Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Debra Crumrine is active.

Publication


Featured researches published by Debra Crumrine.


The Journal of Allergy and Clinical Immunology | 2009

FILAGGRIN DEFICIENCY CONFERS A PARACELLULAR BARRIER ABNORMALITY THAT REDUCES INFLAMMATORY THRESHOLDS TO IRRITANTS AND HAPTENS

Tiffany C. Scharschmidt; Mao-Qiang Man; Yutaka Hatano; Debra Crumrine; Roshan Gunathilake; John P. Sundberg; Kathleen A. Silva; Theodora M. Mauro; Melanie Hupe; Soyun Cho; Yan Wu; Anna Celli; Matthias Schmuth; Kenneth R. Feingold; Peter M. Elias

BACKGROUND Mutations in the human filaggrin gene (FLG) are associated with atopic dermatitis (AD) and are presumed to provoke a barrier abnormality. Yet additional acquired stressors might be necessary because the same mutations can result in a noninflammatory disorder, ichthyosis vulgaris. OBJECTIVE We examined here whether FLG deficiency alone suffices to produce a barrier abnormality, the basis for the putative abnormality, and its proinflammatory consequences. METHODS By using the flaky-tail mouse, which lacks processed murine filaggrin because of a frameshift mutation in the gene encoding profilaggrin that mimics some mutations in human AD, we assessed whether FLG deficiency provokes a barrier abnormality, further localized the defect, identified its subcellular basis, and assessed thresholds to irritant- and hapten-induced dermatitis. RESULTS Flaky-tail mice exhibit low-grade inflammation with increased bidirectional, paracellular permeability of water-soluble xenobiotes caused by impaired lamellar body secretion and altered stratum corneum extracellular membranes. This barrier abnormality correlates with reduced inflammatory thresholds to both topical irritants and haptens. Moreover, when exposed repeatedly to topical haptens at doses that produce no inflammation in wild-type mice, flaky-tail mice experience a severe AD-like dermatosis with a further deterioration in barrier function and features of a T(H)2 immunophenotype (increased CRTH levels plus inflammation, increased serum IgE levels, and reduced antimicrobial peptide [mBD3] expression). CONCLUSIONS FLG deficiency alone provokes a paracellular barrier abnormality in mice that reduces inflammatory thresholds to topical irritants/haptens, likely accounting for enhanced antigen penetration in FLG-associated AD.


American Journal of Pathology | 2011

Filaggrin Genotype in Ichthyosis Vulgaris Predicts Abnormalities in Epidermal Structure and Function

Robert Gruber; Peter M. Elias; Debra Crumrine; Tzu Kai Lin; Johanna M. Brandner; Jean Pierre Hachem; Richard B. Presland; Philip Fleckman; Andreas R. Janecke; Aileen Sandilands; W.H. Irwin McLean; P. Fritsch; Michael Mildner; Erwin Tschachler; Matthias Schmuth

Although it is widely accepted that filaggrin (FLG) deficiency contributes to an abnormal barrier function in ichthyosis vulgaris and atopic dermatitis, the pathomechanism of how FLG deficiency provokes a barrier abnormality in humans is unknown. We report here that the presence of FLG mutations in Caucasians predicts dose-dependent alterations in epidermal permeability barrier function. Although FLG is an intracellular protein, the barrier abnormality occurred solely via a paracellular route in affected stratum corneum. Abnormal barrier function correlated with alterations in keratin filament organization (perinuclear retraction), impaired loading of lamellar body contents, followed by nonuniform extracellular distribution of secreted organelle contents, and abnormalities in lamellar bilayer architecture. In addition, we observed reductions in corneodesmosome density and tight junction protein expression. Thus, FLG deficiency provokes alterations in keratinocyte architecture that influence epidermal functions localizing to the extracellular matrix. These results clarify how FLG mutations impair epidermal permeability barrier function.


Journal of Clinical Investigation | 1997

Activators of the nuclear hormone receptors PPARalpha and FXR accelerate the development of the fetal epidermal permeability barrier.

