial skin infection is Outlook and conclusions
age and sensitization can affect other barrier dependent on dietary vitamin A (61). Together, The application of molecular, culture-independent
sites, such as the intestine and the lung (Fig. 3). these findings strengthen our molecular under- techniques to survey microbial communities
For example, superficial skin damage causes standing of the importance of diet in the de- has reinvigorated the study of skin micro-
keratinocytes to release IL-33 systemically. In velopment of host immunity. biota and its role in dermatological health
synergy with IL-25, IL-33 triggers the activation
of ILC2s in the intestine to generate IL-4. This,
in turn, stimulates the expansion of mast cells in
the intestine, where they are poised to respond to
food allergens and mediate anaphylaxis (53).
Wounding of the skin also augments intestinal Keratinocyte IL-36 Pore forming
Serum IgE toxins TRPV1 Pain
inflammation in dextran sodium sulfate–induced
colitis mouse models, which mimic inflammatory
bowel disease. Cross-talk between the skin and Lung inflammation
A B C
gut depends on the production of hyaluronan
fragments generated in the dermis during injury
that stimulate intestinal fibroblasts to differen- Allergic sensitization S. aureus infection
tiate into proinflammatory adipocytes through plus S. aureus infection Skin injury
D
a process called reactive adipogenesis. These
reactive adipocytes propagate gut inflamma-
tion through the production of AMPs and other Gut inflammation Hyaluronan Dietary fiber
reactive adipocytes gut microbiota
inflammatory mediators (54).
Skin sensitization also affects the lungs. Epi- Cytotoxic
T cell
demiological evidence demonstrates that many Tumor
patients progress through an “atopic march,” cell
first presenting with the skin barrier condition death
AD and subsequently developing allergic rhini- Melanoma response to therapy
tis, food allergies, and asthma (55). Epicutaneous
ILLUSTRATION: KELLIE HOLOSKI/SCIENCE

exposure to S. aureus stimulates keratinocytes to Fig. 3. Skin cross-talk with other organ systems is mediated by the microbiota. Emerging evidence
produce IL-36, which amplifies serum immuno- highlights the role of skin cross-talk with distant organ systems, which is driven by host–microbiota interactions.
globulin E (IgE) levels. Mice lacking the IL-36 Depicted are four examples of cross-talk between skin and other organ systems. (A) Allergic sensitization of skin,
receptor do not develop elevated IgE in response together with S. aureus infection, results in IL-36–dependent lung inflammation, suggesting a potential mechanism
to S. aureus and are also protected from allergen- for the “atopic march.” (B) Skin injury releases hyaluronan fragments systemically, which drives reactive
specific lung inflammation (56). These findings adipogenesis, gut inflammation, and dysbiosis in murine models. (C) During infection, S. aureus releases pore-
provide evidence for skin exposure to microbial forming toxins that are implicated in directly activating nociceptors and causing pain. TRPV1, transient receptor
pathogens as an initiating event in systemic potential cation channel subfamily V member 1. (D) The gut microbiome and dietary fiber contribute to melanoma
inflammation. However, it is notable that the response to immune checkpoint therapy, driving cytotoxic T cell accumulation and killing of tumor cells.

Harris-Tryon et al., Science 376, 940–945 (2022) 27 May 2022 4 of 6


T H E S YS T E M IC MI C R O B I O M E

Introduce to model systems of developed by screening human skin commen-


skin host-microbe interaction sals for AHR activation in keratinocytes (14).
Culture-
dependent Colonization with this consortium improved
barrier function in germ-free mice and re-
duced disease severity in murine models of
Cultured isolate collection AD. The Gram-negative Roseomonas mucosa,
isolated from healthy human skin, was once
In vivo animal models (mouse, pig)
explored for the treatment of AD, but clinical
Culture- A A Ex vivo human skin
independent Recover trials were ultimately discontinued owing to
Collect skin B B isolate(s) In vitro 3D organotypics, failure to meet end points (67). Other microbiota-
microbiota specimens of interest reconstituted human epidermis,
human primary keratinocytes based therapeutic approaches are less developed
C C
in the skin but could include phage-directed
Taxonomic relative therapies to target pathogens, engineering com-
abundance Examine Identify and mensals to express molecules of benefit, and/or
skin test host
phenotypes mechanisms prebiotic approaches to modify the habitat and
thus the microbiota (68).
Although microbes have tremendous ther-
apeutic potential, an ecological perspective of
Community diversity community-level interactions between host and
and richness
microbes is needed to inform efforts to manip-
Identify or Histopathology, ulate the microbiome. Selection of a consortium
Experimental or generate immunostaining with desired functional attributes is only the first

