You are on page 1of 2

Cell Host & Microbe

Previews

Seeing Is Believing: Vitamin A Promotes


Skin Health through a Host-Derived Antibiotic
Jens-M. Schröder1,*
1Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany

*Correspondence: jschroeder@dermatology.uni-kiel.de
https://doi.org/10.1016/j.chom.2019.05.011

In this issue of Cell Host & Microbe, Harris et al. (2019) show that members of the resistin-like molecule
(RELM) family are bactericidal, antimicrobial peptides that promote skin defenses in a vitamin-A-dependent
manner. Administration of vitamin A analogs triggers RELM, which improves host resistance against skin
pathogens in mice.

Body surfaces—including those of the gut, molecule a (RELMa), which belongs to proteins. Indeed, Harris et al. (2019)
the lung, and the skin—are intricate habi- the protein family that encompasses resis- demonstrated that a S. aureus mutant
tats for numerous commensal and environ- tin and the resistin-like molecules lacking STX is more susceptible to killing
mental bacteria, fungi, and viruses. In (RELMs), was found to be strongly by mRELMa and hRETN, indicating that
healthy skin, commensal bacteria reside induced by bacterial exposure. Resistin STX is required for S. aureus resistance
mainly within the different layers of the and other RELMs have been characterized to RELM bactericidal activity. This sug-
dead keratinocyte-containing stratum cor- as hormones that modulate insulin pro- gests that RELMs may disrupt mem-
neum (Zeeuwen et al., 2012) but also grow duction. It, however, was recently found branes similarly to the majority of pore-
within skin appendages like sweat glands that RELMb is a Gram-negative, bacteria- forming amphipathic AMPs. Liposome
and hair follicles with sebaceous glands. killing AMP, promoting host-bacterial disruption assays showed that mRELMa
It is currently believed that—apart from mutualism in the intestine (Propheter and hRETN can permeabilize artificial
environmental conditions like nutrient et al., 2017). Thus, Harris et al. (2019) hy- membranes. Since these assays used
availability, salt, essential trace elements, pothesized that RELMa might be a bacte- liposomes with a lipid composition similar
water, and the pH—antimicrobial compo- ricidal protein of the skin. They found it to to that of bacterial membranes, the au-
nents of the host as well as the commensal be expressed in mouse keratinocytes as thors suggest cellular lysis as a possible
microflora shape the local microbiome. well as in sebocytes, cells of the seba- mechanism for the observed bactericidal
Thus, the skin surface has to manage inter- ceous glands. While the mouse genome activity. Similar findings were obtained
actions with this diverse microbial commu- encodes four RELM family members, the with mRELMb (Propheter et al., 2017),
nity mainly through epidermal-cell-based human genome encodes only two RELM suggesting that membrane lysis could
defense strategies with antimicrobial pep- proteins, resistin-like molecule b (RELMb) be a common mechanism of RELM mem-
tides (AMPs) as effector molecules. In this and resistin (RETN). The location of ber’s bactericidal properties. Interest-
issue of Cell Host & Microbe, Harris et al. RELMa and RETN expression in keratino- ingly, however, ultrastructural analyses
(2019) explore a vitamin-A-dependent cytes and sebaceous glands—similar of P. aeruginosa treated with mRELMb
role for murine resistin-like molecule a to the lipophilic AMP cathelicidin—led (Propheter et al., 2017) do not show the
(RELMa) and its human parlor resistin in them hypothesize that mRELMa and typical morphological signs of lytic
shaping the normal flora and its role as hRETN might function in antimicrobial de- AMPs with membrane disruption and
AMP in skin defense against certain bacte- fense of the skin. With recombinant bleb formation. Instead, intracytoplasmic
rial infections. mRELMa and hRETN, they identified protein aggregates are observed, simi-
Some epidermal AMPs are constitu- bactericidal activity for Streptococcus larly to P. aeruginosa treated with peptide
tively produced and others need to be pyogenes and Pseudomonas aeruginosa fragments of hornerin, an abundant pre-
induced, either upon infection, inflamma- and, of note, the highest sensitivity of Pro- cursor of highly hydrophilic, cationic,
tion, or wounding. One strategy to under- pionibacterium acnes, a skin commensal intrinsically disordered antimicrobial pep-
stand how the skin’s antimicrobial defense linked to acne and residing in a lipid-rich tides (CIDAMPs) found in the stratum cor-
is regulated by environmental factors is to environment. neum of healthy skin (Gerstel et al., 2018;
identify antimicrobial proteins whose Whereas the skin commensal Staphylo- Latendorf et al., 2019). CIDAMPs are ribo-
expression is inducible by bacteria. To coccus epidermidis is susceptible to some-targeting AMPs that have the
achieve this, Harris et al. (2019) used mRELMa and hRETN, a strain of the path- in vitro propensity to form microbicidal
whole-transcriptome RNA sequencing to ogen Staphylococcus aureus is unexpect- amyloid-like nanostructures (Gerstel
compare transcript abundances in the edly resistant, a propensity attributed to et al., 2018). Thus, it would be of interest
skin of germ-free mice to those in the its production of staphyloxanthin (STX), to explore whether members of the
skin of germ-free mice challenged with a yellow pigment that intercalates into RELM family have additional intracellular
the opportunistic skin pathogen Staphylo- the S. aureus membrane and protects it targets possibly also targeting the bacte-
coccus aureus. The protein resistin-like from attack by pore-forming antimicrobial rial ribosome.