Karen Hanley; Y Jiang; Debra Crumrine; N M Bass; R Appel; Peter M. Elias; M L Williams; Kenneth R. Feingold

Members of the superfamily of nuclear hormone receptors which are obligate heterodimeric partners of the retinoid X receptor may be important in epidermal development. Here, we examined the effects of activators of the receptors for vitamin D3 and retinoids, and of the peroxisome proliferator activated receptors (PPARs) and the farnesoid X-activated receptor (FXR), on the development of the fetal epidermal barrier in vitro. Skin explants from gestational day 17 rats (term is 22 d) are unstratified and lack a stratum corneum (SC). After incubation in hormone-free media for 3-4 d, a multilayered SC replete with mature lamellar membranes in the interstices and a functionally competent barrier appear. 9-cis or all-trans retinoic acid, 1,25 dihydroxyvitamin D3, or the PPARgamma ligands prostaglandin J2 or troglitazone did not affect the development of barrier function or epidermal morphology. In contrast, activators of the PPARalpha, oleic acid, linoleic acid, and clofibrate, accelerated epidermal development, resulting in mature lamellar membranes, a multilayered SC, and a competent barrier after 2 d of incubation. The FXR activators, all-trans farnesol and juvenile hormone III, also accelerated epidermal barrier development. Activities of beta-glucocerebrosidase and steroid sulfatase, enzymes previously linked to barrier maturation, also increased after treatment with PPARalpha and FXR activators. In contrast, isoprenoids, such as nerolidol, cis-farnesol, or geranylgeraniol, or metabolites in the cholesterol pathway, such as mevalonate, squalene, or 25-hydroxycholesterol, did not alter barrier development. Finally, additive effects were observed in explants incubated with clofibrate and farnesol together in suboptimal concentrations which alone did not affect barrier development. These data indicate a putative physiologic role for PPARalpha and FXR in epidermal barrier development.


Journal of The American Academy of Dermatology | 1997

Optimal ratios of topical stratum corneum lipids improve barrier recovery in chronologically aged skin

Elizabeth Zettersten; Ruby Ghadially; Kenneth R. Feingold; Debra Crumrine; Peter M. Elias

BACKGROUND Chronologically aged skin exhibits delayed recovery rates after defined barrier insults, with decreased epidermal lipid synthesis, and particularly a reduction in cholesterol synthesis. Prior studies in young mice (< 10 weeks) and humans (20 to 30 years of age) have shown that application of a mixture of cholesterol, ceramides, and essential/nonessential free fatty acids (FFAs) in an equimolar ratio allows normal barrier recovery, whereas any 3:1:1:1 ratio of these four ingredients accelerates barrier recovery. OBJECTIVE AND METHODS Our purpose was to compare the ability of equimolar and cholesterol- and FFA-dominant molar lipid mixtures (2% in propylene glycol/n-propanol, 7:3) versus vehicle alone on barrier recovery rates at 0, 3, 6, 24, 48 hours, and 1 week after tape stripping of aged hairless mouse (> 18 months) and chronologically aged human skin (80 +/- 5 years). RESULTS Whereas a single topical application of the equimolar mixture only allows normal recovery in young mice, it appeared to improve barrier recovery in chronologically aged mice (p < 0.06). Moreover, a 3:1:1:1 mixture with cholesterol as the dominant lipid further accelerated barrier recovery at 3 and 6 hours (p < 0.01 and p < 0.03, respectively, vs 1:1:1:1). Likewise, the cholesterol-dominant, optimal molar ratio mixture significantly accelerated barrier recovery in chronologically aged human skin at 6 hours (p < 0.005; n = 6). In contrast, in aged mice, an FFA-dominant mixture significantly delayed barrier recovery at 3, 6, and 24 hours (p < 0.005, 0.05, and 0.001, respectively), Finally, ultrastructural studies showed that lipid-induced, accelerated recovery in chronologically aged mice is associated with the accelerated replenishment of the stratum corneum interstices with lamellar unit structures. CONCLUSION These findings show that barrier recovery is accelerated in chronologically aged murine epidermis with optimized ratios of physiologic lipids, provided that cholesterol is the dominant lipid and that the same mixture also accelerates barrier recovery in chronologically aged human skin.


