Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

Epidermal Barriers Ken Natsuga Cold Spring Harb Perspect Med 2014; doi: 10.1101/cshperspect.a018218 Subject Collection

The Skin and Its Diseases

The Dermal Papilla: An Instructive Niche for Epithelial Stem and Progenitor Cells in Development and Regeneration of the Hair Follicle Bruce A. Morgan Immunology and Skin in Health and Disease Jillian M. Richmond and John E. Harris Desmosomes: Regulators of Cellular Signaling and Adhesion in Epidermal Health and Disease Jodi L. Johnson, Nicole A. Najor and Kathleen J. Green Markers of Epidermal Stem Cell Subpopulations in Adult Mammalian Skin Kai Kretzschmar and Fiona M. Watt Melanoma: Clinical Features and Genomic Insights Elena B. Hawryluk and Hensin Tsao Psoriasis Paola Di Meglio, Federica Villanova and Frank O. Nestle Cell Therapy in Dermatology Gabriela Petrof, Alya Abdul-Wahab and John A. McGrath Melanocytes and Their Diseases Yuji Yamaguchi and Vincent J. Hearing

Advanced Treatment for Basal Cell Carcinomas Scott X. Atwood, Ramon J. Whitson and Anthony E. Oro Epidermal Polarity Genes in Health and Disease Frederik Tellkamp, Susanne Vorhagen and Carien M. Niessen Induced Pluripotent Stem Cells in Dermatology: Potentials, Advances, and Limitations Ganna Bilousova and Dennis R. Roop The Genetics of Human Skin Disease Gina M. DeStefano and Angela M. Christiano Modeling Cutaneous Squamous Carcinoma Development in the Mouse Phillips Y. Huang and Allan Balmain p53/p63/p73 in the Epidermis in Health and Disease Vladimir A. Botchkarev and Elsa R. Flores Diversification and Specialization of Touch Receptors in Skin David M. Owens and Ellen A. Lumpkin Long Noncoding RNA: Significance and Potential in Skin Biology Derrick C. Wan and Kevin C. Wang

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2014 Cold Spring Harbor Laboratory Press; all rights reserved

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

Epidermal Barriers Ken Natsuga Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan Correspondence: [email protected]

www.perspectivesinmedicine.org

The epidermis functions as a physical barrier to the external environment and works to prevent loss of water from the skin. Numerous factors have been implicated in the formation of epidermal barriers, such as cornified envelopes, corneocytes, lipids, junctional proteins, proteases, protease inhibitors, antimicrobial peptides, and transcription factors. This review illustrates human diseases (ichthyoses) and animal models in which the epidermal barrier is disrupted or dysfunctional at steady state owing to ablation of one or more of the above factors. These diseases and animal models help us to understand the complicated mechanisms of epidermal barrier formation and give further insights on epidermal development.

he skin is the largest organ in the body and is constantly confronted with external stimuli, such as microorganisms, chemicals, heat, and water. Ever since vertebrates started their terrestrial life, skin has had the ability to retain water and to prevent liquids from penetrating into the internal organs. The epidermal barrier is indispensable to the maintenance of organisms; many examples in experimental animals and humans have shown that severe defects in epidermal barrier formation lead to an early demise. A great number of factors play roles in barrier development, including structural and junctional proteins, lipids, and transcription factors. This makes it difficult to determine which factor or factors are affecting barrier function through their impairment. It is noteworthy that barrier development is one aspect of the development of the epidermis itself. The ichthyoses are a heterogeneous group of disorders characterized by thick, scaly skin (Oji et al. 2010; Schmuth et al. 2013). “Icthy”

T

comes from the Greek word for fish. Congenital ichthyosis patients and animal models of these conditions generally show epidermal barrier defects caused by mutations in the genes encoding epidermal barrier components or key regulators. The thick, scaly skin of ichthyoses is thought to result from the skin compensating for barrier dysfunction (Segre 2006). This review presents some ichthyosis diseases and animal models with skin barrier dysfunction to illuminate the factors that are relevant to epidermal barrier formation. ASSESSMENT OF EPIDERMAL BARRIER FUNCTION

Various methods have been used to evaluate epidermal barrier function in animal models and humans. As is true for other experimental procedures, each method has advantages and disadvantages (Indra and Leid 2011). In any case, researchers must recognize two central aspects

Editors: Anthony E. Oro and Fiona M. Watt Additional Perspectives on The Skin and Its Diseases available at www.perspectivesinmedicine.org Copyright # 2014 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a018218 Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

1

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

K. Natsuga

of barrier function: “outside-in” barrier and “inside-out” barrier (Elias et al. 2008a; Proksch et al. 2008). “Outside-in” denotes the epidermal function of hindering the infiltration of external materials through the epidermis to the area inside. “Inside-out” denotes the ability of the epidermis to prevent the leakage of water and electrolytes from inside. As many of the epidermal components are involved in both outsidein and inside-out barrier function, it is not surprising to find many experimental animals with impairment of both barrier functions. Transepidermal water loss (TEWL) is a simple measure of the quantity of water that passes from inside the body through the epidermis to the atmosphere by evaporation and diffusion (Fluhr et al. 2006; Indra and Leid 2011). Skin barrier impairment presents as increased TEWL. The method of taking TEWL as a proxy for barrier function has been used for more than three decades and can be applied not only to animal skin but also to human skin (Elias and Brown 1978; Hammarlund and Sedin 1979; Wilson and Maibach 1980; Aszterbaum et al. 1992). However, when a probe is used for TEWL measurement, the score can vary depending on the probe site (Chilcott and Farrar 2000). In addition, the ambient temperature and humidity greatly affect TEWL. For these reasons, researchers must be cautious in interpreting the results of TEWL, especially for human patients. TEWL reflects only inside-out barrier function and not outside-in barrier function. Lanthanum tracer assay is another way of evaluating inside-out barrier function (Elias and Friend 1975; Elias and Brown 1978). In this assay, lanthanum nitrate is injected intracutaneously into experimental animals and specimens are later observed by electron microscopy. Lanthanum tracer percolates outward to the stratum granulosum (SG) and stops at tight junctions (TJs). The tracer does not reach the stratum corneum (SC) in wild-type skin (Elias and Brown 1978). In contrast, certain barrier impairments, including tight junction abnormalities, allow the tracer to reach the SC via intercellular spaces. Similar to lanthanum tracer assay, the surface biotinylation assay was developed. In this 2

assay, biotin instead of lanthanum is injected into animal skin (Merzdorf et al. 1998; Furuse et al. 2002). Specimens are incubated with avidin conjugated with fluorescein and visualized by fluorescent microscopy. Dye penetration assay is the gold standard for evaluating outside-in epidermal barrier function. In this method, animal fetuses or neonates are simply soaked in a dye such as X-Gal, Lucifer Yellow, toluidine blue, or hematoxylin (Hardman et al. 1998; Indra and Leid 2011). Barrier acquisition presents as non-dye-stained area. The dye penetration assay clearly shows E16.5 as the time point of barrier formation in mice (Hardman et al. 1998). Along with this, the dye penetration assay gives locational information on skin barrier development. There are other ways of evaluating barrier function through physical and chemical challenges to the skin. These include tape stripping and the application of acetone or detergent. Generally these methods are used when experimental animals have only subtle barrier defects or do not show any barrier dysfunction at steady state. When tape stripping or chemical application is used, barrier recovery rates are calculated (Ghadially et al. 1996). Except where otherwise noted, this review addresses experimental animals or human diseases that lead to physiological barrier dysfunction. COMPONENTS CONTRIBUTING TO EPIDERMAL BARRIER FORMATION

As described earlier, there are many factors involved in epidermal barrier function. As it is almost impossible to cover all of the factors, this review focuses on the following components: (1) cornified envelope and corneocytes, (2) TJs and other junctional proteins, (3) lipids, (4) proteases and protease inhibitors, (5) antimicrobial peptides, and (6) transcription factors. Table 1 summarizes the selected mouse models with epidermal barrier defects. Cornified Envelope and Corneocytes

The formation of corneocytes and the cornified envelope (CE) is the final step of epidermal dif-

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

Epidermal Barriers

Table 1. Selected mouse models with physiological barrier defects Gene

Gene product

Cornified envelope, corneocytes Inv/Ppl/Evpl Involucrin/periplakin/ envoplakin Tgm1 Transglutaminase-1

Human counterpart

References

Null

-

Sevilla et al. 2007

Null

LI, CIE

Null Null Null Null

IV EI EI PC

Matsuki et al. 1998; Nakagawa et al. 2012 Kawasaki et al. 2012 Roth et al. 2012 Reichelt et al. 2001 Lessard and Coulombe 2012

Null Null Carboxy-terminal truncation Null-C

IHSC HLHS, ODDD -

Furuse et al. 2002 Chidgey et al. 2001 Maass et al. 2004

Fatty acid transport protein-4

Null, Null-C

IPS

Abca12

ABC transporter A12

Null

HI, LI, CIE

Gba

b-Glucocerebrosidase

Null

Ugcg

UDP-glucose ceramide glucosyltransferase Ceramide synthase-3 Elongation of very long chain fatty acids-like-4 Elongation of very long chain fatty acids-like-1 Lipoxygenase-3 12(R)-lipoxygenase

Null-C

Gaucher syndrome type II -

Herrmann et al. 2003; Moulson et al. 2003; Herrmann et al. 2005 Smyth et al. 2008; Yanagi et al. 2008; Zuo et al. 2008 Holleran et al. 1994

Null Null

ARCI ISQMR

Jennemann et al. 2012 Vasireddy et al. 2007

Null

-

Sassa et al. 2013

Null Null

CIE CIE

Null

DCS

Krieg et al. 2013 Epp et al. 2007; Moran et al. 2007 Radner et al. 2010

Proteases and protease inhibitors Spink5 LECTI (lymphoepithelial Kazal-type inhibitor 5)

Null

NS

St14 Prss8 Casp14

Null Null Null

IHS -

Flg Krt1 Krt10 Krt16

Filaggrin Keratin-1 Keratin-10 Keratin-16

Junctional proteins Cldn1 Claudin-1 Dsc1 Desmocollin-1 Cx43 Connexin-43 Cdh1 Lipids Fatp4 www.perspectivesinmedicine.org

Mutation

Cers3 Elovl4 Elovl1 Aloxe3 Aloxe12

E-cadherin

Cgi58 (Abhd5) Comparative gene identification-58 (abhydrolase domaincontaining-5)

Matriptase Prostasin Caspase-14

Tunggal et al. 2005

Amen et al. 2013

Yang et al. 2004; Descargues et al. 2005; Hewett et al. 2005 List et al. 2002 Leyvraz et al. 2005 Denecker et al. 2007 Continued

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

3

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

K. Natsuga

Table 1. Continued Gene

Gene product

Mutation

Antimicrobial peptides Cramp Cathelicidin-related antimicrobial peptide

Human counterpart

References

Null

-

Nizet et al. 2001

Transcription factors Klf4 Krippel-like factor-4 Gata3 Gata-binding protein-3

Null Null-C

-

Taf10

Null-C

-

Segre et al. 1999 de Guzman Strong et al. 2006 Indra et al. 2005

Null Null-C

-

Null-C

-

Grhl3 Arnt Cebpa/Cebpb

Transcription initiation factor TFIID subunit 10 Grainy head-like-3 Aryl hydrocarbon receptor nuclear translocator CCAAT/enhancer binding protein (C/EBP), a and b