Cell Host & Microbe 25, June 12, 2019 ª 2019 Elsevier Inc. 769
Cell Host & Microbe

Previews

The skin surface has unique physical contrast to several known mouse skin vitamin A, prior work identified a role for
and chemical properties relative to other antimicrobial proteins, which are not vitamin D in the expression of cathelicidin
body sites, including an acidic pH (Zloto- markedly affected. Thus, RELMa is (Schauber et al., 2006). Collectively, the
gorski, 1987), suggesting that an innate unique among known skin antimicrobial findings of Harris et al. (2019) and previ-
skin defense system should have adapted proteins in that its expression requires ous reports linking changes in the skin mi-
to these conditions. Indeed, Harris et al. vitamin A in the diet. This led to the ques- crobiome to skin conditions such as acne
(2019) found that the sensitivity of the tion of whether administration of exoge- vulgaris and hidradenitis suppurativa sug-
acid-resilient bacterial species Listeria nous therapeutic retinoids can stimulate gest that diet plays an important role in
monocytogenes toward mRELMa was RELMa expression in vivo. Harris et al. skin microflora and defense O’Neill and
highest at pH 5 and diminished at pH 7. (2019) now present experimental evi- Gallo, (2018).
This could indicate that skin pathogens dence that a synthetic retinoid rescues
like P. aeruginosa or Cutibacterium the increased susceptibility toward REFERENCES
(formerly Propionibacterium) acnes may S. pyogenes infection of mice fed with
also be more sensitive at acidic pH to- a vitamin-A-deficient diet. Conversely, Felipe, F., Bonet, M.L., Ribot, J., and Palou, A.
(2004). Modulation of resistin expression by reti-
ward RELMs—as seen for skin horny- treatment of RELMa-knockout mice with noic acid and vitamin A status. Diabetes 53,
layer-derived and histidine-rich CIDAMPs therapeutic retinoids does not alter 882–889.
(Latendorf et al., 2019). susceptibility to infection. Gerstel, U., Latendorf, T., Bartels, J., Becker, A.,
To interrogate the in vivo relevance of The study by Harris et al. (2019) provides Tholey, A., and Schröder, J.M. (2018). Hornerin
RELMs in skin innate immunity, Harris convincing evidence of a role for RELMa, contains a linked series of ribosome-targeting pep-
tide antibiotics. Sci. Rep. 8, 16158.
et al. (2019) studied the skin microbiota possibly generated by sebocytes, in
composition of RELMa-knockout mice vitamin-A-dependent antibacterial skin Harder, J., Meyer-Hoffert, U., Wehkamp, K.,
Schwichtenberg, L., and Schröder, J.M. (2004).
and found that wild-type and knockout defense in mice. Curiously, in adipocytes, Differential gene induction of human beta-defen-
mice have distinct skin microbiotas. retinoids inhibit the expression of resistin sins (hBD-1, -2, -3, and -4) in keratinocytes is in-
Consistent with their in vitro findings, the (Felipe et al., 2004), and in human epithelial hibited by retinoic acid. J. Invest. Dermatol. 123,
522–529.
relative abundance of coagulase-nega- cells, this adipokine inhibits beta-defensin
tive Staphylococci and Streptococcus is production (Harder et al., 2004). Thus, the Harris, T.A., Gattu, S., Propheter, D.C., Kuang, Z.,
Bel, S., Ruhn, K.A., Chara, A.L., Edwards, M.,
increased in the knockout mice. Since question of whether vitamin-A-dependent Zhang, C., Jo, J.H., et al. (2019). Resistin-like mole-
other known mouse skin AMPs seem to regulation of RELMs differs in sebocytes, cule a provides vitamin-A-dependent antimicrobial