Experimental Dermatology | 2002

Basis for the permeability barrier abnormality in lamellar ichthyosis

Peter M. Elias; Matthias Schmuth; Yoshikazu Uchida; Robert H. Rice; Martin J. Behne; Debra Crumrine; Kenneth R. Feingold; Walter M. Holleran; D. Pharm

Abstract: The basis for the permeability barrier abnormality in lamellar ichthyosis (LI) is not known. LI is caused by mutations in the gene that encodes the enzyme, transglutaminase 1 (TGI), which is responsible for assembly of the cornified envelope (CE). TG1 also has been suggested recently to catalyze the covalent attachment of omega‐hydroxyceramides (omega‐OHCer) to the CE, forming the corneocyte‐lipid envelope (CLE). We first assessed the barrier function and the permeability pathway of the water‐soluble tracer, colloidal lanthanum, across the stratum corneum (SC) in patients with LI with absent (n = 4) or low (n = 2) TG1 activity/protein. Increased movement of tracer through the SC correlated with increased transcutaneous water loss, and tracer remained restricted to the SC interstices. Enhanced extracellular permeability, in turn, was explicable by truncation and fragmentation of extracellular lamellar membrane arrays. The resultant clefts in the SC interstices represent the likely pathway for increased water permeability. Moreover, tracer movement remained restricted to the interstices, despite the demonstration of increased corneocyte fragility associated with widespread variations in CE structure. Regardless of variability in CE structure, however, CLE structure and bound omega‐OHCer content were normal. The normal CLE in LI may explain both the restriction of tracer to the SC interstices, as well as the presence of foreshortened membrane arrays with near‐normal interlamellar dimensions. Finally, the demonstration of a normal CLE in LI also raises questions about the putative role of TG1 in forming the CLE. These results demonstrate: (1) the extracellular nature of increased permeability in LI; (2) discontinuities in extracellular membrane structures that account for the enhanced permeability in LI; (3) that these membrane abnormalities are both associated with and explained by abnormalities in the subjacent CE scaffold; and (4) an intact CLE is present in LI, despite abnormalities in the CE, which may restrict water movement to the SC interstices in LI.


Journal of Biological Chemistry | 2011

Lipoxygenases Mediate the Effect of Essential Fatty Acid in Skin Barrier Formation: A PROPOSED ROLE IN RELEASING OMEGA-HYDROXYCERAMIDE FOR CONSTRUCTION OF THE CORNEOCYTE LIPID ENVELOPE*♦

Yuxiang Zheng; Huiyong Yin; William E. Boeglin; Peter M. Elias; Debra Crumrine; David R. Beier; Alan R. Brash

A barrier to water loss is vital to maintaining life on dry land. Formation of the mammalian skin barrier requires both the essential fatty acid linoleate and the two lipoxygenases 12R-lipoxygenase (12R-LOX) and epidermal lipoxygenase-3 (eLOX3), although their roles are poorly understood. Linoleate occurs in O-linoleoyl-ω-hydroxyceramide, which, after hydrolysis of the linoleate moiety, is covalently attached to protein via the free ω-hydroxyl of the ceramide, forming the corneocyte lipid envelope, a scaffold between lipid and protein that helps seal the barrier. Here we show using HPLC-UV, LC-MS, GC-MS, and 1H NMR that O-linoleoyl-ω-hydroxyceramide is oxygenated in a regio- and stereospecific fashion by the consecutive actions of 12R-LOX and eLOX3 and that these products occur naturally in pig and mouse epidermis. 12R-LOX forms 9R-hydroperoxy-linoleoyl-ω-hydroxyceramide, further converted by eLOX3 to specific epoxyalcohol (9R,10R-trans-epoxy-11E-13R-hydroxy) and 9-keto-10E,12Z esters of the ceramide; an epoxy-ketone derivative (9R,10R-trans-epoxy-11E-13-keto) is the most prominent oxidized ceramide in mouse skin. These products are absent in 12R-LOX-deficient mice, which crucially display a near total absence of protein-bound ω-hydroxyceramides and of the corneocyte lipid envelope and die shortly after birth from transepidermal water loss. We conclude that oxygenation of O-linoleoyl-ω-hydroxyceramide is required to facilitate the ester hydrolysis and allow bonding of the ω-hydroxyceramide to protein, providing a coherent explanation for the roles of multiple components in epidermal barrier function. Our study uncovers a hitherto unknown biochemical pathway in which the enzymic oxygenation of ceramides is involved in building a crucial structure of the epidermal barrier.