Ting et al. 2005 Takagi et al. 2003; Geng et al. 2006 Lopez et al. 2009

www.perspectivesinmedicine.org

ARCI, autosomal recessive congenital ichthyosis; CIE, congenital ichthyosiform erythroderma; DCS, Dorfman-Chanarin syndrome; EI, epidermolytic ichthyosis; HI, harlequin ichthyosis; HLHS, hypoplastic left heart syndrome; IHS, ichthyosis hypotrichosis syndrome; IHSC, ichthyosis-hypotrichosis-sclerosing cholangitis; IPS, ichthyosis prematurity syndrome; ISQMR, ichthyosis, spastic quadriplegia, and mental retardation; IV, ichthyosis vulgaris; LI, lamellar ichthyosis; NS, Netherton syndrome; Null-C, conditional null; ODDD, oculodentodigital dysplasia.

ferentiation. The CE is formed through crosslinking of insoluble membranous proteins by transglutaminase (Candi et al. 2005). Major components of the CE include involucrin, periplakin, envoplakin, and loricrin (Rice and Green 1977, 1979; Mehrel et al. 1990; Ruhrberg et al. 1996; Ruhrberg et al. 1997). Unexpectedly, mice with loss of each of these proteins did not show overt phenotypic changes (Maatta et al. 2001; Aho et al. 2004; Djalilian et al. 2006), other than loricrin-deficient mice, which developed transient ichthyosiform skin in the neonatal period that disappeared at several days after birth (Koch et al. 2000). In line with the results of the knockout (KO) mice, there have been no reports on human mutations in involucrin, periplakin, or envoplakin genes, although single-base-pair-insertion mutations in the gene encoding loricrin are known to cause loricrin keratoderma in humans, which is characterized by erythrokeratoderma and digital constriction bands ( pseudo-ainhum) (Ishida-Yamamoto 2003). A further transgenic approach to delineating the role of CE components in the epidermal barrier failed to find apparent phenotypic changes in double-KO mice for invo4

lucrin/periplakin or periplakin/envoplakin or envoplakin/involucrin (Sevilla et al. 2007). Barrier defects were observed only when the three proteins (involucrin, periplakin, and envoplakin) were ablated at the same time (Sevilla et al. 2007). The triple-KO mice were viable but developed dry, ichthyotic skin, reflecting delayed permeability barrier acquisition as shown by dye penetration assay. The impaired epidermal barrier of the triple-KO mice might be accounted by for by (1) the fragility of the cornified envelope, (2) reduced epidermal lipid content, and (3) aberrant filaggrin processing. Intriguingly, the triple-KO mice were characterized by dermal inflammatory cell infiltrates, mostly composed of CD4þ T cells, and loss of epidermal dendritic T cells (Sevilla et al. 2007). This inflammatory phenotype in the barrierdefective mice might be consistent with the fact that atopic dermatitis can develop in people with filaggrin mutations, which is discussed below. In contrast to KO mice for components of the CE, the ablation of transglutaminase, the enzyme crosslinking CE proteins, is detrimental. Mutations in TGM1, which encodes trans-

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

Epidermal Barriers

glutminase-1, lead to lamellar ichthyosis (LI) or congenital ichthyosiform erythroderma (CIE) in humans (Huber et al. 1995; Russell et al. 1995; Laiho et al. 1997). Tgm1-null mice were shown to have severe barrier defects and neonatal lethality (Matsuki et al. 1998; Kuramoto et al. 2002). These mice had erythrodermic skin with defective CE assembly, as well as disintegrated lipid bilayers between CEs (Matsuki et al. 1998; Kuramoto et al. 2002). The same research group recently generated knock-in mice with a Tgm1 R142C mutation that is equivalent to TGM1 R143C in human LI or CIE patients (Nakagawa et al. 2012). Those mice showed severe barrier defects and neonatal lethality (Nakagawa et al. 2012). The results of Tgm1 transgenic mice underscore the indispensable role of transglutaminase in epidermal barrier formation. Filaggrin is known to bundle keratin filaments and to be incorporated into corneocytes (Steinert et al. 1981). Loss of filaggrin expression has been observed in the epidermal keratinocytes of ichthyosis vulgaris (IV) patients (Sybert et al. 1985; Nirunsuksiri et al. 1998). However, the many repeats in its sequence hindered the sequencing of FLG, which encodes filaggrin. The McLean group succeeded in sequencing FLG and discovered loss-of-function mutations in IV patients (Smith et al. 2006). As it was known that IV is a risk factor for atopic dermatitis (AD), the same group sequenced FLG in AD patients and found FLG mutations (Palmer et al. 2006). Since then, many reports have addressed the frequency of FLG mutations in AD patients (Rodriguez et al. 2009; McAleer and Irvine 2013). Spontaneous mutant mice called “flaky-tail mice” have been known to show dry, flaky skin and to show abnormal filaggrin processing (Presland et al. 2000). Sequencing technology revealed that flaky-tail mice have a frameshift mutation in Flg (Fallon et al. 2009). Since then, the flaky-tail mouse has been used as an AD model for its spontaneous dermatitis phenotype (Oyoshi et al. 2009; Scharschmidt et al. 2009b; Moniaga et al. 2010). Unexpectedly, however, Flg 2/2 mice show no overt phenotypic changes, including spontaneous dermatitis (Kawasaki et al. 2012). Skin barrier defects in the steady state

were not confirmed in Flg 2/2 mice except in the outside-in barrier against liposome, whereas enhanced irritant and allergic contact dermatitis responses were observed in Flg 2/2 mice on hapten application (Kawasaki et al. 2012). Recently, pyrosequencing of flaky-tail mice revealed another homozygous nonsense mutation in the gene encoding mattrin (Sasaki et al. 2013; Saunders et al. 2013). This mutation was reported to be involved in the spontaneous dermatitis phenotype in the mice (Sasaki et al. 2013; Saunders et al. 2013). A common missense variant in the orghologous human gene, MATT, was identified in AD patients (Saunders et al. 2013). Taken together, it may be that the filaggrin mutations in AD patients do not cause spontaneous dermatitis but, rather, contribute to increased percutaneous immune response caused by penetration of antigens, haptens, and irritants into the deeper SC. In contrast, it may be that the mattrin mutations in AD patients induce spontaneous dermatitis as well as elevated immune response on percutaneous challenge. The implications of human and murine experiments on CE and filaggrin are as follows: (1) loss of one CE component or filaggrin can be compensated by other proteins and does not lead to severe barrier defects, (2) the ablation of several CE components (exemplified in Inv/ Ppl/Evpl triple-KO mice) causes barrier defects, (3) in contrast, transglutaminase, which crosslinks CE proteins, is essential for epidermal barrier formation, and (4) disturbance of the CE (as seen in Inv/Ppl/Evpl triple-KO mice and Tgm1-null mice) affects not only the CE but also the lipid contents in the SC. Keratins are the largest group of intermediate filaments and are expressed in epithelial cells (Schweizer et al. 2006). Keratins are classified into type I (acidic) and type II (basic to neutral). Type I and type II keratins form noncovalent heteropolymers. The specificity of keratin subtypes in each cell has been known (Schweizer et al. 2006). In the epidermis, basal keratinocytes express keratin5/keratin14 (K5/K14) heteropolymers, whereas the keratinocytes in the spinous and granular layers preferentially possess keratin1/keratin10 (K1/K10) (Moll et al. 1982; Nelson and Sun 1983; Roop et al.

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

5

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

K. Natsuga

1983). Dominant-negative mutations in KRT1 or KRT10 (encoding K1 or K10) cause epidermolytic ichthyosis in human, which is characterized by erythroderma and widespread blister formation at birth (Cheng et al. 1992; Rothnagel et al. 1992). Several mice with keratin modifications have been used to investigate keratin’s involvement in the epidermal barrier. Krt10null mice show no severe phenotypes and are viable, which is explained by the compensatory expression of K5/K14 in suprabasal layers (Reichelt et al. 2001). Dye penetration assay revealed a focal outside-in barrier defect in the forepaw pad skin of Krt10-null neonates. In contrast to Krt-10 null mice, Krt1-null mice show perinatal lethality (Roth et al. 2012). Krt1-null have elevated TEWL compared with the wild type, although the outside-in barrier in these mice is intact (Roth et al. 2012). The increased fragility of the CE might be responsible for barrier dysfunction in Krt1- or Krt10null mice (Roth et al. 2012). In contrast to the results for Krt1-null mice, Krt1/Krt10 doubleKO mice do not show altered inside-out barrier by biotin permeability assay (Wallace et al. 2012). The discrepancy between Krt1-null mice and Krt1/Krt10 double-KO mice might be accounted for by the assays used in each experiment. Dominant-negative mutations in the gene encoding keratin 16 (KRT16) cause pachyonychia congenita in humans, characterized by hypertrophic nail dystrophy and palmoplantar keratoderma (PPK) (McLean et al. 2011). Krt16null mice were reported to develop PPK (Lessard and Coulombe 2012). Intriguingly, the front paws of adult Krt16-null mice show a focal loss of outside-in barrier using dye-penetration assay corresponding to hyperkeratotic lesions, suggesting that hyperkeratosis is a compensatory process resulting from barrier dysfunction (Lessard and Coulombe 2012). The cause of focal barrier disruption is not clear, even though the authors showed that there is reduced expression of filaggrin in the affected area of Krt16-null mice (Lessard and Coulombe 2012), which may not necessarily explain barrier disturbance as discussed earlier in Flg-null mice (Kawasaki et al. 2012). 6

Tight Junction and Other Junctional Proteins

The borders of epithelial cells are occupied by several junctional devices, including tight junctions (TJs), desmosomes, gap junctions (GJs), and adherent junctions (AJs). The role of TJs in the epidermal barrier is highlighted in claudin1-deficient mice (Furuse et al. 2002). Claudin-1 has been identified as a TJ protein (Furuse et al. 1998), and copolymers of heterogeneous claudins make TJ strands (Furuse et al. 1999). Claudin-1-deficient mice show neonatal lethality (Furuse et al. 2002). As expected, disruption of normal TJs results in inside-out barrier defects in claudin-1-deficient mice as revealed by increased TEWL and positive biotin permeability assay (Furuse et al. 2002). Furthermore, outsidein barrier dysfunction in these mice is observed; it possibly results from altered intercellular lipid composition (Sugawara et al. 2013). The cause of the altered lipid composition in these mice is not clear. The fact that claudin-1 gene mutations lead to human ichthyosis-hypotrichosissclerosing cholangitis (IHSC syndrome) corroborates the importance of this molecule in the epidermal barrier (Hadj-Rabia et al. 2004). Surprisingly, overexpression of claudin-6 under involucrin promoter in mice (INV-Cldn6 mice) produces neonatal lethality with outside-in and inside-out barrier defects, a phenotype similar to that of claudin-1-deficient mice (Turksen and Troy 2002). These complicated results indicate that differential expression of, or the balance of, claudin proteins might be relevant in the tightness of the TJ barrier (Kubo et al. 2012). Desmosomes are another junctional device involved in cell adhesion, and disturbed desmosomal components have been shown to cause skin, hair, and heart abnormalities in humans (Brooke et al. 2012; Petrof et al. 2012). Desmocollins and desmogleins are the desmosomal cadherins that extend into the extracellular area and adhere to neighboring cells (Delva et al. 2009). Desmocollin1-deficient mice show flaky skin and impaired outside-in/inside-out epidermal barrier (Chidgey et al. 2001). However, this barrier formation may not be due directly to desmocollin-1 in the epidermal barrier; it could simply be a consequence of the acan-

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

Epidermal Barriers

tholytic skin in these mice (Chidgey et al. 2001). Mutations in the gene encoding desmocollin-1 in humans have not been reported. GJs mediate the exchange of ions, metabolites, and second messengers between neighboring cells (Maeda and Tsukihara 2011). Mutations in the gene encoding connexin-43 (Cx43), a major GJ protein, are known to be responsible for oculodentodigital dysplasia (Paznekas et al. 2003) and hypoplastic left heart syndrome (Dasgupta et al. 2001). Mice in which the Cterminus of Cx43 was ablated died shortly after birth (Maass et al. 2004). Unexpectedly, neonates of these mice show scaly skin and constriction bands around the tails as well as outside-in barrier defects shown by toluidine blue dye penetration assay (Maass et al. 2004). The cause of the epidermal barrier impairment is unclear, although these mice show aberrant filaggrin processing. E-cadherin is one of the adhesive components of AJs and is important for tissue morphogenesis and polarity (Perez-Moreno et al. 2003; Yonemura 2011). Keratinocyte-specific E-cadherin-deficient mice show intact outsidein barrier function (Tunggal et al. 2005). However, these mice have impaired inside-out barrier (shown by TEWL measurement and biotin penetration assay) because of aberrant tight junction proteins possibly resulting from the loss or reduced presence of activated atypical PKC (Tunggal et al. 2005). Lipids

Corneocytes and CEs are surrounded by intercellular lipids (ceramides, cholesterols, and free fatty acids) that function as a potent epidermal barrier. Lipids are stored in lamellar bodies in stratum granulosum (SG) keratinocytes and are secreted at the SC – SG interface through cornification steps. Perturbations in lipid metabolism and transport lead to severe barrier dysfunction in humans and animals. Several review papers have recently addressed lipid metabolism disorders (Elias et al. 2008b, 2011, 2012; Feingold and Jiang 2011; Rizzo 2011). In addition to discussing the major players in lipid metabolism and diseases caused by dysfunctional metabolic

pathways, this section addresses recent advances in our understanding of the roles of enzymes in facilitating very-long-chain fatty acid elongation and in synthesizing ceramide on the epidermal barrier. Free fatty acids are transported across cell membranes in several ways (Khnykin et al. 2011). FATP4, encoding fatty acid transport protein 4, was reported to be the causative gene of ichthyosis prematurity syndrome in humans (Klar et al. 2009). The “wrinkle-free” mouse is a natural mutant of Fatp4 that is characterized by neonatal lethality, barrier defects and absence of wrinkles (Moulson et al. 2003). Fatp4-null mice were also generated and confirmed to have phenotypes similar to those of wrinkle-free mice, including outside-in and inside-out epidermal barrier dysfunction (Herrmann et al. 2003). Transgenic mice in which Fatp4 deficiency is inducible in the epidermis show impaired epidermal barrier (Herrmann et al. 2005). In contrast, keratinocyte-specific overexpression of Fatp4 restores the skin phenotype of wrinklefree mice (Moulson et al. 2007). As the proportion of very-long-chain fatty acids (VLFA; C26) in epidermal ceramides is significantly decreased in wrinkle-free mice and Fatp4-null mice and the reduced VLFA in these mice is rescued through keratinocyte-specific overexpression of Fatp4, the fatty acid chain length of skin lipids is thought to be regulated by Fatp4 activity to maintain normal skin integrity (Herrmann et al. 2003; Moulson et al. 2007). ABCA12 is a member of the superfamily of ATP-binding cassette (ABC) transporters (Annilo et al. 2002). ABCA12 (ABC transporter A12) has been characterized as a key protein in keratinocyte lipid transport (Akiyama 2011). ABCA12 is expressed in lamellar bodies and is thought to mediate lipid transport into extracellular spaces via lamellar bodies to establish the lipid barrier in cornified layers (Akiyama et al. 2005). ABCA12 mutations have been reported in lamellar ichthyosis, congenital ichthyosiform erythroderma, and harlequin ichthyosis in humans (Lefevre et al. 2003; Akiyama et al. 2005; Kelsell et al. 2005; Natsuga et al. 2007; Akiyama 2010). Abca12-deficient mice show outside-in and inside-out barrier defects and

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

7

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

K. Natsuga

neonatal lethality, which reproduces the phenotype of harlequin ichthyosis, the most severe congenital ichthyosis subtype, in humans (Smyth et al. 2008; Yanagi et al. 2008; Zuo et al. 2008). Abca12-deficient mice have defective lipid efflux represented as decreased v-O-acylceramides as well as increased accumulation of glucosylceramides (Smyth et al. 2008; Zuo et al. 2008). This defective lipid efflux leads to malformation of the intercellular lipid layers, which mostly accounts for the severe barrier defects in Abca-deficient skin, although aberrant filaggrin processing and fragile CE are also observed in Abca12-deficient mice (Smyth et al. 2008; Yanagi et al. 2008; Zuo et al. 2008; Akiyama 2011). The hydrolysis of glucosylceramides to ceramides is catalyzed by b-glucocerebrosidase (Beutler 1992). Mutations in the human bglucocerebrosidase gene cause Gaucher’s disease type II, which is characterized by cranial nerve and brain abnormalities and hepatosplenomegaly (Tsuji et al. 1987). Targeted disruption of murine b-glucocerebrosidase leads to outside-in and inside-out barrier dysfunction, highlighting the importance of the content of glucosylceramides and ceramides in the intercellular lipid layers (Holleran et al. 1994). This fact is corroborated by the disrupted water permeability barrier seen in tamoxifeninduced deletion of glucosylceramide-synthesizing enzyme UDP-glucose ceramide glucosyltransferase (UGCG) in keratinocytes (Amen et al. 2013). The ceramide synthases (CerS) are a group of enzymes that acylate dihydrosphingosine to produce dihydroceramide (Mizutani et al. 2009). Among CerS subtypes, CerS3 is involved in the production of ceramides with VLFA (C26) (Jennemann et al. 2012). Ablation of CerS3 causes neonatal lethality and severe skin barrier defects (both outside-in and insideout), whereas CerS2-deficiency produces phenotypically normal mice (Jennemann et al. 2012). These results indicate the essential role of ceramides with very-long-chain acyl moieties in epidermal barrier function. It is not surprising that mutations in CERS3 were recently reported to be responsible for human autosomal 8

recessive congenital ichthyosis (Eckl et al. 2013; Radner et al. 2013). As observed in CerS3-deficient mice, VLFA have been implicated in skin development and barrier formation (Kihara 2012). Fatty acid elongation steps involve the production of 3ketoacyl-CoA from acyl-CoA and malonylCoA (Jakobsson et al. 2006). This reaction is catalyzed by fatty acids elongases (Elovl1-7 in mammals) (Kihara 2012). Among Elovl family members, Elovl1 and Elovl4 have been shown to be involved in the elongation of VLFA (Guillou et al. 2010; Ohno et al. 2010). Elovl4-deficient mice show neonatal lethality as well as severe barrier dysfunction (outside-in), which might be explained by depletion of VLFA (C28) in both the ceramide and free fatty acid fractions in the epidermis and by loss of v-O-acylceramides, which are the major components of the hydrophobic lipid bilayers between corneocytes (Vasireddy et al. 2007). Mutations in ELOVL4 were reported in patients suffering from ichthyosis with intellectual disability and spastic quadriplegia (Aldahmesh et al. 2011), the symptoms of which are very similar to those of SjogrenLarsson syndrome (De Laurenzi et al. 1996). Moreover, Elovl1-deficient mice also show neonatal lethality and outside-in/inside-out epidermal barrier defects with depletion of VLFA (C26), and they also show increases in C24 fatty acids (Sassa et al. 2013). Interestingly, Elovl1 was shown to coordinate with CerS3 to produce VLFA (C26) (Sassa et al. 2013). Human ELOVL1 mutations have not been reported. Mutations in ALOXE3 (encoding lipoxygenase-3) and ALOX12B (encoding 12(R)-lipoxygenase) have been detected in CIE patients through linkage analysis (Jobard et al. 2002). Lipoxygenase-3 and 12(R)-lipoxygenase have been implicated in the catalysis of oxygenation of linoleate followed by the esterase-catalyzed hydrolysis of the oxidized linoleate, eventually leading to covalent coupling of the lipids to the cross-linked proteins of the CE (Zheng et al. 2011). Aloxe3-deficient and Alox12b-deficient mice show severe barrier defects (both outsidein and inside-out) accompanied by altered lipid composition (Epp et al. 2007; Moran et al. 2007; Krieg et al. 2013).

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

Epidermal Barriers

Dorfman-Chanarin syndrome is a rare autosomal recessive disorder characterized by congenital ichthyosiform erythroderma and intracellular accumulation of triacylglycerol droplets in many tissues (Dorfman et al. 1974; Chanarin et al. 1975). Linkage analysis identified CGI-58 (also called ABHD5) as a causal gene for Dorfman-Chanarin syndrome (Lefevre et al. 2001). CGI-58 recombinant protein was shown to show acyltransferase activity on lysophosphatidic acid (Ghosh et al. 2008). Cgi-58-deficientmice have severe outside-in barrier defects as well as triacylglycerol accumulation and depletion of acylceramide (Radner et al. 2010).

www.perspectivesinmedicine.org

Proteases and Protease Inhibitors

Serine proteases have been implicated in the process of keratinocyte differentiation and desquamation (Ovaere et al. 2009). The main roles of serine proteases are (1) the processing of profilaggrin into filaggrin monomer and natural moisturizing factor, and (2) the degradation of junctional proteins (desmosomes and corneodesmosomes) during desquamation steps. The cascades and main players in the proteases and inhibitors have been discussed in a recent review paper (Ovaere et al. 2009). The first breakthrough that gained attention was the discovery of SPINK5 (serine protease inhibitor Kazal-type 5) mutations in Netherton syndrome patients (Chavanas et al. 2000; Hovnanian 2013). Netherton syndrome is a rare genodermatosis that is characterized by trichorrhexis nodosa, congenital ichthyosiform erythroderma, and atopic diathesis (Hovnanian 2013). SPINK5 encodes LECTI (lymphoepithelial Kazal-type inhibitor 5) expressed in stratified epithelia (Magert et al. 1999; Bitoun et al. 2003). Spink5-deficient mice show outside-in and inside-out epidermal barrier dysfunction as well as the following: (1) increase in the proteolytic processing of profilaggrin into filaggrin monomers (Descargues et al. 2005; Hewett et al. 2005), (2) abnormal degradation of desmoglein-1 and desmoplakin (Descargues et al. 2005), and (3) premature proteolysis of corneodesmosin (Yang et al. 2004; Descargues et al. 2005). Kallikreins are a family of serine prote-

ases, and many kallikrein members are expressed in human epidermis (Lundwall and Brattsand 2008). As LEKTI is a potent inhibitor of multiple serine proteases, Spink5-deficient epidermis leads to kallikrein 5 (KLK5) and KLK7 hyperactivity, accounting for the degradation of desmosomal proteins (Descargues et al. 2005). The fact that transgenic mice in which KLK7 is overexpressed have increased TEWL confirms the role of LEKTI in epidermal barrier formation (Ny and Egelrud 2004). Furthermore, another target of LEKTI was found to be elastase-2 (Ela2) through mass spectrometry analysis (Bonnart et al. 2010). Overexpression of Ela2 under the control of involucrin promoter produces both outside-in and inside-out barrier dysfunction in mice (Bonnart et al. 2010). Matriptase and prostatin are serine proteases that are known to be involved in epidermal terminal differentiation (Netzel-Arnett et al. 2006; Ovaere et al. 2009). Matriptase is thought to activate prostatin to mediate subsequent processes that lead to filaggrin monomer formation and the generation of natural moisturizing factors (NMFs) (Ovaere et al. 2009; Miller and List 2013). Mutations in ST14 encoding matriptase have been reported in patients with autosomal recessive congenital ichthyosis with hypotrichosis (Basel-Vanagaite et al. 2007; Alef et al. 2009). Matriptase-deficient mice show outside-in and inside-out barrier defects accompanied by hypoplastic lamellar bodies and loss of filaggrin monomer (List et al. 2002, 2003). Furthermore, matriptase activates prokallikreins ( proKLK5 and proKLK7), whereas LEKTI targets neither matriptase nor prostatin (Sales et al. 2010). In accordance with this fact, the epidermal barrier defects in Spink5-deficient mice are compensated for by abrasion of matriptase (Sales et al. 2010). Similar to matriptase-deficient mice, prostasin-deficient mice have both outside-in and inside-out barrier defects (Leyvraz et al. 2005; Netzel-Arnett et al. 2006) that are consistent with the fact that frizzy mouse and hairless rat, natural mutants of Prss8 (encoding prostasin), show abnormal barrier function (Spacek et al. 2010; Frateschi et al. 2012), although PRSS8 mutations in human patients have not

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

9

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

K. Natsuga

been reported. It is noteworthy that overexpression of prostasin in basal keratinocytes also leads to elevated TEWL in the transgenic mice (Frateschi et al. 2011). These facts indicate that a balance between proteases and protease inhibitors is crucial for epidermal barrier formation. Caspase-14, an aspartate-specific protease, has recently been implicated in filaggrin degradation to NMFs (Hoste et al. 2011). TEWL in caspase-14 deficient mice is elevated compared with controls, although the outside-in barrier is intact (Denecker et al. 2007). Caspase-14 gene mutations have not been documented in human ichthyosis patients.

www.perspectivesinmedicine.org

Antimicrobial Peptides

Skin faces external pathogens as a surface tissue; thus, it expresses various kinds of antimicrobial peptides (AMPs) constitutively or in response to physical stresses. Excellent review papers have been published on AMPs (Lai and Gallo 2009; Gallo and Hooper 2012). This section briefly discusses key findings on AMPs and skin barrier function. Many proteins have been listed as having antimicrobial properties in skin, including b defensins, cathelicidin-related antimicrobial peptide (Cramp in mice, LL-37 in humans), S100A7 and S100A8 (Lai and Gallo 2009). Cramp was shown to protect skin from infection by invasive bacteria such as group A Streptococcus pyogens, as revealed by infection experiments on Cramp-deficient mice (Nizet et al. 2001). Cramp is stored in lamellar bodies in granular layer keratinocytes and secreted at the SC – SG interface (Braff et al. 2005). Although Cramp-deficient mice show normal levels of TEWL, lanthanum penetration assay reveals inside-out barrier dysfunction of Cramp-deficient epidermis in electron microscopy (Aberg et al. 2008). This is explained by abnormal lamellar body formation and immature intercellular lipid layers in Cramp-deficient mice (Aberg et al. 2008). This is consistent with the relationship between Cramp expression pattern and barrier status. In a recent paper, techniques for impairing barrier function, including psychological stress, were reported to decrease 10

Cramp expression in the skin (Rodriguez-Martin et al. 2011). Transcription Factor

Although various kinds of enzymes, structural proteins and lipid metabolisms are involved in epidermal barrier formation, some transcription factors are known to regulate those players and eventually govern the development of the epidermal barrier. The first example is krippel-like factor 4 (Klf4). Klf4 is expressed in the epidermis, and Klf4-deficient mice were reported to have outside-in barrier defect and neonatal death (Segre et al. 1999). Microarray analysis of wild-type and Klf4-deficient mice identifies connexin 26 (Cx26) as a strongly upregulated gene in Klf4deficient epidermis. Klf4 binds to the proximal promoter of Cx26 and regulates its transcription level (Djalilian et al. 2006). Cx26 overexpression under involucrin promoter in mice produces severe barrier defects that mimic those of Klf4-deficient mice (Djalilian et al. 2006). In contrast, induced expression of Klf4 under keratin 5 promoter accelerates epidermal barrier formation in mice (Jaubert et al. 2003). These results indicate that Klf4 is one of the major transcription factors that decide the fate of epidermal barrier development in the fetus. Gata3 is a member of the GATA family of transcription factors in the epidermis (Kaufman et al. 2003). Agpat5, a lipid acyltransferase gene, is the direct target of Gata3 (de Guzman Strong et al. 2006). Keratinocyte-specific Gata3deficient mice show outside-in and inside-out barrier defects together with decreased v-Oacylceramides (de Guzman Strong et al. 2006). Taf10 is a component of the TFIID complex, which is essential for the initiation of transcription by RNA polymerase II (Mohan et al. 2003). Keratinocyte-specific ablation of Taf10 in mice reveals severe outside-in and inside-out barrier dysfunction, which might be attributable to the fact that Taf10 regulates the expression of Tgm1, Klf4, and Cldn1(Indra et al. 2005). Grainy head-like 3 (Grhl3) is another example of a transcription factor that is involved in epidermal barrier formation. Grhl3-deficient

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

Epidermal Barriers

mice have severe outside-in and inside-out barrier defects, which is corroborated by the fact that Grhl3 controls the expression of Tgm1 (Ting et al. 2005). Aryl hydrocarbon receptor nuclear translocator (Arnt) is a basic protein that belongs to the Period-Arnt-single-minded (PAS) family. Keratinocyte-specific Arnt-deficient mice show neonatal death and severe outside-in and inside-out barrier dysfunction (Takagi et al. 2003; Geng et al. 2006). Lipid composition is perturbed in these mice. Arnt regulates the expression of epidermal differentiation genes, although the direct target of Arnt has not been clarified (Robertson et al. 2012). CCAAT/enhancer binding protein (C/EBP) a and b are transcription factors expressed in the epidermis (Maytin and Habener 1998; Oh and Smart 1998). Whereas C/EBP-b-deficient mice had mild epidermal hyperplasia, epidermis-specific C/EBP-a-deficient mice showed no skin phenotypic abnormalities in steady state (Zhu et al. 1999; Loomis et al. 2007). Keratinocyte-specific ablation of both C/EBP-a and C/ EBP-b revealed hyperproliferation of basal kerationocytes and halted terminal differentiation (Lopez et al. 2009). These mice showed abnormal inside-out skin barrier function but normal outside-in barrier (Lopez et al. 2009). CONCLUDING REMARKS

This article has briefly reviewed skin-barrierdefective animal models and related human diseases. As seen in the section on lipids, disturbance of lipid metabolism is detrimental and leads to early demise in transgenic mice. Even when epidermal proteases or SC components are ablated, the alteration of lipid composition is observed. Not only do these results highlight the central role of lipids in the epidermal barrier, but they also show the complexity of barrier formation. Furthermore, the epidermal barrier is maintained by a fine balance among the major players; an imbalance of protease/protease inhibitors and overexpression/ablation of tight junction proteins leads to barrier dysfunction. Recent studies have indicated that the barrier is not simply a fortress keeping pathogens

out, but is an integrated interface composed of SCs, keratinocytes structural components and immune cells mediating antigen presentation (Kubo et al. 2012). This is exemplified in AD, which is characterized by epidermal barrier dysfunction and cutaneous sensitization. Nevertheless, the nature of AD remains unclear, because barrier defects allow the penetration of allergens leading to inflammation and inflammation can induce skin barrier dysfunction (Kubo et al. 2012). The key to solving this complicated problem might be in the observation that AD patients with high IgE levels have FLG mutations more frequently than AD patients with low IgE levels (Kabashima-Kubo et al. 2012). This suggests that primary barrier defects induce allergic reaction in a subclass of AD (extrinsic AD), whereas other AD patients have primary immunological abnormalities without barrier impairment (intrinsic AD). As many mutant mice have become available from library sources (Ayadi et al. 2012), we will be able to add many more genes to the list of players in epidermal barrier formation in the near future. Future tasks in research on epidermal barrier function are not limited to the identification of unknown barrier components. Skin infection and epidermal barrier dysfunction promise to rank among the hottest topics in the context of atopic dermatitis, in which staphylococcal infections are frequent (Kong et al. 2012). In fact, one barrier-defective mouse has been shown to have altered skin microbiota compared with controls (Scharschmidt et al. 2009a). This research field will be supported by the Human Microbiome Project (2012a,b). Another topic will be the relationship between epidermal barrier function and predisposition to skin cancer. Although there is controversy over the risk of skin cancer in atopic dermatitis patients, ichthyosis patients (especially with those Keratitis-Ichthyosis-Deafness (KID) syndrome, congenital ichthyosiform erythroderma/lamellar ichthyosis, or Netherton syndrome) might have increased risk of skin cancers, including squamous cell carcinoma (Natsuga et al. 2011). Carcinogenesis experiments on barrier-defective mice could provide

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

11

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

K. Natsuga

clues to understanding the pathomechanisms underlying skin cancer in inherited ichthyoses, given that the model mice do not show neonatal lethality. From the perspective of managing ichthyosis disorders, we have only conservative treatments, such as emollients. Once the mechanisms of epidermal barrier formation are further elucidated, novel and more sophisticated treatment options will be developed.

www.perspectivesinmedicine.org

REFERENCES Aberg KM, Man MQ, Gallo RL, Ganz T, Crumrine D, Brown BE, Choi EH, Kim DK, Schroder JM, Feingold KR, et al. 2008. Co-regulation and interdependence of the mammalian epidermal permeability and antimicrobial barriers. J Invest Dermatol 128: 917– 925. Aho S, Li K, Ryoo Y, McGee C, Ishida-Yamamoto A, Uitto J, Klement JF. 2004. Periplakin gene targeting reveals a constituent of the cornified cell envelope dispensable for normal mouse development. Mol Cell Biol 24: 6410– 6418. Akiyama M. 2010. ABCA12 mutations and autosomal recessive congenital ichthyosis: A review of genotype/phenotype correlations and of pathogenetic concepts. Hum Mutat 31: 1090– 1096. Akiyama M. 2011. The roles of ABCA12 in keratinocyte differentiation and lipid barrier formation in the epidermis. Dermatoendocrinol 3: 107– 112. Akiyama M, Sugiyama-Nakagiri Y, Sakai K, McMillan JR, Goto M, Arita K, Tsuji-Abe Y, Tabata N, Matsuoka K, Sasaki R, et al. 2005. Mutations in lipid transporter ABCA12 in harlequin ichthyosis and functional recovery by corrective gene transfer. J Clin Invest 115: 1777– 1784. Aldahmesh MA, Mohamed JY, Alkuraya HS, Verma IC, Puri RD, Alaiya AA, Rizzo WB, Alkuraya FS. 2011. Recessive mutations in ELOVL4 cause ichthyosis, intellectual disability, and spastic quadriplegia. Am J Hum Genet 89: 745–750. Alef T, Torres S, Hausser I, Metze D, Tursen U, Lestringant GG, Hennies HC. 2009. Ichthyosis, follicular atrophoderma, and hypotrichosis caused by mutations in ST14 is associated with impaired profilaggrin processing. J Invest Dermatol 129: 862 –869. Amen N, Mathow D, Rabionet M, Sandhoff R, Langbein L, Gretz N, Jackel C, Grone HJ, Jennemann R. 2013. Differentiation of epidermal keratinocytes is dependent on glucosylceramide:ceramide processing. Hum Mol Genet 22: 4164–4179. Annilo T, Shulenin S, Chen ZQ, Arnould I, Prades C, Lemoine C, Maintoux-Larois C, Devaud C, Dean M, Denefle P, et al. 2002. Identification and characterization of a novel ABCA subfamily member, ABCA12, located in the lamellar ichthyosis region on 2q34. Cytogenet Genome Res 98: 169 –176. Aszterbaum M, Menon GK, Feingold KR, Williams ML. 1992. Ontogeny of the epidermal barrier to water loss

12

in the rat: Correlation of function with stratum corneum structure and lipid content. Pediatr Res 31: 308 –317. Ayadi A, Birling MC, Bottomley J, Bussell J, Fuchs H, Fray M, Gailus-Durner V, Greenaway S, Houghton R, Karp N, et al. 2012. Mouse large-scale phenotyping initiatives: Overview of the European Mouse Disease Clinic (EUMODIC) and of the Wellcome Trust Sanger Institute Mouse Genetics Project. Mamm Genome 23: 600– 610. Basel-Vanagaite L, Attia R, Ishida-Yamamoto A, Rainshtein L, Ben Amitai D, Lurie R, Pasmanik-Chor M, Indelman M, Zvulunov A, Saban S, et al. 2007. Autosomal recessive ichthyosis with hypotrichosis caused by a mutation in ST14, encoding type II transmembrane serine protease matriptase. Am J Hum Genet 80: 467– 477. Beutler E. 1992. Gaucher disease: New molecular approaches to diagnosis and treatment. Science 256: 794 –799. Bitoun E, Micheloni A, Lamant L, Bonnart C, TartagliaPolcini A, Cobbold C, Al Saati T, Mariotti F, Mazereeuw-Hautier J, Boralevi F, et al. 2003. LEKTI proteolytic processing in human primary keratinocytes, tissue distribution and defective expression in Netherton syndrome. Hum Mol Genet 12: 2417– 2430. Bonnart C, Deraison C, Lacroix M, Uchida Y, Besson C, Robin A, Briot A, Gonthier M, Lamant L, Dubus P, et al. 2010. Elastase 2 is expressed in human and mouse epidermis and impairs skin barrier function in Netherton syndrome through filaggrin and lipid misprocessing. J Clin Invest 120: 871–882. Braff MH, Di Nardo A, Gallo RL. 2005. Keratinocytes store the antimicrobial peptide cathelicidin in lamellar bodies. J Invest Dermatol 124: 394 –400. Brooke MA, Nitoiu D, Kelsell DP. 2012. Cell-cell connectivity: Desmosomes and disease. J Pathol 226: 158 –171. Candi E, Schmidt R, Melino G. 2005. The cornified envelope: A model of cell death in the skin. Nat Rev Mol Cell Biol 6: 328 –340. Chanarin I, Patel A, Slavin G, Wills EJ, Andrews TM, Stewart G. 1975. Neutral-lipid storage disease: A new disorder of lipid metabolism. Br Med J 1: 553–555. Chavanas S, Bodemer C, Rochat A, Hamel-Teillac D, Ali M, Irvine AD, Bonafe JL, Wilkinson J, Taieb A, Barrandon Y, et al. 2000. Mutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome. Nat Genet 25: 141– 142. Cheng J, Syder AJ, Yu QC, Letai A, Paller AS, Fuchs E. 1992. The genetic basis of epidermolytic hyperkeratosis: A disorder of differentiation-specific epidermal keratin genes. Cell 70: 811–819. Chidgey M, Brakebusch C, Gustafsson E, Cruchley A, Hail C, Kirk S, Merritt A, North A, Tselepis C, Hewitt J, et al. 2001. Mice lacking desmocollin 1 show epidermal fragility accompanied by barrier defects and abnormal differentiation. J Cell Biol 155: 821 –832. Chilcott RP, Farrar R. 2000. Biophysical measurements of human forearm skin in vivo: Effects of site, gender, chirality and time. Skin Res Technol 6: 64–69. Dasgupta C, Martinez AM, Zuppan CW, Shah MM, Bailey LL, Fletcher WH. 2001. Identification of connexin43 (a1) gap junction gene mutations in patients with hypoplastic left heart syndrome by denaturing gradient gel electrophoresis (DGGE). Mutat Res 479: 173 –186.

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

Epidermal Barriers

de Guzman Strong C, Wertz PW, Wang C, Yang F, Meltzer PS, Andl T, Millar SE, Ho IC, Pai SY, Segre JA. 2006. Lipid defect underlies selective skin barrier impairment of an epidermal-specific deletion of Gata-3. J Cell Biol 175: 661– 670. De Laurenzi V, Rogers GR, Hamrock DJ, Marekov LN, Steinert PM, Compton JG, Markova N, Rizzo WB. 1996. Sjogren-Larsson syndrome is caused by mutations in the fatty aldehyde dehydrogenase gene. Nat Genet 12: 52–57. Delva E, Tucker DK, Kowalczyk AP. 2009. The desmosome. Cold Spring Harb Perspect Biol 1: a002543. Denecker G, Hoste E, Gilbert B, Hochepied T, Ovaere P, Lippens S, Van den Broecke C, Van Damme P, D’Herde K, Hachem JP, et al. 2007. Caspase-14 protects against epidermal UVB photodamage and water loss. Nat Cell Biol 9: 666– 674. Descargues P, Deraison C, Bonnart C, Kreft M, Kishibe M, Ishida-Yamamoto A, Elias P, Barrandon Y, Zambruno G, Sonnenberg A, et al. 2005. Spink5-deficient mice mimic Netherton syndrome through degradation of desmoglein 1 by epidermal protease hyperactivity. Nat Genet 37: 56– 65. Djalilian AR, McGaughey D, Patel S, Seo EY, Yang C, Cheng J, Tomic M, Sinha S, Ishida-Yamamoto A, Segre JA. 2006. Connexin 26 regulates epidermal barrier and wound remodeling and promotes psoriasiform response. J Clin Invest 116: 1243–1253. Dorfman ML, Hershko C, Eisenberg S, Sagher F. 1974. Ichthyosiform dermatosis with systemic lipidosis. Arch Dermatol 110: 261– 266. Eckl KM, Tidhar R, Thiele H, Oji V, Hausser I, Brodesser S, Preil ML, Onal-Akan A, Stock F, Muller D, et al. 2013. Impaired epidermal ceramide synthesis causes autosomal recessive congenital ichthyosis and reveals the importance of ceramide acyl chain length. J Invest Dermatol 133: 2202–2211. Elias PM, Brown BE. 1978. The mammalian cutaneous permeability barrier: Defective barrier function is essential fatty acid deficiency correlates with abnormal intercellular lipid deposition. Lab Invest 39: 574–583. Elias PM, Friend DS. 1975. The permeability barrier in mammalian epidermis. J Cell Biol 65: 180–191. Elias PM, Hatano Y, Williams ML. 2008a. Basis for the barrier abnormality in atopic dermatitis: Outside-inside-outside pathogenic mechanisms. J Allergy Clin Immunol 121: 1337– 1343. Elias PM, Williams ML, Holleran WM, Jiang YJ, Schmuth M. 2008b. Pathogenesis of permeability barrier abnormalities in the ichthyoses: Inherited disorders of lipid metabolism. J Lipid Res 49: 697 –714. Elias PM, Crumrine D, Paller A, Rodriguez-Martin M, Williams ML. 2011. Pathogenesis of the cutaneous phenotype in inherited disorders of cholesterol metabolism: Therapeutic implications for topical treatment of these disorders. Dermatoendocrinology 3: 100– 106. Elias PM, Williams ML, Feingold KR. 2012. Abnormal barrier function in the pathogenesis of ichthyosis: Therapeutic implications for lipid metabolic disorders. Clin Dermatol 30: 311– 322. Epp N, Furstenberger G, Muller K, de Juanes S, Leitges M, Hausser I, Thieme F, Liebisch G, Schmitz G, Krieg P. 2007.

12R-lipoxygenase deficiency disrupts epidermal barrier function. J Cell Biol 177: 173– 182. Fallon PG, Sasaki T, Sandilands A, Campbell LE, Saunders SP, Mangan NE, Callanan JJ, Kawasaki H, Shiohama A, Kubo A, et al. 2009. A homozygous frameshift mutation in the murine filaggrin gene facilitates enhanced percutaneous allergen priming. Nat Genet 41: 602–608. Feingold KR, Jiang YJ. 2011. The mechanisms by which lipids coordinately regulate the formation of the protein and lipid domains of the stratum corneum: Role of fatty acids, oxysterols, cholesterol sulfate and ceramides as signaling molecules. Dermatoendocrinol 3: 113 –118. Fluhr JW, Feingold KR, Elias PM. 2006. Transepidermal water loss reflects permeability barrier status: Validation in human and rodent in vivo and ex vivo models. Exp Dermatol 15: 483–492. Frateschi S, Camerer E, Crisante G, Rieser S, Membrez M, Charles RP, Beermann F, Stehle JC, Breiden B, Sandhoff K, et al. 2011. PAR2 absence completely rescues inflammation and ichthyosis caused by altered CAP1/Prss8 expression in mouse skin. Nat Commun 2: 161. Frateschi S, Keppner A, Malsure S, Iwaszkiewicz J, Sergi C, Merillat AM, Fowler-Jaeger N, Randrianarison N, Planes C, Hummler E. 2012. Mutations of the serine protease CAP1/Prss8 lead to reduced embryonic viability, skin defects, and decreased ENaC activity. Am J Pathol 181: 605 –615. Furuse M, Fujita K, Hiiragi T, Fujimoto K, Tsukita S. 1998. Claudin-1 and -2: Novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol 141: 1539– 1550. Furuse M, Sasaki H, Tsukita S. 1999. Manner of interaction of heterogeneous claudin species within and between tight junction strands. J Cell Biol 147: 891–903. Furuse M, Hata M, Furuse K, Yoshida Y, Haratake A, Sugitani Y, Noda T, Kubo A, Tsukita S. 2002. Claudin-based tight junctions are crucial for the mammalian epidermal barrier: A lesson from claudin-1-deficient mice. J Cell Biol 156: 1099–1111. Gallo RL, Hooper LV. 2012. Epithelial antimicrobial defence of the skin and intestine. Nat Rev Immunol 12: 503– 516. Geng S, Mezentsev A, Kalachikov S, Raith K, Roop DR, Panteleyev AA. 2006. Targeted ablation of Arnt in mouse epidermis results in profound defects in desquamation and epidermal barrier function. J Cell Sci 119: 4901– 4912. Ghadially R, Brown BE, Hanley K, Reed JT, Feingold KR, Elias PM. 1996. Decreased epidermal lipid synthesis accounts for altered barrier function in aged mice. J Invest Dermatol 106: 1064– 1069. Ghosh AK, Ramakrishnan G, Chandramohan C, Rajasekharan R. 2008. CGI-58, the causative gene for Chanarin-Dorfman syndrome, mediates acylation of lysophosphatidic acid. J Biol Chem 283: 24525–24533. Guillou H, Zadravec D, Martin PG, Jacobsson A. 2010. The key roles of elongases and desaturases in mammalian fatty acid metabolism: Insights from transgenic mice. Prog Lipid Res 49: 186 –199. Hadj-Rabia S, Baala L, Vabres P, Hamel-Teillac D, Jacquemin E, Fabre M, Lyonnet S, De Prost Y, Munnich A, Hadchouel M, et al. 2004. Claudin-1 gene mutations in neo-

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

13

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

K. Natsuga

natal sclerosing cholangitis associated with ichthyosis: A tight junction disease. Gastroenterology 127: 1386– 1390. Hammarlund K, Sedin G. 1979. Transepidermal water loss in newborn infants: III. Relation to gestational age. Acta Paediatr Scand 68: 795 –801. Hardman MJ, Sisi P, Banbury DN, Byrne C. 1998. Patterned acquisition of skin barrier function during development. Development 125: 1541–1552. Herrmann T, van der Hoeven F, Grone HJ, Stewart AF, Langbein L, Kaiser I, Liebisch G, Gosch I, Buchkremer F, Drobnik W, et al. 2003. Mice with targeted disruption of the fatty acid transport protein 4 (Fatp 4, Slc27a4) gene show features of lethal restrictive dermopathy. J Cell Biol 161: 1105–1115. Herrmann T, Grone HJ, Langbein L, Kaiser I, Gosch I, Bennemann U, Metzger D, Chambon P, Stewart AF, Stremmel W. 2005. Disturbed epidermal structure in mice with temporally controlled fatp4 deficiency. J Invest Dermatol 125: 1228–1235. Hewett DR, Simons AL, Mangan NE, Jolin HE, Green SM, Fallon PG, McKenzie AN. 2005. Lethal, neonatal ichthyosis with increased proteolytic processing of filaggrin in a mouse model of Netherton syndrome. Hum Mol Genet 14: 335 –346. Holleran WM, Ginns EI, Menon GK, Grundmann JU, Fartasch M, McKinney CE, Elias PM, Sidransky E. 1994. Consequences of b-glucocerebrosidase deficiency in epidermis. Ultrastructure and permeability barrier alterations in Gaucher disease. J Clin Invest 93: 1756– 1764. Hoste E, Kemperman P, Devos M, Denecker G, Kezic S, Yau N, Gilbert B, Lippens S, De Groote P, Roelandt R, et al. 2011. Caspase-14 is required for filaggrin degradation to natural moisturizing factors in the skin. J Invest Dermatol 131: 2233–2241. Hovnanian A. 2013. Netherton syndrome: Skin inflammation and allergy by loss of protease inhibition. Cell Tissue Res 351: 289– 300. Huber M, Rettler I, Bernasconi K, Frenk E, Lavrijsen SP, Ponec M, Bon A, Lautenschlager S, Schorderet DF, Hohl D. 1995. Mutations of keratinocyte transglutaminase in lamellar ichthyosis. Science 267: 525– 528. Human Microbiome Project C. 2012a. A framework for human microbiome research. Nature 486: 215– 221. Human Microbiome Project C. 2012b. Structure, function and diversity of the healthy human microbiome. Nature 486: 207– 214. Indra AK, Leid M. 2011. Epidermal permeability barrier measurement in mammalian skin. Methods Mol Biol 763: 73–81. Indra AK, Mohan WS 2nd, Frontini M, Scheer E, Messaddeq N, Metzger D, Tora L. 2005. TAF10 is required for the establishment of skin barrier function in foetal, but not in adult mouse epidermis. Dev Biol 285: 28– 37. Ishida-Yamamoto A. 2003. Loricrin keratoderma: A novel disease entity characterized by nuclear accumulation of mutant loricrin. J Dermatol Sci 31: 3– 8. Jakobsson A, Westerberg R, Jacobsson A. 2006. Fatty acid elongases in mammals: Their regulation and roles in metabolism. Prog Lipid Res 45: 237–249. Jaubert J, Cheng J, Segre JA. 2003. Ectopic expression of kruppel like factor 4 (Klf4) accelerates formation of the

14

epidermal permeability barrier. Development 130: 2767– 2777. Jennemann R, Rabionet M, Gorgas K, Epstein S, Dalpke A, Rothermel U, Bayerle A, van der Hoeven F, Imgrund S, Kirsch J, et al. 2012. Loss of ceramide synthase 3 causes lethal skin barrier disruption. Hum Mol Genet 21: 586– 608. Jobard F, Lefevre C, Karaduman A, Blanchet-Bardon C, Emre S, Weissenbach J, Ozguc M, Lathrop M, Prud’homme JF, Fischer J. 2002. Lipoxygenase-3 (ALOXE3) and 12(R)-lipoxygenase (ALOX12B) are mutated in nonbullous congenital ichthyosiform erythroderma (NCIE) linked to chromosome 17p13.1. Hum Mol Genet 11: 107– 113. Kabashima-Kubo R, Nakamura M, Sakabe J, Sugita K, Hino R, Mori T, Kobayashi M, Bito T, Kabashima K, Ogasawara K, et al. 2012. A group of atopic dermatitis without IgE elevation or barrier impairment shows a high Th1 frequency: Possible immunological state of the intrinsic type. J Dermatol Sci 67: 37– 43. Kaufman CK, Zhou P, Pasolli HA, Rendl M, Bolotin D, Lim KC, Dai X, Alegre ML, Fuchs E. 2003. GATA-3: An unexpected regulator of cell lineage determination in skin. Genes Dev 17: 2108–2122. Kawasaki H, Nagao K, Kubo A, Hata T, Shimizu A, Mizuno H, Yamada T, Amagai M. 2012. Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrin-null mice. J Allergy Clin Immunol 129: 1538– 1546 e1536. Kelsell DP, Norgett EE, Unsworth H, Teh MT, Cullup T, Mein CA, Dopping-Hepenstal PJ, Dale BA, Tadini G, Fleckman P, et al. 2005. Mutations in ABCA12 underlie the severe congenital skin disease harlequin ichthyosis. Am J Hum Genet 76: 794– 803. Khnykin D, Miner JH, Jahnsen F. 2011. Role of fatty acid transporters in epidermis: Implications for health and disease. Dermatoendocrinology 3: 53–61. Kihara A. 2012. Very long-chain fatty acids: Elongation, physiology and related disorders. J Biochem 152: 387– 395. Klar J, Schweiger M, Zimmerman R, Zechner R, Li H, Torma H, Vahlquist A, Bouadjar B, Dahl N, Fischer J. 2009. Mutations in the fatty acid transport protein 4 gene cause the ichthyosis prematurity syndrome. Am J Hum Genet 85: 248– 253. Koch PJ, de Viragh PA, Scharer E, Bundman D, Longley MA, Bickenbach J, Kawachi Y, Suga Y, Zhou Z, Huber M, et al. 2000. Lessons from loricrin-deficient mice: Compensatory mechanisms maintaining skin barrier function in the absence of a major cornified envelope protein. J Cell Biol 151: 389 –400. Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, Nomicos E, Polley EC, Komarow HD, Program NCS, et al. 2012. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res 22: 850 –859. Krieg P, Rosenberger S, de Juanes S, Latzko S, Hou J, Dick A, Kloz U, van der Hoeven F, Hausser I, Esposito I, et al. 2013. Aloxe3 knockout mice reveal a function of epidermal lipoxygenase-3 as hepoxilin synthase and its pivotal role in barrier formation. J Invest Dermatol 133: 172– 180.

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

Epidermal Barriers

Kubo A, Nagao K, Amagai M. 2012. Epidermal barrier dysfunction and cutaneous sensitization in atopic diseases. J Clin Invest 122: 440– 447. Kuramoto N, Takizawa T, Takizawa T, Matsuki M, Morioka H, Robinson JM, Yamanishi K. 2002. Development of ichthyosiform skin compensates for defective permeability barrier function in mice lacking transglutaminase 1. J Clin Invest 109: 243– 250. Lai Y, Gallo RL. 2009. AMPed up immunity: How antimicrobial peptides have multiple roles in immune defense. Trends Immunol 30: 131– 141. Laiho E, Ignatius J, Mikkola H, Yee VC, Teller DC, Niemi KM, Saarialho-Kere U, Kere J, Palotie A. 1997. Transglutaminase 1 mutations in autosomal recessive congenital ichthyosis: Private and recurrent mutations in an isolated population. Am J Hum Genet 61: 529– 538. Lefevre C, Jobard F, Caux F, Bouadjar B, Karaduman A, Heilig R, Lakhdar H, Wollenberg A, Verret JL, Weissenbach J, et al. 2001. Mutations in CGI-58, the gene encoding a new protein of the esterase/lipase/thioesterase subfamily, in Chanarin-Dorfman syndrome. Am J Hum Genet 69: 1002–1012. Lefevre C, Audebert S, Jobard F, Bouadjar B, Lakhdar H, Boughdene-Stambouli O, Blanchet-Bardon C, Heilig R, Foglio M, Weissenbach J, et al. 2003. Mutations in the transporter ABCA12 are associated with lamellar ichthyosis type 2. Hum Mol Genet 12: 2369–2378. Lessard JC, Coulombe PA. 2012. Keratin 16-null mice develop palmoplantar keratoderma, a hallmark feature of pachyonychia congenita and related disorders. J Invest Dermatol 132: 1384–1391. Leyvraz C, Charles RP, Rubera I, Guitard M, Rotman S, Breiden B, Sandhoff K, Hummler E. 2005. The epidermal barrier function is dependent on the serine protease CAP1/Prss8. J Cell Biol 170: 487– 496. List K, Haudenschild CC, Szabo R, Chen W, Wahl SM, Swaim W, Engelholm LH, Behrendt N, Bugge TH. 2002. Matriptase/MT-SP1 is required for postnatal survival, epidermal barrier function, hair follicle development, and thymic homeostasis. Oncogene 21: 3765– 3779. List K, Szabo R, Wertz PW, Segre J, Haudenschild CC, Kim SY, Bugge TH. 2003. Loss of proteolytically processed filaggrin caused by epidermal deletion of Matriptase/ MT-SP1. J Cell Biol 163: 901– 910. Loomis KD, Zhu S, Yoon K, Johnson PF, Smart RC. 2007. Genetic ablation of CCAAT/enhancer binding protein a in epidermis reveals its role in suppression of epithelial tumorigenesis. Cancer Res 67: 6768–6776. Lopez RG, Garcia-Silva S, Moore SJ, Bereshchenko O, Martinez-Cruz AB, Ermakova O, Kurz E, Paramio JM, Nerlov C. 2009. C/EBP-a and b couple interfollicular keratinocyte proliferation arrest to commitment and terminal differentiation. Nat Cell Biol 11: 1181– 1190. Lundwall A, Brattsand M. 2008. Kallikrein-related peptidases. Cell Mol Life Sci 65: 2019– 2038. Maass K, Ghanem A, Kim JS, Saathoff M, Urschel S, Kirfel G, Grummer R, Kretz M, Lewalter T, Tiemann K, et al. 2004. Defective epidermal barrier in neonatal mice lacking the C-terminal region of connexin43. Mol Biol Cell 15: 4597–4608.

Maatta A, DiColandrea T, Groot K, Watt FM. 2001. Gene targeting of envoplakin, a cytoskeletal linker protein and precursor of the epidermal cornified envelope. Mol Cell Biol 21: 7047– 7053. Maeda S, Tsukihara T. 2011. Structure of the gap junction channel and its implications for its biological functions. Cell Mol Life Sci 68: 1115–1129. Magert HJ, Standker L, Kreutzmann P, Zucht HD, Reinecke M, Sommerhoff CP, Fritz H, Forssmann WG. 1999. LEKTI, a novel 15-domain type of human serine proteinase inhibitor. J Biol Chem 274: 21499–21502. Matsuki M, Yamashita F, Ishida-Yamamoto A, Yamada K, Kinoshita C, Fushiki S, Ueda E, Morishima Y, Tabata K, Yasuno H, et al. 1998. Defective stratum corneum and early neonatal death in mice lacking the gene for transglutaminase 1 (keratinocyte transglutaminase). Proc Natl Acad Sci 95: 1044–1049. Maytin EV, Habener JF. 1998. Transcription factors C/EBPa, C/EBP-b, and CHOP (Gadd153) expressed during the differentiation program of keratinocytes in vitro and in vivo. J Invest Dermatol 110: 238 –246. McAleer MA, Irvine AD. 2013. The multifunctional role of filaggrin in allergic skin disease. J Allergy Clin Immunol 131: 280–291. McLean WH, Hansen CD, Eliason MJ, Smith FJ. 2011. The phenotypic and molecular genetic features of pachyonychia congenita. J Invest Dermatol 131: 1015–1017. Mehrel T, Hohl D, Rothnagel JA, Longley MA, Bundman D, Cheng C, Lichti U, Bisher ME, Steven AC, Steinert PM, et al. 1990. Identification of a major keratinocyte cell envelope protein, loricrin. Cell 61: 1103–1112. Merzdorf CS, Chen YH, Goodenough DA. 1998. Formation of functional tight junctions in Xenopus embryos. Dev Biol 195: 187 –203. Miller GS, List K. 2013. The matriptase-prostasin proteolytic cascade in epithelial development and pathology. Cell Tissue Res 351: 245– 253. Mizutani Y, Mitsutake S, Tsuji K, Kihara A, Igarashi Y. 2009. Ceramide biosynthesis in keratinocyte and its role in skin function. Biochimie 91: 784– 790. Mohan WS Jr, Scheer E, Wendling O, Metzger D, Tora L. 2003. TAF10 (TAFII30) is necessary for TFIID stability and early embryogenesis in mice. Mol Cell Biol 23: 4307–4318. Moll R, Franke WW, Schiller DL, Geiger B, Krepler R. 1982. The catalog of human cytokeratins: Patterns of expression in normal epithelia, tumors and cultured cells. Cell 31: 11–24. Moniaga CS, Egawa G, Kawasaki H, Hara-Chikuma M, Honda T, Tanizaki H, Nakajima S, Otsuka A, Matsuoka H, Kubo A, et al. 2010. Flaky tail mouse denotes human atopic dermatitis in the steady state and by topical application with Dermatophagoides pteronyssinus extract. Am J Pathol 176: 2385– 2393. Moran JL, Qiu H, Turbe-Doan A, Yun Y, Boeglin WE, Brash AR, Beier DR. 2007. A mouse mutation in the 12R-lipoxygenase, Alox12b, disrupts formation of the epidermal permeability barrier. J Invest Dermatol 127: 1893–1897. Moulson CL, Martin DR, Lugus JJ, Schaffer JE, Lind AC, Miner JH. 2003. Cloning of wrinkle-free, a previously uncharacterized mouse mutation, reveals crucial roles

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

15

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

K. Natsuga

for fatty acid transport protein 4 in skin and hair development. Proc Natl Acad Sci 100: 5274–5279. Moulson CL, Lin MH, White JM, Newberry EP, Davidson NO, Miner JH. 2007. Keratinocyte-specific expression of fatty acid transport protein 4 rescues the wrinkle-free phenotype in Slc27a4/Fatp4 mutant mice. J Biol Chem 282: 15912– 15920. Nakagawa N, Yamamoto M, Imai Y, Sakaguchi Y, Takizawa T, Ohta N, Yagi N, Hatta I, Hitomi K, Takizawa T, et al. 2012. Knocking-in the R142C mutation in transglutaminase 1 disrupts the stratum corneum barrier and postnatal survival of mice. J Dermatol Sci 65: 196– 206. Natsuga K, Akiyama M, Kato N, Sakai K, Sugiyama-Nakagiri Y, Nishimura M, Hata H, Abe M, Arita K, Tsuji-Abe Y, et al. 2007. Novel ABCA12 mutations identified in two cases of non-bullous congenital ichthyosiform erythroderma associated with multiple skin malignant neoplasia. J Invest Dermatol 127: 2669– 2673. Natsuga K, Akiyama M, Shimizu H. 2011. Malignant skin tumours in patients with inherited ichthyosis. Br J Dermatol 165: 263– 268. Nelson WG, Sun TT. 1983. The 50- and 58-kdalton keratin classes as molecular markers for stratified squamous epithelia: Cell culture studies. J Cell Biol 97: 244– 251. Netzel-Arnett S, Currie BM, Szabo R, Lin CY, Chen LM, Chai KX, Antalis TM, Bugge TH, List K. 2006. Evidence for a matriptase-prostasin proteolytic cascade regulating terminal epidermal differentiation. J Biol Chem 281: 32941–32945. Nirunsuksiri W, Zhang SH, Fleckman P. 1998. Reduced stability and bi-allelic, coequal expression of profilaggrin mRNA in keratinocytes cultured from subjects with ichthyosis vulgaris. J Invest Dermatol 110: 854 –861. Nizet V, Ohtake T, Lauth X, Trowbridge J, Rudisill J, Dorschner RA, Pestonjamasp V, Piraino J, Huttner K, Gallo RL. 2001. Innate antimicrobial peptide protects the skin from invasive bacterial infection. Nature 414: 454– 457. Ny A, Egelrud T. 2004. Epidermal hyperproliferation and decreased skin barrier function in mice overexpressing stratum corneum chymotryptic enzyme. Acta Derm Venereol 84: 18–22. Oh HS, Smart RC. 1998. Expression of CCAAT/enhancer binding proteins (C/EBP) is associated with squamous differentiation in epidermis and isolated primary keratinocytes and is altered in skin neoplasms. J Invest Dermatol 110: 939 –945. Ohno Y, Suto S, Yamanaka M, Mizutani Y, Mitsutake S, Igarashi Y, Sassa T, Kihara A. 2010. ELOVL1 production of C24 acyl-CoAs is linked to C24 sphingolipid synthesis. Proc Natl Acad Sci 107: 18439– 18444. Oji V, Tadini G, Akiyama M, Blanchet Bardon C, Bodemer C, Bourrat E, Coudiere P, DiGiovanna JJ, Elias P, Fischer J, et al. 2010. Revised nomenclature and classification of inherited ichthyoses: Results of the First Ichthyosis Consensus Conference in Soreze 2009. J Am Acad Dermatol 63: 607 –641. Ovaere P, Lippens S, Vandenabeele P, Declercq W. 2009. The emerging roles of serine protease cascades in the epidermis. Trends Biochem Sci 34: 453 –463. Oyoshi MK, Murphy GF, Geha RS. 2009. Filaggrin-deficient mice exhibit TH17-dominated skin inflammation and

16

permissiveness to epicutaneous sensitization with protein antigen. J Allergy Clin Immunol 124: 485–493, 493.e1. Palmer CN, Irvine AD, Terron-Kwiatkowski A, Zhao Y, Liao H, Lee SP, Goudie DR, Sandilands A, Campbell LE, Smith FJ, et al. 2006. Common loss-of-function variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nat Genet 38: 441 –446. Paznekas WA, Boyadjiev SA, Shapiro RE, Daniels O, Wollnik B, Keegan CE, Innis JW, Dinulos MB, Christian C, Hannibal MC, et al. 2003. Connexin 43 (GJA1) mutations cause the pleiotropic phenotype of oculodentodigital dysplasia. Am J Hum Genet 72: 408– 418. Perez-Moreno M, Jamora C, Fuchs E. 2003. Sticky business: Orchestrating cellular signals at adherens junctions. Cell 112: 535–548. Petrof G, Mellerio JE, McGrath JA. 2012. Desmosomal genodermatoses. Br J Dermatol 166: 36–45. Presland RB, Boggess D, Lewis SP, Hull C, Fleckman P, Sundberg JP. 2000. Loss of normal profilaggrin and filaggrin in flaky tail (ft/ft) mice: An animal model for the filaggrin-deficient skin disease ichthyosis vulgaris. J Invest Dermatol 115: 1072– 1081. Proksch E, Brandner JM, Jensen JM. 2008. The skin: An indispensable barrier. Exp Dermatol 17: 1063– 1072. Radner FP, Streith IE, Schoiswohl G, Schweiger M, Kumari M, Eichmann TO, Rechberger G, Koefeler HC, Eder S, Schauer S, et al. 2010. Growth retardation, impaired triacylglycerol catabolism, hepatic steatosis, and lethal skin barrier defect in mice lacking comparative gene identification-58 (CGI-58). J Biol Chem 285: 7300–7311. Radner FP, Marrakchi S, Kirchmeier P, Kim GJ, Ribierre F, Kamoun B, Abid L, Leipoldt M, Turki H, Schempp W, et al. 2013. Mutations in CERS3 cause autosomal recessive congenital ichthyosis in humans. PLoS Genet 9: e1003536. Reichelt J, Bussow H, Grund C, Magin TM. 2001. Formation of a normal epidermis supported by increased stability of keratins 5 and 14 in keratin 10 null mice. Mol Biol Cell 12: 1557–1568. Rice RH, Green H. 1977. The cornified envelope of terminally differentiated human epidermal keratinocytes consists of cross-linked protein. Cell 11: 417–422. Rice RH, Green H. 1979. Presence in human epidermal cells of a soluble protein precursor of the cross-linked envelope: Activation of the cross-linking by calcium ions. Cell 18: 681– 694. Rizzo WB. 2011. The role of fatty aldehyde dehydrogenase in epidermal structure and function. Dermatoendocrinology 3: 91–99. Robertson ED, Weir L, Romanowska M, Leigh IM, Panteleyev AA. 2012. ARNT controls the expression of epidermal differentiation genes through HDAC- and EGFR-dependent pathways. J Cell Sci 125: 3320–3332. Rodriguez E, Baurecht H, Herberich E, Wagenpfeil S, Brown SJ, Cordell HJ, Irvine AD, Weidinger S. 2009. Meta-analysis of filaggrin polymorphisms in eczema and asthma: Robust risk factors in atopic disease. J Allergy Clin Immunol 123: 1361–1370 e1367. Rodriguez-Martin M, Martin-Ezquerra G, Man MQ, Hupe M, Youm JK, Mackenzie DS, Cho S, Trullas C, Holleran WM, Radek KA, et al. 2011. Expression of epidermal

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

www.perspectivesinmedicine.org

Epidermal Barriers

CAMP changes in parallel with permeability barrier status. J Invest Dermatol 131: 2263–2270. Roop DR, Hawley-Nelson P, Cheng CK, Yuspa SH. 1983. Keratin gene expression in mouse epidermis and cultured epidermal cells. Proc Natl Acad Sci 80: 716– 720. Roth W, Kumar V, Beer HD, Richter M, Wohlenberg C, Reuter U, Thiering S, Staratschek-Jox A, Hofmann A, Kreusch F, et al. 2012. Keratin 1 maintains skin integrity and participates in an inflammatory network in skin through interleukin-18. J Cell Sci 125: 5269–5279. Rothnagel JA, Dominey AM, Dempsey LD, Longley MA, Greenhalgh DA, Gagne TA, Huber M, Frenk E, Hohl D, Roop DR. 1992. Mutations in the rod domains of keratins 1 and 10 in epidermolytic hyperkeratosis. Science 257: 1128– 1130. Ruhrberg C, Hajibagheri MA, Simon M, Dooley TP, Watt FM. 1996. Envoplakin, a novel precursor of the cornified envelope that has homology to desmoplakin. J Cell Biol 134: 715– 729. Ruhrberg C, Hajibagheri MA, Parry DA, Watt FM. 1997. Periplakin, a novel component of cornified envelopes and desmosomes that belongs to the plakin family and forms complexes with envoplakin. J Cell Biol 139: 1835– 1849. Russell LJ, DiGiovanna JJ, Rogers GR, Steinert PM, Hashem N, Compton JG, Bale SJ. 1995. Mutations in the gene for transglutaminase 1 in autosomal recessive lamellar ichthyosis. Nat Genet 9: 279–283. Sales KU, Masedunskas A, Bey AL, Rasmussen AL, Weigert R, List K, Szabo R, Overbeek PA, Bugge TH. 2010. Matriptase initiates activation of epidermal pro-kallikrein and disease onset in a mouse model of Netherton syndrome. Nat Genet 42: 676 –683. Sasaki T, Shiohama A, Kubo A, Kawasaki H, Ishida-Yamamoto A, Yamada T, Hachiya T, Shimizu A, Okano H, Kudoh J, et al. 2013. A homozygous nonsense mutation in the gene for Tmem79, a component for the lamellar granule secretory system, produces spontaneous eczema in an experimental model of atopic dermatitis. J Allergy Clin Immunol 132: 1111– 1120. Sassa T, Ohno Y, Suzuki S, Nomura T, Nishioka C, Kashiwagi T, Hirayama T, Akiyama M, Taguchi R, Shimizu H, et al. 2013. Impaired epidermal permeability barrier in mice lacking the Elovl1 gene responsible for very longchain fatty acid production. Mol Cell Biol 33: 2787– 2796. Saunders SP, Goh CS, Brown SJ, Palmer CN, Porter RM, Cole C, Campbell LE, Gierlinski M, Barton GJ, Schneider G, et al. 2013. Tmem79/Matt is the matted mouse gene and is a predisposing gene for atopic dermatitis in human subjects. J Allergy Clin Immunol 132: 1121– 1129. Scharschmidt TC, List K, Grice EA, Szabo R, Program NCS, Renaud G, Lee CC, Wolfsberg TG, Bugge TH, Segre JA. 2009a. Matriptase-deficient mice exhibit ichthyotic skin with a selective shift in skin microbiota. J Invest Dermatol 129: 2435–2442. Scharschmidt TC, Man MQ, Hatano Y, Crumrine D, Gunathilake R, Sundberg JP, Silva KA, Mauro TM, Hupe M, Cho S, et al. 2009b. Filaggrin deficiency confers a paracellular barrier abnormality that reduces inflammatory thresholds to irritants and haptens. J Allergy Clin Immunol 124: 496 –506, 506.e1–6.

Schmuth M, Martinz V, Janecke AR, Fauth C, Schossig A, Zschocke J, Gruber R. 2013. Inherited ichthyoses/generalized Mendelian disorders of cornification. Eur J Hum Genet 21: 123– 133. Schweizer J, Bowden PE, Coulombe PA, Langbein L, Lane EB, Magin TM, Maltais L, Omary MB, Parry DAD, Rogers MA, et al. 2006. New consensus nomenclature for mammalian keratins. J Cell Biol 174: 169–174. Segre JA. 2006. Epidermal barrier formation and recovery in skin disorders. J Clin Invest 116: 1150–1158. Segre JA, Bauer C, Fuchs E. 1999. Klf4 is a transcription factor required for establishing the barrier function of the skin. Nat Genet 22: 356–360. Sevilla LM, Nachat R, Groot KR, Klement JF, Uitto J, Djian P, Maatta A, Watt FM. 2007. Mice deficient in involucrin, envoplakin, and periplakin have a defective epidermal barrier. J Cell Biol 179: 1599–1612. Smith FJ, Irvine AD, Terron-Kwiatkowski A, Sandilands A, Campbell LE, Zhao Y, Liao H, Evans AT, Goudie DR, Lewis-Jones S, et al. 2006. Loss-of-function mutations in the gene encoding filaggrin cause ichthyosis vulgaris. Nat Genet 38: 337 –342. Smyth I, Hacking DF, Hilton AA, Mukhamedova N, Meikle PJ, Ellis S, Satterley K, Collinge JE, de Graaf CA, Bahlo M, et al. 2008. A mouse model of harlequin ichthyosis delineates a key role for Abca12 in lipid homeostasis. PLoS Genet 4: e1000192. Spacek DV, Perez AF, Ferranti KM, Wu LK, Moy DM, Magnan DR, King TR. 2010. The mouse frizzy (fr) and rat “hairless” (frCR) mutations are natural variants of protease serine S1 family member 8 (Prss8). Exp Dermatol 19: 527– 532. Steinert PM, Cantieri JS, Teller DC, Lonsdale-Eccles JD, Dale BA. 1981. Characterization of a class of cationic proteins that specifically interact with intermediate filaments. Proc Natl Acad Sci 78: 4097– 4101. Sugawara T, Iwamoto N, Akashi M, Kojima T, Hisatsune J, Sugai M, Furuse M. 2013. Tight junction dysfunction in the stratum granulosum leads to aberrant stratum corneum barrier function in claudin-1-deficient mice. J Dermatol Sci 70: 12– 18. Sybert VP, Dale BA, Holbrook KA. 1985. Ichthyosis vulgaris: Identification of a defect in synthesis of filaggrin correlated with an absence of keratohyaline granules. J Invest Dermatol 84: 191–194. Takagi S, Tojo H, Tomita S, Sano S, Itami S, Hara M, Inoue S, Horie K, Kondoh G, Hosokawa K, et al. 2003. Alteration of the 4-sphingenine scaffolds of ceramides in keratinocyte-specific Arnt-deficient mice affects skin barrier function. J Clin Invest 112: 1372– 1382. Ting SB, Caddy J, Hislop N, Wilanowski T, Auden A, Zhao LL, Ellis S, Kaur P, Uchida Y, Holleran WM, et al. 2005. A homolog of Drosophila grainy head is essential for epidermal integrity in mice. Science 308: 411–413. Tsuji S, Choudary PV, Martin BM, Stubblefield BK, Mayor JA, Barranger JA, Ginns EI. 1987. A mutation in the human glucocerebrosidase gene in neuronopathic Gaucher’s disease. N Engl J Med 316: 570– 575. Tunggal JA, Helfrich I, Schmitz A, Schwarz H, Gunzel D, Fromm M, Kemler R, Krieg T, Niessen CM. 2005. E-cadherin is essential for in vivo epidermal barrier function by regulating tight junctions. EMBO J 24: 1146– 1156.

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

17

Downloaded from http://perspectivesinmedicine.cshlp.org/ at WASHINGTON UNIV SCHOFMED on December 22, 2014 - Published by Cold Spring Harbor Laboratory Press

K. Natsuga

sociation, and destabilization of corneodesmosin in Spink5 – / – mice. Genes Dev 18: 2354–2358. Yonemura S. 2011. Cadherin-actin interactions at adherens junctions. Curr Opin Cell Biol 23: 515– 522. Zheng Y, Yin H, Boeglin WE, Elias PM, Crumrine D, Beier DR, Brash AR. 2011. Lipoxygenases mediate the effect of essential fatty acid in skin barrier formation: A proposed role in releasing v-hydroxyceramide for construction of the corneocyte lipid envelope. J Biol Chem 286: 24046– 24056. Zhu S, Oh HS, Shim M, Sterneck E, Johnson PF, Smart RC. 1999. C/EBP-b modulates the early events of keratinocyte differentiation involving growth arrest and keratin 1 and keratin 10 expression. Mol Cell Biol 19: 7181– 7190. Zuo Y, Zhuang DZ, Han R, Isaac G, Tobin JJ, McKee M, Welti R, Brissette JL, Fitzgerald ML, Freeman MW. 2008. ABCA12 maintains the epidermal lipid permeability barrier by facilitating formation of ceramide linoleic esters. J Biol Chem 283: 36624– 36635.

www.perspectivesinmedicine.org

Turksen K, Troy TC. 2002. Permeability barrier dysfunction in transgenic mice overexpressing claudin 6. Development 129: 1775–1784. Vasireddy V, Uchida Y, Salem N Jr, Kim SY, Mandal MN, Reddy GB, Bodepudi R, Alderson NL, Brown JC, Hama H, et al. 2007. Loss of functional ELOVL4 depletes very long-chain fatty acids (. or ¼C28) and the unique v-Oacylceramides in skin leading to neonatal death. Hum Mol Genet 16: 471– 482. Wallace L, Roberts-Thompson L, Reichelt J. 2012. Deletion of K1/K10 does not impair epidermal stratification but affects desmosomal structure and nuclear integrity. J Cell Sci 125: 1750–1758. Wilson DR, Maibach HI. 1980. Transepidermal water loss in vivo. Premature andterminfants.BiolNeonate 37: 180–185. Yanagi T, Akiyama M, Nishihara H, Sakai K, Nishie W, Tanaka S, Shimizu H. 2008. Harlequin ichthyosis model mouse reveals alveolar collapse and severe fetal skin barrier defects. Hum Mol Genet 17: 3075– 3083. Yang T, Liang D, Koch PJ, Hohl D, Kheradmand F, Overbeek PA. 2004. Epidermal detachment, desmosomal dis-

18

Cite this article as Cold Spring Harb Perspect Med 2014;4:a018218

Epidermal barriers.

The epidermis functions as a physical barrier to the external environment and works to prevent loss of water from the skin. Numerous factors have been...
692KB Sizes 2 Downloads 3 Views