be unaltered, RELM may contribute to adipocytes, and epithelial cells merits protection in the skin. Cell Host Microbe 25, this
issue, 777–788.
shaping the composition of mouse skin future studies.
microbiota by its direct bactericidal prop- Further, additional investigations will Latendorf, T., Gerstel, U., Wu, Z., Bartels, J.,
Becker, A., Tholey, A., and Schröder, J.M. (2019).
erties and thus protect the skin from bac- be required for a more comprehensive
Cationic intrinsically disordered antimicrobial pep-
terial infection. understanding of the range of microor- tides (CIDAMPs) represent a new paradigm of
Dietary vitamin A deficiency causes a ganisms that are targeted by mouse innate defense with a potential for novel anti-infec-
tives. Sci. Rep. 9, 3331.
marked susceptibility to infection and RELMa and human resistin and to identify
inflammation; as such, synthetic retinoids which bacterial species (in addition to O’Neill, A.M., and Gallo, R.L. (2018). Host-micro-
biome interactions and recent progress into under-
are widely used for treatment of inflam- S. aureus) can trigger RELMa and RETN
standing the biology of acne vulgaris. Microbiome
matory skin diseases. To delineate expression. 6, 177.
whether RELM family members are regu- An important remaining question is
Propheter, D.C., Chara, A.L., Harris, T.A., Ruhn,
lated by vitamin A, Harris et al. (2019) how skin bacteria trigger RELM expres- K.A., and Hooper, L.V. (2017). Resistin-like mole-
analyzed the human resistin promoter for sion in skin. The finding that the vitamin cule b is a bactericidal protein that promotes
retinoic acid response elements (RARE) A derivative retinol drives resistin expres- spatial segregation of the microbiota and the
colonic epithelium. Proc. Natl. Acad. Sci. USA
and found binding to several predicted sion through RAR(s) suggests that skin 114, 11027–11033.
RAREs. Consistently, the authors show bacteria could similarly regulate retinol
Schauber, J., Dorschner, R.A., Yamasaki, K.,
that retinol, a pro-form of vitamin A, or retinoic acid levels in keratinocytes Brouha, B., and Gallo, R.L. (2006). Control of the
induces the resistin gene in human and sebocytes and thus promote RAR- innate epithelial antimicrobial response is cell-
sebocytes synergistically with IL1b, which dependent transcription of RELM-encod- type specific and dependent on relevant microen-
vironmental stimuli. Immunology 118, 509–519.
suggests that resistin protein may be ing genes.
generated in sebaceous glands upon This study suggests that fat-soluble Zeeuwen, P.L., Boekhorst, J., van den Bogaard,
E.H., de Koning, H.D., van de Kerkhof, P.M.,
inflammation. vitamins appear to be of particular impor- Saulnier, D.M., van Swam, I.I., van Hijum, S.A.,
Harris et al. (2019) then explored tance for the expression of lipophilic skin Kleerebezem, M., Schalkwijk, J., and Timmerman,
whether mouse RELMa expression is antimicrobial proteins, which is possibly H.M. (2012). Microbiome dynamics of human
epidermis following skin barrier disruption.
similarly dependent on vitamin A. Mice of relevance to defend bacteria, like Genome Biol. 13, R101.
fed with a vitamin-A-deficient diet have C. acnes, residing in a lipid-rich environ-
Zlotogorski, A. (1987). Distribution of skin surface
less abundant RELMa transcripts and ment. While RELMa is a unique example pH on the forehead and cheek of adults. Arch.
RELMa protein levels in the skin —in of an antimicrobial protein regulated by Dermatol. Res. 279, 398–401.

770 Cell Host & Microbe 25, June 12, 2019

You might also like