Journal of Investigative Dermatology | 2009

pH-regulated mechanisms account for pigment-type differences in epidermal barrier function.

Roshan Gunathilake; Nanna Schürer; Brenda A. Shoo; Anna Celli; Jean-Pierre Hachem; Debra Crumrine; Ganga Sirimanna; Kenneth R. Feingold; Theodora M. Mauro; Peter M. Elias

To determine whether pigment type determines differences in epidermal function, we studied stratum corneum (SC) pH, permeability barrier homeostasis, and SC integrity in three geographically disparate populations with pigment type I-II versus IV-V skin (Fitzpatrick I-VI scale). Type IV-V subjects showed: (i) lower surface pH (approximately 0.5 U); (ii) enhanced SC integrity (transepidermal water loss change with sequential tape strippings); and (iii) more rapid barrier recovery than type I-II subjects. Enhanced barrier function could be ascribed to increased epidermal lipid content, increased lamellar body production, and reduced acidity, leading to enhanced lipid processing. Compromised SC integrity in type I-II subjects could be ascribed to increased serine protease activity, resulting in accelerated desmoglein-1 (DSG-1)/corneodesmosome degradation. In contrast, DSG-1-positive CDs persisted in type IV-V subjects, but due to enhanced cathepsin-D activity, SC thickness did not increase. Adjustment of pH of type I-II SC to type IV-V levels improved epidermal function. Finally, dendrites from type IV-V melanocytes were more acidic than those from type I-II subjects, and they transfer more melanosomes to the SC, suggesting that melanosome secretion could contribute to the more acidic pH of type IV-V skin. These studies show marked pigment-type differences in epidermal structure and function that are pH driven.


Experimental Dermatology | 2006

Topical treatment with thiazolidinediones, activators of peroxisome proliferator-activated receptor-γ, normalizes epidermal homeostasis in a murine hyperproliferative disease model

Marianne Demerjian; Mao-Qiang Man; Eung-Ho Choi; Barbara E. Brown; Debra Crumrine; Sandra Chang; Theodora M. Mauro; Peter M. Elias; Kenneth R. Feingold

Abstract:  In a murine model of epidermal hyperplasia reproducing some of the abnormalities of several common skin disorders, we previously demonstrated the antiproliferative and pro‐differentiating effects of peroxisome proliferator‐activated receptor (PPAR)α, PPARβ/δ, and liver X receptor activators. Unlike other subgroups of PPAR activators, thiazolidinediones (TZDs), a family of PPARγ ligands, did not inhibit keratinocyte proliferation in normal murine skin. Here, we studied the effects of two TZDs, namely ciglitazone (10 mM) and troglitazone (1 mM), in the same murine model where epidermal hyperproliferation was reproduced by repeated barrier abrogation with tape stripping. Topical treatment with ciglitazone and troglitazone resulted in a marked and significant decrease in epidermal thickness. Furthermore, in all TZD‐treated groups, we observed a significant decrease in keratinocyte proliferation using proliferating cell nuclear antigen, 5‐bromo‐2′‐deoxyuridine, and tritiated thymidine incorporation. However, using the terminal deoxynucleotidyl transferase‐mediated dUTP nick end‐labeling assay, we found no difference in apoptosis between different treatments, emphasizing that it is the antiproliferative role of these activators that accounts for the decrease of epidermal thickness. Finally, using immunohistochemical methods, we determined the effects of ciglitazone on keratinocyte differentiation in this hyperproliferative model. We observed an increased expression of involucrin and filaggrin following ciglitazone treatment, suggesting a pro‐differentiating action of TZDs in this model. In summary, topical TZDs significantly reduce epidermal keratinocyte proliferation while promoting differentiation in a murine model of hyperproliferative epidermis. Together, these results suggest that in addition to their metabolic effects currently in use in the treatment of type 2 diabetes, topical TZDs could be considered as potential alternative therapeutic agents in hyperproliferative skin diseases such as psoriasis.