Downloaded from https://www.science.org on December 13, 2023


disease condition mutant of Barrier function (TEER, TEWL) step, as major obstacles to the delivery and sta-
Healthy or unaffected interest
control skin Gene expression, protein, ble engraftment of transplanted communities
Metabolic and genetic metabolite profiling remain. The host and the endogenous micro-
Negative (contamination)
controls pathway enrichment Immune and microbiome profiling biota have powerful effects on the establish-
ment, persistence, growth, and long-term impact
Fig. 4. Framework for examining functional and mechanistic attributes of the skin microbiota.
of a transplanted community and are likely
Illustrated is an example pipeline for using skin microbiome profiles to inform functional and mechanistic
factors that influence the engraftment of a
investigations. Applying a combination of culture-independent and culture-dependent approaches allows
transplant (69). Spatial architecture of the
examination of specific microbes or consortia in the context of skin model systems. Furthermore, shotgun
pilosebaceous unit likely limits the complete
metagenomic sequencing can inform which metabolic or genetic pathways to target in the microbiota, which
removal of skin microbes, even with topical
then can be functionally interrogated by examining mutants for these pathways. Deep phenotyping of skin or
treatments meant to sterilize skin. These pro-
skin models colonized or associated with clinical isolates, mutants, and/or consortia facilitates hypothesis
tected structures may serve as reservoirs to
generation regarding host interactive pathways, which then can be tested in this pipeline. TEER,
“reseed” the skin microbiome following dis-
transepithelial electrical resistance; TEWL, transepidermal water loss.
turbance. Additionally, it is now apparent that
skin microbes are highly specialized to their
and disease. Interpreting results from skin micro- isolates or obtained from collections of labora- niches, reflecting millions of years of adaptation
biota surveys, such as 16S rRNA gene sequencing tory isolates, to test the role of these pathways in to human skin, and not only interact with the
or shotgun metagenomics, and using these data driving skin phenotypes. Deep-skin phenotyp- local tissue microenvironment but drive sig-
to guide a deeper understanding of functional ing, for example, using unbiased assays to mea- nals at distant organs as well. Unsurprisingly,
and mechanistic attributes of the microbiota sure gene expression can further define how disrupting microbe–host relationships in the
remains a challenge for the field. We posit that the host responds to the skin microbiota. skin has consequences on organ structure and
application of both culture-independent and Therapeutic advancement based on the skin function. Introducing a new member to the
culture-dependent approaches, within an ecolog- microbiome will rely on such approaches to community undoubtedly triggers responses
ical framework of community-wide interactions, identify candidates for either enhancement or from both host and microbial cells. Under-
can advance our understanding of homeo- depletion in the community. Bacteriotherapy, standing how skin microbial communities
static and pathological mechanisms at the host– or transplantation of live, defined bacteria, is interact with the host and each other is cru-
microbiota interface (Fig. 4). Skin microbiome currently under development for the treatment cial to inform transplantation strategies and
surveys can reveal the composition and diver- of AD using a strain of S. hominis that was all types of microbial-based therapeutics that
sity of the community and be combined with isolated from healthy human skin and inhibits target this interface for the prevention and
culture-based approaches to identify isolates S. aureus (9). Phase 1 trials in S. aureus–positive treatment of skin disorders.
or target the isolation of community members. AD patients (n = 54) indicate the safety of
These isolates can then be used individually or S. hominis A9 and demonstrate a reduction of REFERENCES AND NOTES

in combination to test their effects on the skin S. aureus colonization, although overall clin- 1. G. D. Hannigan et al., mBio 6, e01578-15 (2015).
2. J. Oh et al., Temporal stability of the human skin microbiome.
ILLUSTRATION: KELLIE HOLOSKI/SCIENCE

in vivo with model organisms, in vitro using ical severity of disease was not significantly Cell 165, 854–866 (2016).
human skin cells and constructs, or ex vivo affected (65). Moreover, a placebo-controlled trial 3. S. Saheb Kashaf et al., Nat. Microbiol. 7, 169–179 (2022).
4. D. M. Chu et al., Nat. Med. 23, 314–326 (2017).
using human skin explants. In this framework, using lysates of the Gram-negative Vitreoscilla 5. J. Park et al., J. Invest. Dermatol. 142, 212–219 (2022).
shotgun metagenomics of ample read depths filiformis has proven beneficial through the 6. A. Conwill et al., Cell Host Microbe 30, 171–182.e7 (2022).
can be a useful approach for strain-level charac- stimulation of IL-10–producing dendritic cells 7. A. J. SanMiguel et al., J. Invest. Dermatol. 138, 2234–2243 (2018).
8. J.-H. Jo et al., Sci. Transl. Med. 13, eabd8077 (2021).
terization of the community or can identify within the skin (66). Screening strategies may 9. T. Nakatsuji et al., Sci. Transl. Med. 9, eaah4680 (2017).
functional genetic pathways that are enriched also be tailored to identify microbes that ac- 10. A. E. Paharik et al., Cell Host Microbe 22, 746–756.e5 (2017).
11. M. R. Williams et al., Sci. Transl. Med. 11, eaat8329 (2019).
within a sample. Genetic deletion of these path- tivate or repress host pathways of interest. The 12. K. Bitschar et al., Nat. Commun. 10, 2730 (2019).
ways or genes can then be targeted in clinical “Flowers’ Flora” consortium, for example, was 13. J. Claesen et al., Sci. Transl. Med. 12, eaay5445 (2020).

Harris-Tryon et al., Science 376, 940–945 (2022) 27 May 2022 5 of 6


14. A. Uberoi et al., Cell Host Microbe 29, 1235–1248.e8 (2021). 39. J. Yu et al., J. Allergy Clin. Immunol. 143, 2108–2119.e12 (2019). 62. K. J. Blake et al., Nat. Commun. 9, 37 (2018).
15. Y. Zheng et al., Cell Host Microbe 30, 301–313.e9 (2022). 40. J. P. H. Smits et al., J. Invest. Dermatol. 140, 415–424.e10 (2020). 63. S. W. Kashem et al., Immunity 43, 515–526 (2015).
16. L. Bomar, S. D. Brugger, B. H. Yost, S. S. Davies, K. P. Lemon, 41. L. Guenin-Macé et al., JCI Insight 5, e140598 (2020). 64. F. A. Pinho-Ribeiro et al., Cell 173, 1083–1097.e22 (2018).
mBio 7, e01725–e15 (2016). 42. M. R. Williams et al., Cell Rep. 30, 2923–2933.e7 (2020). 65. T. Nakatsuji et al., Nat. Med. 27, 700–709 (2021).
17. V. K. Ridaura et al., J. Exp. Med. 215, 785–799 (2018). 43. O. Tirosh et al., Nat. Med. 24, 1815–1821 (2018). 66. T. Volz et al., J. Invest. Dermatol. 134, 96–104 (2014).
18. T. Nakatsuji et al., J. Invest. Dermatol. 130, 985–994 (2010). 44. T. Kobayashi et al., Cell 176, 982–997.e16 (2019).
67. I. A. Myles et al., Sci. Transl. Med. 12, eaaz8631 (2020).
19. S. W. Kashem et al., Immunity 42, 356–366 (2015). 45. Ö. Uluçkan et al., Cell Rep. 29, 844–859.e3 (2019).
68. M. T. Sorbara, E. G. Pamer, Nat. Rev. Microbiol. (2022).
20. Y. E. Chen et al., bioRxiv 664656 [Preprint] (2019). https://doi. 46. T. Johnson, D. Kang, E. Barnard, H. Li, MSphere 1, e00023-15
69. J. Walter, M. X. Maldonado-Gómez, I. Martínez, Curr. Opin.
org/10.1101/664656. (2016).
47. D. Kang, B. Shi, M. C. Erfe, N. Craft, H. Li, Sci. Transl. Med. 7, Biotechnol. 49, 129–139 (2018).
21. P. V. Chang, L. Hao, S. Offermanns, R. Medzhitov, Proc. Natl.
Acad. Sci. U.S.A. 111, 2247–2252 (2014). 293ra103 (2015).
48. S. C. Williams, J. W. Frew, J. G. Krueger, Exp. Dermatol. 30, AC KNOWLED GME NTS
22. J. A. Sanford et al., Sci. Immunol. 1, eaah4609 (2016).
23. J. A. Sanford, A. M. O’Neill, C. C. Zouboulis, R. L. Gallo, J. Immunol. 1388–1397 (2021). We thank the members of the Grice lab and the Harris-Tryon lab
202, 1767–1776 (2019). 49. S. Verbanic, Y. Shen, J. Lee, J. M. Deacon, I. A. Chen, NPJ for their underlying contributions. Funding: This work was
24. Y. Lai et al., J. Invest. Dermatol. 130, 2211–2221 (2010). Biofilms Microbiomes 6, 21 (2020). supported by National Institutes of Health grants R01NR015639
25. C. Zhang et al., eLife 11, e76729 (2022). 50. H.-W. Chang et al., Microbiome 6, 154 (2018). and R01AI143790 (E.A.G.) and K08AR076459 (T.A.H.-T.), a Linda
26. T. Latendorf et al., Sci. Rep. 9, 3331 (2019). 51. M. A. Loesche et al., J. Invest. Dermatol. 138, 1973–1981 Pechenik Montague Investigator Award (E.A.G.), a Burroughs
27. J. Di Domizio et al., Nat. Immunol. 21, 1034–1045 (2020). (2018). Wellcome Fund PATH Award (E.A.G.), a Burroughs Wellcome Fund
28. N. D. Ubags et al., J. Allergy Clin. Immunol. 147, 1049–1062.e7 52. C. Hurabielle et al., Proc. Natl. Acad. Sci. U.S.A. 117, 16465–16474 CAMS Award (T.A.H.-T.), a Dermatology Foundation Sun Pharma
(2021). (2020). Research Award (E.A.G.), and a Robert Wood Johnson Foundation
29. G. Wang et al., Cell Host Microbe 29, 777–791.e6 (2021). 53. J.-M. Leyva-Castillo et al., Immunity 50, 1262–1275.e4 (2019). Harold Amos Award (T.A.H.-T.). Competing interests: The
30. T. C. Scharschmidt et al., Immunity 43, 1011–1021 (2015). 54. T. Dokoshi et al., J. Clin. Invest. 131, e147614 (2021).
authors declare that they have no competing interests. License
31. T. C. Scharschmidt et al., Cell Host Microbe 21, 467–477.e5 (2017). 55. A. S. Paller, J. M. Spergel, P. Mina-Osorio, A. D. Irvine, J. Allergy
32. M. G. Constantinides et al., Science 366, eaax6624 (2019). information: Copyright © 2022 the authors, some rights reserved;
Clin. Immunol. 143, 46–55 (2019).
33. F. Legoux et al., Science 366, 494–499 (2019). 56. G. J. Patrick et al., J. Clin. Invest. 131, e143334 (2021). exclusive licensee American Association for the Advancement of
34. A. L. Byrd et al., Sci. Transl. Med. 9, eaal4651 (2017). 57. S. Naik et al., Science 337, 1115–1119 (2012). Science. No claim to original US government works. https://www.
35. N. Fyhrquist et al., Nat. Commun. 10, 4703 (2019). 58. Z. Zákostelská et al., PLOS ONE 11, e0159539 (2016). science.org/about/science-licenses-journal-article-reuse
36. L. R. Kalan et al., Cell Host Microbe 25, 641–655.e5 (2019). 59. M. K. Oyoshi et al., J. Clin. Invest. 121, 2210–2220 (2011).
37. L. Cau et al., J. Allergy Clin. Immunol. 147, 955–966.e16 (2021). 60. C. N. Spencer et al., Science 374, 1632–1640 (2021). Submitted 28 February 2022; accepted 26 April 2022
38. J. M. Leech et al., Cell Host Microbe 26, 795–809.e5 (2019). 61. T. A. Harris et al., Cell Host Microbe 25, 777–788.e8 (2019). 10.1126/science.abo0693

Downloaded from https://www.science.org on December 13, 2023

Harris-Tryon et al., Science 376, 940–945 (2022) 27 May 2022 6 of 6


Microbiota and maintenance of skin barrier function
Tamia A. Harris-Tryon and Elizabeth A. Grice

Science 376 (6596), . DOI: 10.1126/science.abo0693

View the article online


https://www.science.org/doi/10.1126/science.abo0693

Downloaded from https://www.science.org on December 13, 2023


Permissions
https://www.science.org/help/reprints-and-permissions

Use of this article is subject to the Terms of service

Science (ISSN 1095-9203) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW,
Washington, DC 20005. The title Science is a registered trademark of AAAS.

Copyright © 2022 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim
to original U.S. Government Works

You might also like