Journal of Clinical Investigation | 1996

Hormonal basis for the gender difference in epidermal barrier formation in the fetal rat. Acceleration by estrogen and delay by testosterone.

Karen Hanley; U Rassner; Y Jiang; D Vansomphone; Debra Crumrine; L Komüves; Peter M. Elias; Kenneth R. Feingold; M L Williams

Previous studies have shown that ontogeny of the epidermal permeability barrier and lung occur in parallel in the fetal rat, and that pharmacologic agents, such as glucocorticoids and thyroid hormone, accelerate maturation at comparable developmental time points. Gender also influences lung maturation, i.e., males exhibit delayed development. Sex steroid hormones exert opposite effects on lung maturation, with estrogens accelerating and androgens inhibiting. In this study, we demonstrate that cutaneous barrier formation, measured as transepidermal water loss, is delayed in male fetal rats. Administration of estrogen to pregnant mothers accelerates fetal barrier development both morphologically and functionally. Competent barriers also form sooner in skin explants incubated in estrogen-supplemented media in vitro. In contrast, administration of dihydrotestosterone delays barrier formation both in vivo and in vitro. Finally, treatment of pregnant rats with the androgen antagonist flutamide eliminates the gender difference in barrier formation. These studies indicate that (a) estrogen accelerates and testosterone delays cutaneous barrier formation, (b) these hormones exert their effects directly on the skin, and (c) sex differences in rates of barrier development in vivo may be mediated by testosterone.


Journal of Investigative Dermatology | 2009

Vitamin D Receptor and Coactivators SRC2 and 3 Regulate Epidermis-Specific Sphingolipid Production and Permeability Barrier Formation

Yuko Oda; Yoshikazu Uchida; Sam Moradian; Debra Crumrine; Peter M. Elias; Daniel D. Bikle

The vitamin D receptor (VDR) is a nuclear hormone receptor that controls transcription of target genes. It exerts its biological effects through transcriptional coactivators. Previously, we identified two distinct classes of VDR coactivators, VDR-interacting protein (DRIP) and steroid receptor coactivator (SRC) at different stages of keratinocyte differentiation. Here, we determined the functions of VDR and coactivators in lipid production and permeability barrier formation. Silencing of either VDR, SRC2, or SRC3 resulted in decreases in specific glucosylceramide (GlcCer) species but not other lipids such as cholesterol and free fatty acids. Their silencing also caused decreased transcription of fatty acid elongase and ceramide glucosyltransferase, which are critical for the synthesis of epidermis-unique GlcCer species, and defects in lamellar body formation associated with decreased expression of the lipid transporter ATP-binding cassette transporter protein 12. VDR null mice exhibit abnormal barrier function with altered lipid composition in vivo. These results demonstrate that VDR and coactivators SRC2 and SRC3, which are also involved in other nuclear receptors as well, are critical for epidermis-specific sphingolipid production and barrier formation. In contrast, DRIP silencing had no apparent effect on these processes indicating that the two classes of coactivators are differentially utilized.

Collaboration


Dive into the Debra Crumrine's collaboration.

Top Co-Authors

Avatar

Peter M. Elias

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mao-Qiang Man

University of California

View shared research outputs
Top Co-Authors

Avatar

Melanie Hupe

University of California

View shared research outputs
Top Co-Authors

Avatar

Matthias Schmuth

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge