1
|
Nüsken M, Heinemeier F, Matzke SS, Porebski P, Forkel S, Dasari P, Braun A, Zautner AE, Schön MP, Buhl T. Immune response to topical sodium lauryl sulfate differs from classical irritant and allergic contact dermatitis. Eur J Immunol 2024; 54:e2350798. [PMID: 39498726 DOI: 10.1002/eji.202350798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024]
Abstract
Sodium lauryl sulfate (SLS) is used as a control irritant in patch testing for allergic contact dermatitis (ACD). However, up to 20% of those tested react to SLS, whereby the pathophysiological basis of this reaction is still unclear. To mimic patch test reactions, we repeatedly applied SLS to the skin of wild-type mice. Reactions were compared with those in a classical ACD model induced by oxazolone and an irritant contact dermatitis (ICD) model induced by croton oil. Skin inflammation was assessed with ear thickness measurements, immunohistochemistry, qRT-PCR, and flow cytometry. Topical SLS treatment was further investigated in Flg/Hrnr-/-, Myd88/Tlr3-/-, and Rag1-/- mouse models. All three compounds caused ear swelling with different courses. Oxazolone treatment, compared with the ICD model, resulted in a greater influx of immune cells (CD4+, MHCII+, CD11b+). Similarly, SLS did not induce immune cell infiltration or expression of selected inflammatory and regulatory cytokines. SLS induced the most pronounced keratinocyte proliferation. Compared with wild-type mice, topical SLS application did not increase ear swelling in skin barrier deficient Flg/Hrnr-/- mice, but led to significantly delayed swelling in mice with defects in innate or adaptive immune functions (Myd88/Tlr3-/-, Rag1-/-). SLS-induced contact dermatitis differed from classical ACD and ICD, as it elicited less pronounced immune alterations. Skin barrier impairment does not affect SLS-induced contact dermatitis, whereas both innate and adaptive components are involved in SLS skin reactions.
Collapse
Affiliation(s)
- Marvin Nüsken
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Fabian Heinemeier
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Silke Sabina Matzke
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Patryk Porebski
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Susann Forkel
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Prasad Dasari
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Andrea Braun
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Erich Zautner
- Institute of Medical Microbiology and Hospital Hygiene, Otto von Guericke University Magdeburg, Magdeburg, Germany
- CHaMP, Center for Health and Medical Prevention, Otto von Guericke-University Magdeburg, Magdeburg, Germany
| | - Michael Peter Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
- Lower Saxony Institute of Occupational Dermatology, University Medical Centre Göttingen, Göttingen, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
- Lower Saxony Institute of Occupational Dermatology, University Medical Centre Göttingen, Göttingen, Germany
| |
Collapse
|
2
|
Cohen E, Johnson CN, Wasikowski R, Billi AC, Tsoi LC, Kahlenberg JM, Gudjonsson JE, Coulombe PA. Significance of stress keratin expression in normal and diseased epithelia. iScience 2024; 27:108805. [PMID: 38299111 PMCID: PMC10828818 DOI: 10.1016/j.isci.2024.108805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
A group of keratin intermediate filament genes, the type II KRT6A-C and type I KRT16 and KRT17, are deemed stress responsive as they are induced in keratinocytes of surface epithelia in response to environmental stressors, in skin disorders (e.g., psoriasis) and in carcinomas. Monitoring stress keratins is widely used to identify keratinocytes in an activated state. Here, we analyze single-cell transcriptomic data from healthy and diseased human skin to explore the properties of stress keratins. Relative to keratins occurring in healthy skin, stress-induced keratins are expressed at lower levels and show lesser type I-type II pairwise regulation. Stress keratins do not "replace" the keratins expressed during normal differentiation nor reflect cellular proliferation. Instead, stress keratins are consistently co-regulated with genes with roles in differentiation, inflammation, and/or activation of innate immunity at the single-cell level. These findings provide a roadmap toward explaining the broad diversity and contextual regulation of keratins.
Collapse
Affiliation(s)
- Erez Cohen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Craig N. Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rachael Wasikowski
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Allison C. Billi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - J. Michelle Kahlenberg
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pierre A. Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
3
|
A Kaleidoscope of Keratin Gene Expression and the Mosaic of Its Regulatory Mechanisms. Int J Mol Sci 2023; 24:ijms24065603. [PMID: 36982676 PMCID: PMC10052683 DOI: 10.3390/ijms24065603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Keratins are a family of intermediate filament-forming proteins highly specific to epithelial cells. A combination of expressed keratin genes is a defining property of the epithelium belonging to a certain type, organ/tissue, cell differentiation potential, and at normal or pathological conditions. In a variety of processes such as differentiation and maturation, as well as during acute or chronic injury and malignant transformation, keratin expression undergoes switching: an initial keratin profile changes accordingly to changed cell functions and location within a tissue as well as other parameters of cellular phenotype and physiology. Tight control of keratin expression implies the presence of complex regulatory landscapes within the keratin gene loci. Here, we highlight patterns of keratin expression in different biological conditions and summarize disparate data on mechanisms controlling keratin expression at the level of genomic regulatory elements, transcription factors (TFs), and chromatin spatial structure.
Collapse
|
4
|
Kuonen F, Li NY, Haensel D, Patel T, Gaddam S, Yerly L, Rieger K, Aasi S, Oro AE. c-FOS drives reversible basal to squamous cell carcinoma transition. Cell Rep 2021; 37:109774. [PMID: 34610301 PMCID: PMC8515919 DOI: 10.1016/j.celrep.2021.109774] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/28/2021] [Accepted: 09/08/2021] [Indexed: 01/22/2023] Open
Abstract
While squamous transdifferentiation within subpopulations of adenocarcinomas represents an important drug resistance problem, its underlying mechanism remains poorly understood. Here, using surface markers of resistant basal cell carcinomas (BCCs) and patient single-cell and bulk transcriptomic data, we uncover the dynamic roadmap of basal to squamous cell carcinoma transition (BST). Experimentally induced BST identifies activator protein 1 (AP-1) family members in regulating tumor plasticity, and we show that c-FOS plays a central role in BST by regulating the accessibility of distinct AP-1 regulatory elements. Remarkably, despite prominent changes in cell morphology and BST marker expression, we show using inducible model systems that c-FOS-mediated BST demonstrates reversibility. Blocking EGFR pathway activation after c-FOS induction partially reverts BST in vitro and prevents BST features in both mouse models and human tumors. Thus, by identifying the molecular basis of BST, our work reveals a therapeutic opportunity targeting plasticity as a mechanism of tumor resistance.
Collapse
MESH Headings
- Animals
- Carcinoma, Basal Cell/metabolism
- Carcinoma, Basal Cell/pathology
- Carcinoma, Basal Cell/veterinary
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/veterinary
- Cell Transdifferentiation/drug effects
- Chromatin Assembly and Disassembly
- Drug Resistance, Neoplasm/genetics
- Humans
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mucin-1/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-fos/antagonists & inhibitors
- Proto-Oncogene Proteins c-fos/genetics
- Proto-Oncogene Proteins c-fos/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Signal Transduction/drug effects
- Transcription Factor AP-1/metabolism
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- ras Proteins/genetics
- ras Proteins/metabolism
Collapse
Affiliation(s)
- François Kuonen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, 1011 Lausanne, Switzerland.
| | - Nancy Yanzhe Li
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Haensel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tiffany Patel
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sadhana Gaddam
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Yerly
- Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, 1011 Lausanne, Switzerland
| | - Kerri Rieger
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sumaira Aasi
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony E Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Suo L, VanBuren C, Hovland ED, Kedishvili NY, Sundberg JP, Everts HB. Dietary Vitamin A Impacts Refractory Telogen. Front Cell Dev Biol 2021; 9:571474. [PMID: 33614636 PMCID: PMC7892905 DOI: 10.3389/fcell.2021.571474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Hair follicles cycle through periods of growth (anagen), regression (catagen), rest (telogen), and release (exogen). Telogen is further divided into refractory and competent telogen based on expression of bone morphogenetic protein 4 (BMP4) and wingless-related MMTV integration site 7A (WNT7A). During refractory telogen hair follicle stem cells (HFSC) are inhibited. Retinoic acid synthesis proteins localized to the hair follicle and this localization pattern changed throughout the hair cycle. In addition, excess retinyl esters arrested hair follicles in telogen. The purpose of this study was to further define these hair cycle changes. BMP4 and WNT7A expression was also used to distinguish refractory from competent telogen in C57BL/6J mice fed different levels of retinyl esters from two previous studies. These two studies produced opposite results; and differed in the amount of retinyl esters the dams consumed and the age of the mice when the different diet began. There were a greater percentage of hair follicles in refractory telogen both when mice were bred on an unpurified diet containing copious levels of retinyl esters (study 1) and consumed excess levels of retinyl esters starting at 12 weeks of age, as well as when mice were bred on a purified diet containing adequate levels of retinyl esters (study 2) and remained on this diet at 6 weeks of age. WNT7A expression was consistent with these results. Next, the localization of vitamin A metabolism proteins in the two stages of telogen was examined. Keratin 6 (KRT6) and cellular retinoic acid binding protein 2 (CRABP2) localized almost exclusively to refractory telogen hair follicles in study 1. However, KRT6 and CRABP2 localized to both competent and refractory telogen hair follicles in mice fed adequate and high levels of retinyl esters in study 2. In mice bred and fed an unpurified diet retinol dehydrogenase SDR16C5, retinal dehydrogenase 2 (ALDH1A2), and cytochrome p450 26B1 (CYP26B1), enzymes and proteins involved in RA metabolism, localized to BMP4 positive refractory telogen hair follicles. This suggests that vitamin A may contribute to the inhibition of HFSC during refractory telogen in a dose dependent manner.
Collapse
Affiliation(s)
- Liye Suo
- Department of Human Nutrition, The Ohio State University, Columbus, OH, United States
| | - Christine VanBuren
- Department of Nutrition and Food Sciences, Texas Woman's University, Denton, TX, United States
| | - Eylul Damla Hovland
- Department of Nutrition and Food Sciences, Texas Woman's University, Denton, TX, United States
| | - Natalia Y Kedishvili
- Department of Biochemistry and Molecular Genetics, Schools of Medicine and Dentistry, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Helen B Everts
- Department of Human Nutrition, The Ohio State University, Columbus, OH, United States.,Department of Nutrition and Food Sciences, Texas Woman's University, Denton, TX, United States
| |
Collapse
|
6
|
Ehrlich F, Fischer H, Langbein L, Praetzel-Wunder S, Ebner B, Figlak K, Weissenbacher A, Sipos W, Tschachler E, Eckhart L. Differential Evolution of the Epidermal Keratin Cytoskeleton in Terrestrial and Aquatic Mammals. Mol Biol Evol 2019; 36:328-340. [PMID: 30517738 PMCID: PMC6367960 DOI: 10.1093/molbev/msy214] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Keratins are the main intermediate filament proteins of epithelial cells. In keratinocytes of the mammalian epidermis they form a cytoskeleton that resists mechanical stress and thereby are essential for the function of the skin as a barrier against the environment. Here, we performed a comparative genomics study of epidermal keratin genes in terrestrial and fully aquatic mammals to determine adaptations of the epidermal keratin cytoskeleton to different environments. We show that keratins K5 and K14 of the innermost (basal), proliferation-competent layer of the epidermis are conserved in all mammals investigated. In contrast, K1 and K10, which form the main part of the cytoskeleton in the outer (suprabasal) layers of the epidermis of terrestrial mammals, have been lost in whales and dolphins (cetaceans) and in the manatee. Whereas in terrestrial mammalian epidermis K6 and K17 are expressed only upon stress-induced epidermal thickening, high levels of K6 and K17 are consistently present in dolphin skin, indicating constitutive expression and substitution of K1 and K10. K2 and K9, which are expressed in a body site-restricted manner in human and mouse suprabasal epidermis, have been lost not only in cetaceans and manatee but also in some terrestrial mammals. The evolution of alternative splicing of K10 and differentiation-dependent upregulation of K23 have increased the complexity of keratin expression in the epidermis of terrestrial mammals. Taken together, these results reveal evolutionary diversification of the epidermal cytoskeleton in mammals and suggest a complete replacement of the quantitatively predominant epidermal proteins of terrestrial mammals by originally stress-inducible keratins in cetaceans.
Collapse
Affiliation(s)
- Florian Ehrlich
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Heinz Fischer
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Lutz Langbein
- Department of Genetics of Skin Carcinogenesis, German Cancer Research Center, Heidelberg, Germany
| | - Silke Praetzel-Wunder
- Department of Genetics of Skin Carcinogenesis, German Cancer Research Center, Heidelberg, Germany
| | - Bettina Ebner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katarzyna Figlak
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | | | - Wolfgang Sipos
- Clinical Department for Farm Animals and Herd Management, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Erwin Tschachler
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Leopold Eckhart
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Heikinheimo K, Jee KJ, Morgan PR, Nagy B, Knuutila S, Leivo I. Genetic Changes in Sporadic Keratocystic Odontogenic Tumors (Odontogenic Keratocysts). J Dent Res 2016; 86:544-9. [PMID: 17525355 DOI: 10.1177/154405910708600611] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Little is known about the genetic background of keratocystic odontogenic tumors (KCOT, odontogenic keratocysts). Our aim was to characterize genomic aberrations in sporadic KCOT using cDNA-expression arrays and array-comparative genomic hybridization. For cDNA-expression arrays, 10 KCOT specimens and 20 fetal tooth germs were studied. Quantitative real-time reverse-transcription/polymerase chain-reaction and immunohistochemical studies were also undertaken. Several genes were over-expressed in 12q13, including cytokeratin 6B ( KRT6B) (≈ 10-fold), epidermal growth factor receptor ERBB3 (~ 4.7-fold), and glioma-associated oncogene homologue 1 ( GLI1) (~ 5- to 12-fold). One amplicon (~ 0.7 Mega base pairs [Mbp]), covering several genes involved in the regulation of cell growth, was found in 12q13.2. Deletions were found in 3q13.1, 5p14.3, and 7q31.3, including the cell-adhesion-related gene cadherin 18 ( CDH18) and leukocyte cell adhesion molecule ( ALCAM, MEMD). Over-expressed and amplified genes in 12q13, also reported in several other tumors and cell lines, may contribute to the persistent growth characteristics of KCOT.
Collapse
Affiliation(s)
- K Heikinheimo
- Department of Oral and Maxillofacial Surgery, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, FIN-20520 Turku, Finland.
| | | | | | | | | | | |
Collapse
|
8
|
van Buul JD, Timmerman I. Small Rho GTPase-mediated actin dynamics at endothelial adherens junctions. Small GTPases 2016; 7:21-31. [PMID: 26825121 DOI: 10.1080/21541248.2015.1131802] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
VE-cadherin-based cell-cell junctions form the major restrictive barrier of the endothelium to plasma proteins and blood cells. The function of VE-cadherin and the actin cytoskeleton are intimately linked. Vascular permeability factors and adherent leukocytes signal through small Rho GTPases to tightly regulate actin cytoskeletal rearrangements in order to open and re-assemble endothelial cell-cell junctions in a rapid and controlled manner. The Rho GTPases are activated by guanine nucleotide exchange factors (GEFs), conferring specificity and context-dependent control of cell-cell junctions. Although the molecular mechanisms that couple cadherins to actin filaments are beginning to be elucidated, specific stimulus-dependent regulation of the actin cytoskeleton at VE-cadherin-based junctions remains unexplained. Accumulating evidence has suggested that depending on the vascular permeability factor and on the subcellular localization of GEFs, cell-cell junction dynamics and organization are differentially regulated by one specific Rho GTPase. In this Commentary, we focus on new insights how the junctional actin cytoskeleton is specifically and locally regulated by Rho GTPases and GEFs in the endothelium.
Collapse
Affiliation(s)
- Jaap D van Buul
- a Department of Molecular Cell Biology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| | - Ilse Timmerman
- b Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory , Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| |
Collapse
|
9
|
Velarde MC, Demaria M, Melov S, Campisi J. Pleiotropic age-dependent effects of mitochondrial dysfunction on epidermal stem cells. Proc Natl Acad Sci U S A 2015; 112:10407-12. [PMID: 26240345 PMCID: PMC4547253 DOI: 10.1073/pnas.1505675112] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tissue homeostasis declines with age partly because stem/progenitor cells fail to self-renew or differentiate. Because mitochondrial damage can accelerate aging, we tested the hypothesis that mitochondrial dysfunction impairs stem cell renewal or function. We developed a mouse model, Tg(KRT14-cre/Esr1) (20Efu/J) × Sod2 (tm1Smel) , that generates mitochondrial oxidative stress in keratin 14-expressing epidermal stem/progenitor cells in a temporally controlled manner owing to deletion of Sod2, a nuclear gene that encodes the mitochondrial antioxidant enzyme superoxide dismutase 2 (Sod2). Epidermal Sod2 loss induced cellular senescence, which irreversibly arrested proliferation in a fraction of keratinocytes. Surprisingly, in young mice, Sod2 deficiency accelerated wound closure, increasing epidermal differentiation and reepithelialization, despite the reduced proliferation. In contrast, at older ages, Sod2 deficiency delayed wound closure and reduced epidermal thickness, accompanied by epidermal stem cell exhaustion. In young mice, Sod2 deficiency accelerated epidermal thinning in response to the tumor promoter 12-O-tetradecanoylphorbol-13-acetate, phenocopying the reduced regeneration of older Sod2-deficient skin. Our results show a surprising beneficial effect of mitochondrial dysfunction at young ages, provide a potential mechanism for the decline in epidermal regeneration at older ages, and identify a previously unidentified age-dependent role for mitochondria in skin quality and wound closure.
Collapse
Affiliation(s)
| | - Marco Demaria
- Buck Institute for Research on Aging, Novato, CA 94945
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA 94945
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945 Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| |
Collapse
|
10
|
Reijnders CMA, van Lier A, Roffel S, Kramer D, Scheper RJ, Gibbs S. Development of a Full-Thickness Human Skin Equivalent In Vitro Model Derived from TERT-Immortalized Keratinocytes and Fibroblasts. Tissue Eng Part A 2015; 21:2448-59. [PMID: 26135533 PMCID: PMC4554934 DOI: 10.1089/ten.tea.2015.0139] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Currently, human skin equivalents (HSEs) used for in vitro assays (e.g., for wound healing) make use of primary human skin cells. Limitations of primary keratinocytes and fibroblasts include availability of donor skin and donor variation. The use of physiologically relevant cell lines could solve these limitations. The aim was to develop a fully differentiated HSE constructed entirely from human skin cell lines, which could be applied for in vitro wound-healing assays. Skin equivalents were constructed from human TERT-immortalized keratinocytes and fibroblasts (TERT-HSE) and compared with native skin and primary HSEs. HSEs were characterized by hematoxylin–eosin and immunohistochemical stainings with markers for epidermal proliferation and differentiation, basement membrane (BM), fibroblasts, and the extracellular matrix (ECM). Ultrastructure was determined with electron microscopy. To test the functionality of the TERT-HSE, burn and cold injuries were applied, followed by immunohistochemical stainings, measurement of reepithelialization, and determination of secreted wound-healing mediators. The TERT-HSE was composed of a fully differentiated epidermis and a fibroblast-populated dermis comparable to native skin and primary HSE. The epidermis consisted of proliferating keratinocytes within the basal layer, followed by multiple spinous layers, a granular layer, and cornified layers. Within the TERT-HSE, the membrane junctions such as corneosomes, desmosomes, and hemidesmosomes were well developed as shown by ultrastructure pictures. Furthermore, the BM consisted of a lamina lucida and lamina densa comparable to native skin. The dermal matrix of the TERT-HSE was more similar to native skin than the primary construct, since collagen III, an ECM marker, was present in TERT-HSEs and absent in primary HSEs. After wounding, the TERT-HSE was able to reepithelialize and secrete inflammatory wound-healing mediators. In conclusion, the novel TERT-HSE, constructed entirely from human cell lines, provides an excellent opportunity to study in vitro skin biology and can also be used for drug targeting and testing new therapeutics, and ultimately, for incorporating into skin-on-a chip in the future.
Collapse
Affiliation(s)
| | - Amanda van Lier
- 1 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Sanne Roffel
- 1 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Duco Kramer
- 2 Department of Dermatology, University Medical Centre Groningen , Groningen, The Netherlands
| | - Rik J Scheper
- 3 Department of Pathology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Susan Gibbs
- 1 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands .,4 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam , Amsterdam, The Netherlands
| |
Collapse
|
11
|
Rorke EA, Adhikary G, Young CA, Rice RH, Elias PM, Crumrine D, Meyer J, Blumenberg M, Eckert RL. Structural and biochemical changes underlying a keratoderma-like phenotype in mice lacking suprabasal AP1 transcription factor function. Cell Death Dis 2015; 6:e1647. [PMID: 25695600 PMCID: PMC4669787 DOI: 10.1038/cddis.2015.21] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/06/2015] [Indexed: 01/07/2023]
Abstract
Epidermal keratinocyte differentiation on the body surface is a carefully choreographed process that leads to assembly of a barrier that is essential for life. Perturbation of keratinocyte differentiation leads to disease. Activator protein 1 (AP1) transcription factors are key controllers of this process. We have shown that inhibiting AP1 transcription factor activity in the suprabasal murine epidermis, by expression of dominant-negative c-jun (TAM67), produces a phenotype type that resembles human keratoderma. However, little is understood regarding the structural and molecular changes that drive this phenotype. In the present study we show that TAM67-positive epidermis displays altered cornified envelope, filaggrin-type keratohyalin granule, keratin filament, desmosome formation and lamellar body secretion leading to reduced barrier integrity. To understand the molecular changes underlying this process, we performed proteomic and RNA array analysis. Proteomic study of the corneocyte cross-linked proteome reveals a reduction in incorporation of cutaneous keratins, filaggrin, filaggrin2, late cornified envelope precursor proteins, hair keratins and hair keratin-associated proteins. This is coupled with increased incorporation of desmosome linker, small proline-rich, S100, transglutaminase and inflammation-associated proteins. Incorporation of most cutaneous keratins (Krt1, Krt5 and Krt10) is reduced, but incorporation of hyperproliferation-associated epidermal keratins (Krt6a, Krt6b and Krt16) is increased. RNA array analysis reveals reduced expression of mRNA encoding differentiation-associated cutaneous keratins, hair keratins and associated proteins, late cornified envelope precursors and filaggrin-related proteins; and increased expression of mRNA encoding small proline-rich proteins, protease inhibitors (serpins), S100 proteins, defensins and hyperproliferation-associated keratins. These findings suggest that AP1 factor inactivation in the suprabasal epidermal layers reduces expression of AP1 factor-responsive genes expressed in late differentiation and is associated with a compensatory increase in expression of early differentiation genes.
Collapse
Affiliation(s)
- E A Rorke
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - G Adhikary
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - C A Young
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - R H Rice
- Department of Environmental Toxicology, University of California, Davis, CA, USA
| | - P M Elias
- Dermatology Service, Veterans Affairs Medical Center, San Francisco and Department of Dermatology, University of California, San Francisco, CA, USA
| | - D Crumrine
- Dermatology Service, Veterans Affairs Medical Center, San Francisco and Department of Dermatology, University of California, San Francisco, CA, USA
| | - J Meyer
- Dermatology Service, Veterans Affairs Medical Center, San Francisco and Department of Dermatology, University of California, San Francisco, CA, USA
| | - M Blumenberg
- The R.O. Perelman Department of Dermatology, Department of Biochemistry and Molecular Pharmacology, New York University Cancer Institute, New York City, NY, USA
| | - R L Eckert
- 1] Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA [2] Dermatology, University of Maryland School of Medicine, Baltimore, MD, USA [3] Obstetrics and Gynecology, University of Maryland School of Medicine, Baltimore, MD, USA [4] Greenebaum Cancer Center University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
|
13
|
Lineage-restricted function of the pluripotency factor NANOG in stratified epithelia. Nat Commun 2014; 5:4226. [PMID: 24979572 DOI: 10.1038/ncomms5226] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 05/23/2014] [Indexed: 12/22/2022] Open
Abstract
NANOG is a pluripotency transcription factor in embryonic stem cells; however, its role in adult tissues remains largely unexplored. Here we show that mouse NANOG is selectively expressed in stratified epithelia, most notably in the oesophagus where the Nanog promoter is hypomethylated. Interestingly, inducible ubiquitous overexpression of NANOG in mice causes hyperplasia selectively in the oesophagus, in association with increased cell proliferation. NANOG transcriptionally activates the mitotic programme, including Aurora A kinase (Aurka), in stratified epithelia, and endogenous NANOG directly binds to the Aurka promoter in primary keratinocytes. Interestingly, overexpression of Nanog or Aurka in mice increased proliferation and aneuploidy in the oesophageal basal epithelium. Finally, inactivation of NANOG in cell lines from oesophageal or head and neck squamous cell carcinomas (ESCCs or HNSCCs, respectively) results in lower levels of AURKA and decreased proliferation, and NANOG and AURKA expression are positively correlated in HNSCCs. Together, these results indicate that NANOG has a lineage-restricted mitogenic function in stratified epithelia.
Collapse
|
14
|
Preclinical Studies of a Specific PPARγ Modulator in the Control of Skin Inflammation. J Invest Dermatol 2014; 134:1001-1011. [DOI: 10.1038/jid.2013.448] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 08/27/2013] [Accepted: 09/10/2013] [Indexed: 12/29/2022]
|
15
|
Meyer W, Schoennagel B, Kacza J, Busche R, Hornickel IN, Hewicker-Trautwein M, Schnapper A. Keratinization of the esophageal epithelium of domesticated mammals. Acta Histochem 2014; 116:235-42. [PMID: 23948668 DOI: 10.1016/j.acthis.2013.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/12/2013] [Accepted: 07/14/2013] [Indexed: 11/25/2022]
Abstract
We studied the esophageal epithelium for keratinization characteristics from samples of domesticated mammals of three nutrition groups (herbivores: horse, cattle, sheep; omnivores: pig, dog, rat; carnivores: cat) using histochemistry (keratins, disulfides), sulfur measurements, and cryo-SEM. Keratins were found in all esophageal layers of all species, except for the equine Stratum corneum. The positive reaction staining of Pan-keratin was remarkable, but decreased in intensity toward the outer layers, whereas in the pig and cat, staining was confined to the corneal layer. The herbivores revealed positive staining reactions in the upper Stratum spinosum, particularly in the sheep. Regarding single keratins, CK6 immunostating was found in most esophageal layers, but only weakly or negatively in the porcine and equine Stratum corneum. CK13 staining was restricted to the sheep and here was found in all layers. CK14 could be detected in the equine and feline Stratum basale, and upper vital layers of the dog and rat. CK17 appeared only in the Stratum spinosum and Stratum granulosum, but in all layers of the dog and cat. Disulfides reacted strongest in the Stratum corneum of the herbivores, as corroborated by the sulfur concentrations in the esophagus. Our study emphasized that keratins are very important for the mechanical stability of the epithelial cells and cell layers of the mammalian esophagus. The role of these keratins in the esophageal epithelia is of specific interest owing to the varying feed qualities and mechanical loads of different nutrition groups, which have to be countered.
Collapse
|
16
|
Lee S, Kong Y, Weatherbee SD. Forward genetics identifies Kdf1/1810019J16Rik as an essential regulator of the proliferation-differentiation decision in epidermal progenitor cells. Dev Biol 2013; 383:201-13. [PMID: 24075906 DOI: 10.1016/j.ydbio.2013.09.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 09/16/2013] [Indexed: 10/26/2022]
Abstract
Cell fate decisions during embryogenesis and adult life govern tissue formation, homeostasis and repair. Two key decisions that must be tightly coordinated are proliferation and differentiation. Overproliferation can lead to hyperplasia or tumor formation while premature differentiation can result in a depletion of proliferating cells and organ failure. Maintaining this balance is especially important in tissues that undergo rapid turnover like skin however, despite recent advances, the genetic mechanisms that balance cell differentiation and proliferation are still unclear. In an unbiased genetic screen to identify genes affecting early development, we identified an essential regulator of the proliferation-differentiation balance in epidermal progenitor cells, the Keratinocyte differentiation factor 1 (Kdf1; 1810019J16Rik) gene. Kdf1 is expressed in epidermal cells from early stages of epidermis formation through adulthood. Specifically, Kdf1 is expressed both in epidermal progenitor cells where it acts to curb the rate of proliferation as well as in their progeny where it is required to block proliferation and promote differentiation. Consequently, Kdf1 mutants display both uncontrolled cell proliferation in the epidermis and failure to develop terminal fates. Our findings reveal a dual role for the novel gene Kdf1 both as a repressive signal for progenitor cell proliferation through its inhibition of p63 and a strong inductive signal for terminal differentiation through its interaction with the cell cycle regulator Stratifin.
Collapse
Affiliation(s)
- Sunjin Lee
- Department of Genetics, Yale University, New Haven, CT 06520, USA
| | | | | |
Collapse
|
17
|
Hyter S, Indra AK. Nuclear hormone receptor functions in keratinocyte and melanocyte homeostasis, epidermal carcinogenesis and melanomagenesis. FEBS Lett 2013; 587:529-41. [PMID: 23395795 PMCID: PMC3670764 DOI: 10.1016/j.febslet.2013.01.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 12/12/2012] [Accepted: 01/18/2013] [Indexed: 12/19/2022]
Abstract
Skin homeostasis is maintained, in part, through regulation of gene expression orchestrated by type II nuclear hormone receptors in a cell and context specific manner. This group of transcriptional regulators is implicated in various cellular processes including epidermal proliferation, differentiation, permeability barrier formation, follicular cycling and inflammatory responses. Endogenous ligands for the receptors regulate actions during skin development and maintenance of tissue homeostasis. Type II nuclear receptor signaling is also important for cellular crosstalk between multiple cell types in the skin. Overall, these nuclear receptors are critical players in keratinocyte and melanocyte biology and present targets for cutaneous disease management.
Collapse
Affiliation(s)
- Stephen Hyter
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
- Environmental Health Science Center, Oregon State University, Corvallis, Oregon, USA
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
18
|
Ramot Y, Sugawara K, Zákány N, Tóth BI, Bíró T, Paus R. A novel control of human keratin expression: cannabinoid receptor 1-mediated signaling down-regulates the expression of keratins K6 and K16 in human keratinocytes in vitro and in situ. PeerJ 2013; 1:e40. [PMID: 23638377 PMCID: PMC3628749 DOI: 10.7717/peerj.40] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 01/28/2013] [Indexed: 12/23/2022] Open
Abstract
Cannabinoid receptors (CB) are expressed throughout human skin epithelium. CB1 activation inhibits human hair growth and decreases proliferation of epidermal keratinocytes. Since psoriasis is a chronic hyperproliferative, inflammatory skin disease, it is conceivable that the therapeutic modulation of CB signaling, which can inhibit both proliferation and inflammation, could win a place in future psoriasis management. Given that psoriasis is characterized by up-regulation of keratins K6 and K16, we have investigated whether CB1 stimulation modulates their expression in human epidermis. Treatment of organ-cultured human skin with the CB1-specific agonist, arachidonoyl-chloro-ethanolamide (ACEA), decreased K6 and K16 staining intensity in situ. At the gene and protein levels, ACEA also decreased K6 expression of cultured HaCaT keratinocytes, which show some similarities to psoriatic keratinocytes. These effects were partly antagonized by the CB1-specific antagonist, AM251. While CB1-mediated signaling also significantly inhibited human epidermal keratinocyte proliferation in situ, as shown by K6/Ki-67-double immunofluorescence, the inhibitory effect of ACEA on K6 expression in situ was independent of its anti-proliferative effect. Given recent appreciation of the role of K6 as a functionally important protein that regulates epithelial wound healing in mice, it is conceivable that the novel CB1-mediated regulation of keratin 6/16 revealed here also is relevant to wound healing. Taken together, our results suggest that cannabinoids and their receptors constitute a novel, clinically relevant control element of human K6 and K16 expression.
Collapse
Affiliation(s)
- Yuval Ramot
- Department of Dermatology, University of Luebeck, Luebeck, Germany.,Department of Dermatology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Koji Sugawara
- Department of Dermatology, University of Luebeck, Luebeck, Germany.,Department of Dermatology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Nóra Zákány
- Department of Dermatology, University of Luebeck, Luebeck, Germany.,DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, MHSC, RCMM, University of Debrecen, Debrecen, Hungary
| | - Balázs I Tóth
- DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, MHSC, RCMM, University of Debrecen, Debrecen, Hungary.,Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Tamás Bíró
- DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, MHSC, RCMM, University of Debrecen, Debrecen, Hungary
| | - Ralf Paus
- Department of Dermatology, University of Luebeck, Luebeck, Germany.,Institute of Inflammation and Repair, and Dermatology Centre, University of Manchester, Manchester, UK
| |
Collapse
|
19
|
Hickman HD, Reynoso GV, Ngudiankama BF, Rubin EJ, Magadán JG, Cush SS, Gibbs J, Molon B, Bronte V, Bennink JR, Yewdell JW. Anatomically restricted synergistic antiviral activities of innate and adaptive immune cells in the skin. Cell Host Microbe 2013; 13:155-68. [PMID: 23414756 PMCID: PMC3591514 DOI: 10.1016/j.chom.2013.01.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 11/30/2012] [Accepted: 01/09/2013] [Indexed: 12/14/2022]
Abstract
Despite extensive ex vivo investigation, the spatiotemporal organization of immune cells interacting with virus-infected cells in tissues remains uncertain. To address this, we used intravital multiphoton microscopy to visualize immune cell interactions with virus-infected cells following epicutaneous vaccinia virus (VV) infection of mice. VV infects keratinocytes in epidermal foci and numerous migratory dermal inflammatory monocytes that outlie the foci. We observed Ly6G(+) innate immune cells infiltrating and controlling foci, while CD8(+) T cells remained on the periphery killing infected monocytes. Most antigen-specific CD8(+) T cells in the skin did not interact with virus-infected cells. Blocking the generation of reactive nitrogen species relocated CD8(+) T cells into foci, modestly reducing viral titers. Depletion of Ly6G(+) and CD8(+) cells dramatically increased viral titers, consistent with their synergistic but spatially segregated viral clearance activities. These findings highlight previously unappreciated differences in the anatomic specialization of antiviral immune cell subsets.
Collapse
Affiliation(s)
- Heather D Hickman
- Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Bar-Or D. The Filamentous Actin Cytoskeleton Organization and the Endothelial Cell Barrier*. Crit Care Med 2013; 41:686-7. [DOI: 10.1097/ccm.0b013e3182758863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Chioni AM, Grose R. FGFR1 cleavage and nuclear translocation regulates breast cancer cell behavior. ACTA ACUST UNITED AC 2012; 197:801-17. [PMID: 22665522 PMCID: PMC3373409 DOI: 10.1083/jcb.201108077] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
FGFR1 cleavage by Granzyme B induces its nuclear translocation, in which it stimulates cell migration through effects on gene expression. FGF-10 and its receptors, FGFR1 and FGFR2, have been implicated in breast cancer susceptibility and progression, suggesting that fibroblast growth factor (FGF) signaling may be co-opted by breast cancer cells. We identify a novel pathway downstream of FGFR1 activation, whereby the receptor is cleaved and traffics to the nucleus, where it can regulate specific target genes. We confirm Granzyme B (GrB) as the protease responsible for cleavage and show that blocking GrB activity stopped FGFR1 trafficking to the nucleus and abrogates the promigratory effect of FGF stimulation. We confirm the in vivo relevance of our findings, showing that FGFR1 localized to the nucleus specifically in invading cells in both clinical material and a three-dimensional model of breast cancer. We identify target genes for FGFR1, which exert significant effects on cell migration and may represent an invasive signature. Our experiments identify a novel mechanism by which FGF signaling can regulate cancer cell behavior and provide a novel therapeutic target for treatment of invasive breast cancer.
Collapse
Affiliation(s)
- Athina-Myrto Chioni
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, England, UK
| | | |
Collapse
|
22
|
Törmä H. Regulation of keratin expression by retinoids. DERMATO-ENDOCRINOLOGY 2011; 3:136-40. [PMID: 22110773 DOI: 10.4161/derm.3.3.15026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 01/25/2011] [Indexed: 11/19/2022]
Abstract
Vitamin A and its natural and synthetic metabolites (retinoids) affect growth and differentiation of human skin and among the genes affected by retinoids in epidermis are keratin genes. Keratins are intermediate filament proteins that have essential functions in maintaining the structural integrity of epidermis and its appendages. Their expressions are under strict control to produce keratins that are optimally adapted to their environment. In this article, retinoid regulation of keratin expression in cultured human epidermal keratinocytes and in human skin in vivo will be reviewed. The direct and indirect mechanisms involved will be discussed and novel therapeutic strategies will be proposed for utilizing retinoids in skin disorders due to keratin mutations (e.g., epidermolysis bullosa simplex and epidermolytic ichthyosis).
Collapse
Affiliation(s)
- Hans Törmä
- Department of Medical Sciences/Dermatology; Uppsala University; Uppsala, Sweden
| |
Collapse
|
23
|
Kerns M, DePianto D, Yamamoto M, Coulombe PA. Differential modulation of keratin expression by sulforaphane occurs via Nrf2-dependent and -independent pathways in skin epithelia. Mol Biol Cell 2010; 21:4068-75. [PMID: 20926689 PMCID: PMC2993737 DOI: 10.1091/mbc.e10-02-0153] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Treatment with the natural chemical sulforaphane (SF) ameliorates skin blistering in keratin 14 (K14)-deficient mice, correlating with the induction of K16 and K17 in the basal layer of epidermis (Kerns et al., PNAS 104:14460, 2007). Here we address the basis for the SF-mediated K16 and K17 induction in mouse epidermis in vivo. As expected, induction of K16 partly depends on the transcription factor Nrf2, which is activated by SF exposure. Strikingly, K17 induction occurs independently of Nrf2 activity and parallels the decrease in glutathione occurring shortly after epidermal exposure to SF. Pharmacological manipulation of glutathione levels in mouse epidermis in vivo alters K17 and K16 expression in the expected manner. We present findings suggesting that select MAP kinases participate in mediating the Nrf2- and glutathione-dependent alterations in K16 and K17 levels in SF-treated epidermis. These findings advance our understanding of the effect of SF on gene expression in epidermis, point to a role for glutathione in mediating some of these effects, and establish that SF induces the expression of two contiguous and highly related genes, K16 and K17, via distinct mechanisms.
Collapse
Affiliation(s)
- Michelle Kerns
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
24
|
Bragulla HH, Homberger DG. Structure and functions of keratin proteins in simple, stratified, keratinized and cornified epithelia. J Anat 2010; 214:516-59. [PMID: 19422428 DOI: 10.1111/j.1469-7580.2009.01066.x] [Citation(s) in RCA: 419] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Historically, the term 'keratin' stood for all of the proteins extracted from skin modifications, such as horns, claws and hooves. Subsequently, it was realized that this keratin is actually a mixture of keratins, keratin filament-associated proteins and other proteins, such as enzymes. Keratins were then defined as certain filament-forming proteins with specific physicochemical properties and extracted from the cornified layer of the epidermis, whereas those filament-forming proteins that were extracted from the living layers of the epidermis were grouped as 'prekeratins' or 'cytokeratins'. Currently, the term 'keratin' covers all intermediate filament-forming proteins with specific physicochemical properties and produced in any vertebrate epithelia. Similarly, the nomenclature of epithelia as cornified, keratinized or non-keratinized is based historically on the notion that only the epidermis of skin modifications such as horns, claws and hooves is cornified, that the non-modified epidermis is a keratinized stratified epithelium, and that all other stratified and non-stratified epithelia are non-keratinized epithelia. At this point in time, the concepts of keratins and of keratinized or cornified epithelia need clarification and revision concerning the structure and function of keratin and keratin filaments in various epithelia of different species, as well as of keratin genes and their modifications, in view of recent research, such as the sequencing of keratin proteins and their genes, cell culture, transfection of epithelial cells, immunohistochemistry and immunoblotting. Recently, new functions of keratins and keratin filaments in cell signaling and intracellular vesicle transport have been discovered. It is currently understood that all stratified epithelia are keratinized and that some of these keratinized stratified epithelia cornify by forming a Stratum corneum. The processes of keratinization and cornification in skin modifications are different especially with respect to the keratins that are produced. Future research in keratins will provide a better understanding of the processes of keratinization and cornification of stratified epithelia, including those of skin modifications, of the adaptability of epithelia in general, of skin diseases, and of the changes in structure and function of epithelia in the course of evolution. This review focuses on keratins and keratin filaments in mammalian tissue but keratins in the tissues of some other vertebrates are also considered.
Collapse
Affiliation(s)
- Hermann H Bragulla
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, 70803, USA.
| | | |
Collapse
|
25
|
Radner FPW, Streith IE, Schoiswohl G, Schweiger M, Kumari M, Eichmann TO, Rechberger G, Koefeler HC, Eder S, Schauer S, Theussl HC, Preiss-Landl K, Lass A, Zimmermann R, Hoefler G, Zechner R, Haemmerle G. Growth retardation, impaired triacylglycerol catabolism, hepatic steatosis, and lethal skin barrier defect in mice lacking comparative gene identification-58 (CGI-58). J Biol Chem 2010; 285:7300-11. [PMID: 20023287 PMCID: PMC2844178 DOI: 10.1074/jbc.m109.081877] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 12/14/2009] [Indexed: 11/06/2022] Open
Abstract
Comparative gene identification-58 (CGI-58), also designated as alpha/beta-hydrolase domain containing-5 (ABHD-5), is a lipid droplet-associated protein that activates adipose triglyceride lipase (ATGL) and acylates lysophosphatidic acid. Activation of ATGL initiates the hydrolytic catabolism of cellular triacylglycerol (TG) stores to glycerol and nonesterified fatty acids. Mutations in both ATGL and CGI-58 cause "neutral lipid storage disease" characterized by massive accumulation of TG in various tissues. The analysis of CGI-58-deficient (Cgi-58(-/-)) mice, presented in this study, reveals a dual function of CGI-58 in lipid metabolism. First, systemic TG accumulation and severe hepatic steatosis in newborn Cgi-58(-/-) mice establish a limiting role for CGI-58 in ATGL-mediated TG hydrolysis and supply of nonesterified fatty acids as energy substrate. Second, a severe skin permeability barrier defect uncovers an essential ATGL-independent role of CGI-58 in skin lipid metabolism. The neonatal lethal skin barrier defect is linked to an impaired hydrolysis of epidermal TG. As a consequence, sequestration of fatty acids in TG prevents the synthesis of acylceramides, which are essential lipid precursors for the formation of a functional skin permeability barrier. This mechanism may also underlie the pathogenesis of ichthyosis in neutral lipid storage disease patients lacking functional CGI-58.
Collapse
Affiliation(s)
- Franz P. W. Radner
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Ingo E. Streith
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Gabriele Schoiswohl
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Martina Schweiger
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Manju Kumari
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Thomas O. Eichmann
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Gerald Rechberger
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | | | - Sandra Eder
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Silvia Schauer
- the Institute of Pathology, Medical University of Graz, A-8010 Graz, and
| | | | - Karina Preiss-Landl
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Achim Lass
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Robert Zimmermann
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Gerald Hoefler
- the Institute of Pathology, Medical University of Graz, A-8010 Graz, and
| | - Rudolf Zechner
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| | - Guenter Haemmerle
- From the Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz
| |
Collapse
|
26
|
Induction of focal epithelial hyperplasia in tongue of young bk6-E6/E7 HPV16 transgenic mice. Transgenic Res 2009; 18:513-27. [PMID: 19165615 DOI: 10.1007/s11248-009-9243-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 01/04/2009] [Indexed: 10/21/2022]
Abstract
Squamous cell carcinoma (SCC) of the oral cavity is one of the most common neoplasms in the world. During the past 2 decades, the role of high-risk human papilloma virus (HR-HPV) has been studied and the data supporting HPV as a one of the causative agents in the development and progression of a sub-set of head and neck squamous cell carcinomas (HNSCC) has accumulated. In order to investigate the role of HR-HPV oncogene expression in early epithelial alterations in vivo, we produced transgenic mice expressing HPV16 early region genes from the promoter of the bovine keratin 6 gene (Tg[bK6-E6/E7]). In this article, we demonstrate that E6/E7 transgene was abundantly expressed and cellular proliferation was increased in the middle tongue epithelia of transgenic mice, and that in the same region young (27 weeks old) Tg[bK6-E6/E7] mice spontaneously developed histological alterations, mainly focal epithelial hyperplasia (FEH).
Collapse
|
27
|
Pavithra L, Singh S, Sreenath K, Chattopadhyay S. Tumor suppressor SMAR1 downregulates Cytokeratin 8 expression by displacing p53 from its cognate site. Int J Biochem Cell Biol 2008; 41:862-71. [PMID: 18822384 DOI: 10.1016/j.biocel.2008.08.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 08/13/2008] [Accepted: 08/21/2008] [Indexed: 11/19/2022]
Abstract
Intermediary filaments play a crucial role in transformation of cells to a malignant phenotype. Here, we report that tumor suppressor SMAR1 downregulates Cytokeratin 8 gene expression by modulating p53-mediated transactivation of this gene. Moreover, the cell surface cytokeratin expression was downregulated leading to a decreased migration and invasiveness of cells. We further validated these results using genotoxic stress agents that lead to an increase in the levels of SMAR1 protein. This subsequently represses the transcription of Cytokeratin 8 gene by local chromatin condensation mediated by histone methylation and deacetylation. Evaluation of SMAR1 and Cytokeratin 8 proteins in different grades of cancer using tissue microarray point out at the inverse expression profiles of these genes (i.e. low levels of SMAR1 correlating with high expression of Cytokeratin 8) in higher grades of breast cancer. Therefore, the results presented here highlight the mechanism of Cytokeratin 8 gene regulation by interplay of tumor suppressor proteins SMAR1 and p53.
Collapse
|
28
|
Human papillomavirus E6/E7 oncogenes promote mouse ear regeneration by increasing the rate of wound re-epithelization and epidermal growth. J Invest Dermatol 2008; 128:2894-903. [PMID: 18548112 DOI: 10.1038/jid.2008.156] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Mammals have limited regeneration capacity. We report here that, in transgenic mice (Tg(bK6-E6/E7)), the expression of the E6/E7 oncogenes of human papilloma virus type 16 (HPV16) under the control of the bovine keratin 6 promoter markedly improves the mouse's capacity to repair portions of the ear after being wounded. Increased repair capacity correlates with an increased number of epidermal proliferating cells. In concordance with the expected effects of the E6 and E7 oncogenes, levels of p53 decreased and those of p16 in epidermal cells increased. In addition, we observed that wound re-epithelization proceeded faster in transgenic than in wild-type animals. After the initial re-epithelization, epidermal cell migration from the intact surrounding tissue appears to be a major contributor to the growing epidermis, especially in the repairing tissue of transgenic mice. We also found that there is a significantly higher number of putative epidermal stem cells in Tg(bK6-E6/E7) than in wild-type mice. Remarkably, hair follicles and cartilage regenerated within the repaired ear tissue, without evidence of tumor formation. We propose that the ability to regenerate ear portions is limited by the capacity of the epidermis to repair itself and grow.
Collapse
|
29
|
Charles RP, Guitard M, Leyvraz C, Breiden B, Haftek M, Haftek-Terreau Z, Stehle JC, Sandhoff K, Hummler E. Postnatal requirement of the epithelial sodium channel for maintenance of epidermal barrier function. J Biol Chem 2007; 283:2622-30. [PMID: 18039670 DOI: 10.1074/jbc.m708829200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In skin, the physiological consequence of an epithelial sodium channel (ENaC) deficiency is not obvious directly at birth. Nevertheless, within hours after birth, mice deficient for the alpha-subunit of the highly amiloride-sensitive epithelial sodium channel (alphaENaC/Scnn1a) suffer from a significant increased dehydration. This is characterized by a loss of body weight (by 6% in 6 h) and an increased transepidermal water loss, which is accompanied by a higher skin surface pH in 1-day-old pups. Although early and late differentiation markers, as well as tight junction protein distribution and function, seem unaffected, deficiency of alphaENaC severely disturbs the stratum corneum lipid composition with decreased ceramide and cholesterol levels, and increased pro-barrier lipids, whereas covalently bound lipids are drastically reduced. Ultrastructural analysis revealed morphological changes in the formation of intercellular lamellar lipids and the lamellar body secretion. Extracellular formation of the lamellar lipids proved to be abnormal in the knockouts. In conclusion, ENaC deficiency results in progressive dehydration and, consequently, weight loss due to severe impairment of lipid formation and secretion. Our data demonstrate that ENaC expression is required for the postnatal maintenance of the epidermal barrier function but not for its generation.
Collapse
Affiliation(s)
- Roch-Philippe Charles
- Département de Pharmacologie & de Toxicologie, Université de Lausanne, Rue du Bugnon 27, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Körver JEM, van Duijnhoven MWFM, Pasch MC, van Erp PEJ, van de Kerkhof PCM. Assessment of epidermal subpopulations and proliferation in healthy skin, symptomless and lesional skin of spreading psoriasis. Br J Dermatol 2007; 155:688-94. [PMID: 16965416 DOI: 10.1111/j.1365-2133.2006.07403.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The margin zone in spreading psoriatic lesions has frequently been used as a model to study the changes in epidermal proliferation, keratinization and inflammation during the transition from symptomless to lesional skin. However, the dynamics of the changes in the epidermal subpopulations-basal cells, transit amplifying cells and differentiated cells-have not been studied in the transition between symptomless and lesional skin. OBJECTIVES To quantify in a dynamic model of the margin zone in psoriasis the characteristics of these subpopulations with respect to epidermal proliferation and differentiation. METHODS From seven patients with active psoriasis, biopsies were taken from the distant uninvolved skin, outer margin, inner margin and centre of a spreading psoriatic plaque. Frozen sections were labelled immunofluorescently using direct immunofluorescence for Ki-67 and beta1 integrin and the Zenon labelling technique for keratin 6, 10 and 15. Digital photographs of the stained sections were quantitatively analysed. RESULTS In the distant uninvolved skin the expression of beta1 integrin was decreased and keratin 15 expression was lost. In this area suprabasal cells expressed beta1 integrin and in the outer margin suprabasal cells expressed Ki-67. From the outer to the inner margin of the psoriasis plaque, which coincided with the appearance of the clinical lesion, there was a significant change in the various markers. The patchy expression of keratin 6 in the inner margin became homogeneous in the centre of the psoriasis plaque and here was also coexpression of keratin 6 and keratin 10 in a single cell. CONCLUSIONS The present study provides additional evidence that the distant uninvolved skin has a prepsoriatic phenotype, which is the first step in a psoriatic cascade. The cascade between symptomless and lesional skin comprises first an abnormality in inflammation with involvement of beta1 integrin-dim cells (transit amplifying cells) subsequently eliciting an enlarged germinative compartment with increased recruitment of cycling epidermal cells and focal expression of proliferation-associated keratins, ultimately culminating in a more-or-less homogeneous epidermis with massive recruitment of cycling epidermal cells and proliferation-associated keratinization.
Collapse
Affiliation(s)
- J E M Körver
- Department of Dermatology, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands.
| | | | | | | | | |
Collapse
|
31
|
Rodgers KE, Ellefson DD, Espinoza T, Hsu YH, diZerega GS, Mehrian-Shai R. Expression of intracellular filament, collagen, and collagenase genes in diabetic and normal skin after injury. Wound Repair Regen 2006; 14:298-305. [PMID: 16808808 DOI: 10.1111/j.1743-6109.2006.00124.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Reports have shown differences in gene expression in the skin of diabetic and normal mice both at baseline and after injury. Cluster analysis identified distinct expression patterns within intermediate filaments and extracellular proteins. This report addresses the effect of diabetes and injury on the expression of keratin-associated proteins, keratin complexes, procollagen, and collagenase (matrix metalloproteinase; MMP) genes. At baseline keratin-associated proteins and keratin complexes gene expression was increased in diabetic mice. After surgery, the level of expression for keratin-associated proteins and keratin complexes genes decreased in diabetic mice, but did not change in normal mice. If the expression of a procollagen gene differed between diabetic and normal mice, the expression was lower in diabetic mice. Procollagen gene expression was elevated after skin excision compared with noninjured skin. At baseline, the level of MMP and tissue inhibitor of metalloproteinase gene expression was comparable between mouse strains. With injury, the expression of several MMP genes was increased in both mouse strains, but to higher levels in diabetic mice. At day 7, the level of MMP-9 activity in granulation tissue was elevated. This alteration may contribute to delayed healing in diabetic mice. Therefore, differences in gene expression exist between mouse strains and can assist in understanding of physiological manifestations, including delayed healing, in diabetic mice.
Collapse
Affiliation(s)
- Kathleen E Rodgers
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California 90033, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Sadagurski M, Yakar S, Weingarten G, Holzenberger M, Rhodes CJ, Breitkreutz D, Leroith D, Wertheimer E. Insulin-like growth factor 1 receptor signaling regulates skin development and inhibits skin keratinocyte differentiation. Mol Cell Biol 2006; 26:2675-87. [PMID: 16537911 PMCID: PMC1430337 DOI: 10.1128/mcb.26.7.2675-2687.2006] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The insulin-like growth factor 1 receptor (IGF-1R) is a multifunctional receptor that mediates signals for cell proliferation, differentiation, and survival. Genetic experiments showed that IGF-1R inactivation in skin results in a disrupted epidermis. However, because IGF-1R-null mice die at birth, it is difficult to study the effects of IGF-1R on skin. By using a combined approach of conditional gene ablation and a three-dimensional organotypic model, we demonstrate that IGF-1R-deficient skin cocultures show abnormal maturation and differentiation patterns. Furthermore, IGF-1R-null keratinocytes exhibit accelerated differentiation and decreased proliferation. Investigating the signaling pathway downstream of IGF-1R reveals that insulin receptor substrate 2 (IRS-2) overexpression compensates for the lack of IGF-1R, whereas IRS-1 overexpression does not. We also demonstrate that phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1 and 2 are involved in the regulation of skin keratinocyte differentiation and take some part in mediating the inhibitory signal of IGF-1R on differentiation. In addition, we show that mammalian target of rapamycin plays a specific role in mediating IGF-1R impedance of action on keratinocyte differentiation. In conclusion, these results reveal that IGF-1R plays an inhibitory role in the regulation of skin development and differentiation.
Collapse
Affiliation(s)
- Marianna Sadagurski
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Patel GK, Wilson CH, Harding KG, Finlay AY, Bowden PE. Numerous keratinocyte subtypes involved in wound re-epithelialization. J Invest Dermatol 2006; 126:497-502. [PMID: 16374449 DOI: 10.1038/sj.jid.5700101] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The expression of different keratin intermediate filaments has been used to define keratinocyte maturation and different phenotypic subtypes involved in acute wound (AW) healing. Immunohistochemistry with specific anti-keratin monoclonal and polyclonal antibodies was used to examine AW in normal healthy volunteers (n = 16). In all wounds examined, basal keratinocytes and cells at the leading edge of the wound expressed keratins K5 and K14. However, suprabasal cells had a more complex pattern of keratin expression, which was dependent on their position relative to the wound and location within the suprabasal compartment of the epidermis. In general, K10 was expressed in suprabasal keratinocytes at the wound edge, but not in keratinocytes covering the wound center, which expressed K6, K16, and K17 in a complex fashion. Ki67 expression, a marker of cell proliferation, was restricted to basal and immediate suprabasal layers at the wound edge. Keratinocytes populated the wound bed below the scab by migration, which was supported by keratinocyte proliferation in the surrounding epidermis both at and adjacent to the wound edge.
Collapse
Affiliation(s)
- Girish K Patel
- Department of Dermatology, School of Medicine,Cardiff University, Cardiff, UK
| | | | | | | | | |
Collapse
|
34
|
Haass NK, Wladykowski E, Kief S, Moll I, Brandner JM. Differential induction of connexins 26 and 30 in skin tumors and their adjacent epidermis. J Histochem Cytochem 2005; 54:171-82. [PMID: 16046668 DOI: 10.1369/jhc.5a6719.2005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Gap junctions (GJs) have been shown to play a role in tumor progression including a variety of keratinocyte-derived and non-keratinocyte-derived skin tumors. Here we show that the synthesis of the GJ proteins connexin 26 and connexin 30 (Cx26 and Cx30) is induced in keratinocyte-derived epithelial skin tumors whereas there is either no change or a downregulation of Cx43. Cx26, Cx30, and Cx43 are absent in non-epithelial skin tumors. Further, Cx26 and Cx30 are induced in the epidermis adjacent to malignant melanoma but absent in the epidermis adjacent to benign non-epithelial skin lesions (melanocytic nevi and angioma). The keratinocyte-derived skin tumors are very heterogeneous regarding the Cx26/Cx30 pattern in the epidermis at the periphery of the tumors. We did not observe any difference in the localization of the very similar proteins Cx26 and Cx30 but a variation in intensity of immunoreactivity. As the staining patterns of Cx26 and Cx30 antibodies are not identical to those of CK6, a marker for hyperproliferation, and CK17, a marker for trauma, we discuss that the induction of these gap junctional proteins exceeds a reflection of reactive hyperproliferative or traumatized epidermis. We further discuss the putative roles of these gap junctional proteins in tumor progression.
Collapse
Affiliation(s)
- Nikolas K Haass
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA.
| | | | | | | | | |
Collapse
|
35
|
Boniface K, Bernard FX, Garcia M, Gurney AL, Lecron JC, Morel F. IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2005; 174:3695-702. [PMID: 15749908 DOI: 10.4049/jimmunol.174.6.3695] [Citation(s) in RCA: 649] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-22 belongs to a family of cytokines structurally related to IL-10, including IL-19, IL-20, IL-24, and IL-26. In contrast to IL-10, IL-22 has proinflammatory activities. IL-22 signals through a class II cytokine receptor composed of an IL-22-binding chain, IL-22RA1, and the IL-10RB subunit, which is shared with the IL-10R. In the present study, we show that short-term cultured human epidermal keratinocytes express a functional IL-22R but no IL-10R. Accordingly, IL-22 but not IL-10 induces STAT3 activation in keratinocytes. Using a cDNA array screening approach, real-time RT-PCR, and Western blot analysis, we demonstrate that IL-22 up-regulates, in a dose-dependent manner, the expression of S100A7, S100A8, S100A9, a group of proinflammatory molecules belonging to the S100 family of calcium-binding proteins, as well as the matrix metalloproteinase 3, the platelet-derived growth factor A, and the CXCL5 chemokine. In addition, IL-22 induces keratinocyte migration in an in vitro injury model and down-regulates the expression of at least seven genes associated with keratinocyte differentiation. Finally, we show that IL-22 strongly induces hyperplasia of reconstituted human epidermis. Taken together, these results suggest that IL-22 plays an important role in skin inflammatory processes and wound healing.
Collapse
Affiliation(s)
- Katia Boniface
- Laboratoire Cytokines et Inflammation, UPRES EA 3806, Pôle Biologie Santé, Université de Poitiers, Poitiers, France
| | | | | | | | | | | |
Collapse
|
36
|
Karchoudhury TK. Disruption of hydrophobic stability of biomembranes is the earliest event in several clinical disorders. Med Hypotheses 2004; 63:502-6. [PMID: 15288377 DOI: 10.1016/j.mehy.2004.02.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2003] [Accepted: 02/25/2004] [Indexed: 10/26/2022]
Abstract
Hydrophobic attractive force is the major force in maintaining the stability of biomembranes, yielding coordinated functionality to the embedded proteins that they contain. This force between the composite linear hydrocarbons of the biomembranes is a function of their length and their mutual parallel distance from each other, and is extremely sensitive to this distance. Extracellular, natural linear hydrocarbons of certain length and shape can intercalate into lipid matrix of the biomembranes, reducing their innate hydrophobic net strength in a concentration-dependent manner, making them loose, leaky, and thus gaining the credence of stimulus-generating agents. In physiological circulatory concentration, these molecules may have a role for the maintenance of cellular homeostasis. However, in stagnating physiological excess, these same agents can become acutely or chronically stimulating and, therefore, disease-precipitating. Such situations do exist in the clinical disorders of acne, atherosclerosis, acute pancreatitis, diabetes, diabetic retinopathy, homocysteinemea, and stress. A systematic approach, beginning with surface film studies with the suspect linear hydrocarbons, can be followed up with in vitro and in vivo studies. This should substantiate or negate the view presented here. Isolated information, along these lines, already exist in literature. The example of acne is a suitable starting point to elaborate this view, for sebaceous gland of the human pilosebaceous unit (PSU) contains all the exemplary, stimulus-inducing linear hydrocarbons to generate surface-reaction on the pilosebaceous ductal surface.
Collapse
|
37
|
Schmuth M, Elias PM, Hanley K, Lau P, Moser A, Willson TM, Bikle DD, Feingold KR. The Effect of LXR Activators on AP-1 Proteins in Keratinocytes. J Invest Dermatol 2004; 123:41-8. [PMID: 15191540 DOI: 10.1111/j.0022-202x.2004.22707.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oxysterols, via activation of liver X receptor (LXR), regulate keratinocyte differentiation by stimulating transglutaminase cross-linking of several constituent proteins leading to the formation of the cornified envelope. We previously reported that oxysterols increase the expression of one of these cross-linked proteins, involucrin, and that this effect can be abolished by mutations of the distal activator protein (AP)-1 response element in the involucrin promoter. Furthermore, oxysterols increase AP-1 binding in an electrophoretic gel mobility shift assay and increase the expression of an AP-1 reporter. In this study, we describe the individual components of the AP-1 complex that are involved in the oxysterol-mediated AP-1 activation and stimulation of keratinocyte differentiation. We identified Fra-1 within the AP-1 DNA binding complex by supershift analysis of nuclear extracts from oxysterol-treated, cultured keratinocytes and confirmed that oxysterol treatment increased the levels of Fra-1 by western blot analysis. Additionally, on Western and Northern analysis, oxysterol treatment increased two other AP-1 proteins, Jun-D and c-Fos, whereas Fra-2, Jun-B, and c-Jun were not changed. Similar alterations in AP-1 proteins occurred when 25-OH-cholesterol or non-steroidal LXR agonists (GW3965, TO-901317) were used. These results indicate that oxysterols induce specific AP-1 proteins, thereby activating involucrin, one of the genes required for epidermal differentiation.
Collapse
Affiliation(s)
- Matthias Schmuth
- Department of Medicine, University of California, San Francisco, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Herrmann H, Hesse M, Reichenzeller M, Aebi U, Magin TM. Functional complexity of intermediate filament cytoskeletons: from structure to assembly to gene ablation. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 223:83-175. [PMID: 12641211 DOI: 10.1016/s0074-7696(05)23003-6] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The cell biology of intermediate filament (IF) proteins and their filaments is complicated by the fact that the members of the gene family, which in humans amount to at least 65, are differentially expressed in very complex patterns during embryonic development. Thus, different tissues and cells express entirely different sets and amounts of IF proteins, the only exception being the nuclear B-type lamins, which are found in every cell. Moreover, in the course of evolution the individual members of this family have, within one species, diverged so much from each other with regard to sequence and thus molecular properties that it is hard to envision a unifying kind of function for them. The known epidermolytic diseases, caused by single point mutations in keratins, have been used as an argument for a role of IFs in mechanical "stress resistance," something one would not have easily ascribed to the beaded chain filaments, a special type of IF in the eye lens, or to nuclear lamins. Therefore, the power of plastic dish cell biology may be limited in revealing functional clues for these structural elements, and it may therefore be of interest to go to the extreme ends of the life sciences, i.e., from the molecular properties of individual molecules including their structure at the atomic level to targeted inactivation of their genes in living animals, mouse, and worm to define their role more precisely in metazoan cell physiology.
Collapse
Affiliation(s)
- Harald Herrmann
- Division of Cell Biology, German Cancer Research Center, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
39
|
Pol A, Pfundt R, Zeeuwen P, Molhuizen H, Schalkwijk J. Transcriptional regulation of the elafin gene in human keratinocytes. J Invest Dermatol 2003; 120:301-7. [PMID: 12542536 DOI: 10.1046/j.1523-1747.2003.12043.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Elafin (also known as skin-derived anti-leukoproteinase/trappin-2) is an epithelial host-defense protein that is absent in normal skin but highly induced in keratinocytes of inflamed skin (e.g., psoriasis), in epidermal skin tumors, and after wounding. Previously, it was shown that in cultured keratinocytes, elafin expression is induced by serum or tumor necrosis factor-alpha, and that expression is suppressed by retinoids, dithranol, and p38 mitogen-activated protein kinase inhibitors. Here we have studied the regulation of elafin gene expression in epidermal keratinocytes at the molecular level. First we determined the transcription start site of the elafin gene and found that the elafin mRNA possesses an unusually short 5'-untranslated region. Using transient transfection of luciferase reporter constructs of the elafin promoter, we mapped a 440 bp region upstream of the translation start site that conferred high-level expression in keratinocytes, but not in A431 cells or cells of mesenchymal origin. We observed that the promoter constructs were not subjected to the same regulation as the endogenous elafin gene as these constructs were highly active independent of keratinocyte stimulation. When elafin promoter constructs were stably transfected in the HaCaT keratinocyte cell line, tumor necrosis factor-alpha inducible expression of both the endogenous elafin gene and the transgene was observed, suggesting that regulation of the elafin gene is also dependent on chromatin structure. We found, however, that a stably transfected 4 kb elafin promoter fragment did not confer retinoid sensitivity indicating that additional sequences are required for proper regulation. This study reveals the complex regulation of a gene that can be used as a paradigm for the specific differentiation program of activated epidermal keratinocytes.
Collapse
Affiliation(s)
- Arno Pol
- Department of Dermatology, University Medical Center St Radboud, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
40
|
Hattori N, Komine M, Yano S, Kaneko T, Hanakawa Y, Hashimoto K, Tamaki K. Interferon-gamma, a strong suppressor of cell proliferation, induces upregulation of keratin K6, one of the inflammatory- and proliferation-associated keratins. J Invest Dermatol 2002; 119:403-10. [PMID: 12190863 DOI: 10.1046/j.1523-1747.2002.01843.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Keratin K6 is known as an inflammatory and hyperproliferative keratin, and is induced by an inflammatory and hyperproliferative agent. In this study, we demonstrated that interferon-gamma, an antiproliferative agent, also induces keratin K6. We used normal human ex vivo skin, normal human cultured keratinocytes, HaCaT keratinocytes, and DJM cells to examine the induction of K6 by interferon-gamma, by immunohistochemical staining, Western blot analysis, promoter chloramphenicol acetyl transferase assay, and reverse transcriptase polymerase chain reaction of mRNA. We succeeded in demonstrating the induction of keratin K6 by interferon-gamma in ex vivo human skin and HaCaT keratinocytes at the protein and message level, and in cultured normal human keratinocytes at the promoter level. The inhibition of the signal transducing activator of transcription 1 pathway by a dominant-negative transfer gene caused the inhibition of K6 induction by interferon-gamma, and the blocking of nuclear factor kappaB using antisense oligonucleotides also inhibited the K6 induction. We also blocked the released interleukin-1alpha from keratinocytes after stimulation with interferon-gamma by neutralizing antibodies, which showed a decrease in the K6 induction. Our results suggest that a small amount of interleukin-1alpha, which cannot induce K6 by itself, is secreted upon stimulation by interferon-gamma, and that the induction of K6 occurs through the synergistic effect of the interferon-gamma/signal transducing activator of transcription 1 and interleukin-1alpha/nuclear factor kappaB pathways. This is the first report to describe K6 induction in epidermal keratinocytes by interferon-gamma and indicate a probable signal transduction pathway, and demonstrates that K6 is a possible partner of K17 in the inflammatory process.
Collapse
Affiliation(s)
- Naoko Hattori
- Department of Dermatology, Faculty of Medicine, University of Tokyo, and University of Ehime, Japan.
| | | | | | | | | | | | | |
Collapse
|
41
|
Sonis ST, Scherer J, Phelan S, Lucey CA, Barron JE, O'Donnell KE, Brennan RJ, Pan H, Busse P, Haley JD. The gene expression sequence of radiated mucosa in an animal mucositis model. Cell Prolif 2002; 35 Suppl 1:93-102. [PMID: 12139712 PMCID: PMC6496665 DOI: 10.1046/j.1365-2184.35.s1.10.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Oral mucositis is a common, dose-limiting, acute toxicity of radiation therapy administered for the treatment of cancers of the head and neck. Accumulating data would suggest that the pathogenesis of mucositis is complex and involves the sequential interaction of all cell types of the oral mucosa, as well as a number of cytokines and elements of the oral environment. While a number of studies have reported on gene expression of particular cell types in response to radiation, the overall response of irradiated mucosa has only been evaluated in a limited way. The present study was undertaken to evaluate the expression of a target group of genes using RNA quantification assays and, more broadly, to assess patterns of mucosal gene expression using DNA microarray hybridization. Our results demonstrate the sequential upregulation of a series of genes that, when taken collectively, suggest an intricate functional interaction.
Collapse
Affiliation(s)
- S T Sonis
- Division of Oral Medicine, Oral and Maxillofacial Surgery and Dentistry, Brigham and Women's Hospital and the Department of Otal Medicine and Diagnostic Sciences, Harvard School of Dental Medicine, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Turchi L, Chassot AA, Rezzonico R, Yeow K, Loubat A, Ferrua B, Lenegrate G, Ortonne JP, Ponzio G. Dynamic characterization of the molecular events during in vitro epidermal wound healing. J Invest Dermatol 2002; 119:56-63. [PMID: 12164925 DOI: 10.1046/j.1523-1747.2002.01805.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The aim of this study was to characterize some of the molecular events stimulated in vitro in response to injury within a confluent culture of normal epidermal keratinocytes as a model to understand the mechanisms of wound healing. To this end, an original device was developed specifically designed to perform calibrated injuries of great lengths within mono-stratified or pluri-stratified keratinocyte cultures. The experiments performed in this study validate this device as an appropriate tool for studying epidermal wound healing; this is because it performs mechanical injuries that stimulate the expression of multiple healing markers also known to be upregulated during wound healing in vivo (growth factors, cytokines, proteinases, extracellular matrix proteins). Using this device, it was demonstrated in human keratinocytes: mechanical injuries (i) immediately stimulate the tyrosine phosphorylation of numerous cellular proteins; (ii) induce molecular cascades leading to the activation of p21ras, mitogen-activated protein kinases, extracellular signal-regulated kinases 1/2, c-Jun NH2 terminal kinase, and p38 mitogen-activated protein kinase; and (iii) increase the phosphorylation of their respective substrates, c-jun and activator transcription factor 1. Wounding of these cells also results in increases in the DNA binding activities of several jun/fos activator protein-1 transcription factor complexes. It is important to note that the development of an appropriate wounding system was essential for performing this study, as use of a classical wounding procedure did not enable the detection of the biologic parameters reported above. In conclusion, these data indicate that using the appropriate system, it is possible to identify the signaling pathways activated in normal human keratinocyte cells after injury. In this study, it was shown that the mitogen-activated protein kinase pathways and activator protein-1 are stimulated in response to physical injury, and may be involved in regulating the expression of healing markers.
Collapse
Affiliation(s)
- Laurent Turchi
- INSERM U 385, "Biologie et Physiopathologie de la Peau", Faculté de Médecine, Avenue de Valombrose, Nice cedex 02, France
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sesto A, Navarro M, Burslem F, Jorcano JL. Analysis of the ultraviolet B response in primary human keratinocytes using oligonucleotide microarrays. Proc Natl Acad Sci U S A 2002; 99:2965-70. [PMID: 11867738 PMCID: PMC122456 DOI: 10.1073/pnas.052678999] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2001] [Accepted: 12/17/2001] [Indexed: 02/06/2023] Open
Abstract
UV radiation is the most important environmental skin aggressor, causing cancer and other problems. This paper reports the use of oligonucleotide microarray technology to determine changes in gene expression in human keratinocytes after UVB treatment. Examination of the effects of different doses at different times after irradiation gave a global picture of the keratinocyte response to this type of insult. Five hundred thirty-nine regulated transcripts were found and organized into nine different clusters depending on behavior patterns. Classification of these genes into 23 functional categories revealed that several biological processes are globally affected by UVB. In addition to confirming a majority up-regulation of the transcripts related to the UV-specific inflammatory and stress responses, significant increases were seen in the expression of genes involved in basal transcription, splicing, and translation as well as in the proteasome-mediated degradation category. On the other hand, those transcripts belonging to the metabolism and adhesion categories were strongly downregulated. These results demonstrate the complexity of the transcriptional profile of the UVB response, describe several cellular processes previously not known to be affected by UV irradiation, and serve as a basis for the global characterization of UV-regulated genes and pathways.
Collapse
Affiliation(s)
- Angela Sesto
- Department of Molecular and Cell Biology, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Avenida Complutense 22, 28040 Madrid, Spain
| | | | | | | |
Collapse
|
44
|
Bernerd F, Del Bino S, Asselineau D. Regulation of keratin expression by ultraviolet radiation: differential and specific effects of ultraviolet B and ultraviolet a exposure. J Invest Dermatol 2001; 117:1421-9. [PMID: 11886503 DOI: 10.1046/j.0022-202x.2001.01589.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Skin, the most superficial tissue of our body, is the first target of environmental stimuli, among which is solar ultraviolet radiation. Very little is known about the regulation of keratin gene expression by ultraviolet radiation, however, although (i) it is well established that ultraviolet exposure is involved in skin cancers and photoaging and (ii) keratins represent the major epidermal proteins. The aim of this study was to analyze the regulation of human keratin gene expression under ultraviolet B (290-320 nm) or ultraviolet A (320-400 nm) irradiation using a panel of constructs comprising different human keratin promoters cloned upstream of a chloramphenicol acetyl transferase reporter gene and transfected into normal epidermal keratinocytes. By this approach, we demonstrated that ultraviolet B upregulated the transcription of keratin 19 gene and to a lesser extent the keratin 6, keratin 5, and keratin 14 genes. The DNA sequence responsible for keratin 19 induction was localized between -130 and +1. In contrast to ultraviolet B, ultraviolet A irradiation induced only an increase in keratin 17, showing a differential gene regulation between these two ultraviolet ranges. The induction of keratin 19 was confirmed by studying the endogenous protein in keratinocytes in classical cultures as well as in skin reconstructed in vitro and normal human skin. These data show for the first time that keratin gene expression is regulated by ultraviolet radiation at the transcriptional level with a specificity regarding the ultraviolet domain of solar light.
Collapse
Affiliation(s)
- F Bernerd
- L'Oréal, Life Sciences Research, Clichy, France.
| | | | | |
Collapse
|
45
|
Presland RB, Tomic-Canic M, Lewis SP, Dale BA. Regulation of human profilaggrin promoter activity in cultured epithelial cells by retinoic acid and glucocorticoids. J Dermatol Sci 2001; 27:192-205. [PMID: 11641059 DOI: 10.1016/s0923-1811(01)00136-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vitamin A and other retinoids profoundly inhibit both morphological and biochemical aspects of epidermal differentiation in vitro. Profilaggrin, like most other markers of keratinocyte differentiation, is negatively regulated by retinoic acid in vitro, both at the level of mRNA synthesis and by inhibiting the activity of endoproteases that convert profilaggrin to filaggrin. Profilaggrin is an abundant component of keratohyalin granules and forms the precursor of filaggrin, the keratin associated protein of the stratum corneum. In this report, we identify a region of the human profilaggrin promoter that is involved in the transcriptional regulation of expression by retinoic acid (RA). A series of promoter deletions linked to the chloramphenicol acetyl transferase (CAT) reporter gene were prepared and analyzed by transfection into Hela cells and keratinocytes. We also cotransfected vectors expressing retinoic acid receptor and cultured the transfected cells in the presence and absence of ligand. The region responsive to retinoic acid was localized to a 53 bp sequence between -1109 and -1056 (relative to the mRNA start site at +1) that contains a cluster of five retinoic acid response elements with variable spacing and orientation. In vitro gel shift analysis demonstrated that nuclear retinoid receptors do not bind directly to the identified sequence, suggesting that the mode of regulation by RA may be indirect or that binding requires another cofactor in addition to retinoid receptors. Whereas in keratin genes retinoic acid and glucocorticoid responsive sequences frequently coincide, the glucocorticoid response element in the profilaggrin promoter was located downstream of the RARE cluster between -965 and -951. These studies demonstrate that RA and glucocorticoids regulate profilaggrin expression at least in part by transcriptional mechanisms, via a region of the promoter that contains both retinoid and glucocorticoid responsive elements.
Collapse
Affiliation(s)
- R B Presland
- Department of Oral Biology, University of Washington, Box 357132, Seattle, WA 98195-7132, USA.
| | | | | | | |
Collapse
|
46
|
Michalik L, Desvergne B, Tan NS, Basu-Modak S, Escher P, Rieusset J, Peters JM, Kaya G, Gonzalez FJ, Zakany J, Metzger D, Chambon P, Duboule D, Wahli W. Impaired skin wound healing in peroxisome proliferator-activated receptor (PPAR)alpha and PPARbeta mutant mice. J Cell Biol 2001; 154:799-814. [PMID: 11514592 PMCID: PMC2196455 DOI: 10.1083/jcb.200011148] [Citation(s) in RCA: 316] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We show here that the alpha, beta, and gamma isotypes of peroxisome proliferator-activated receptor (PPAR) are expressed in the mouse epidermis during fetal development and that they disappear progressively from the interfollicular epithelium after birth. Interestingly, PPARalpha and beta expression is reactivated in the adult epidermis after various stimuli, resulting in keratinocyte proliferation and differentiation such as tetradecanoylphorbol acetate topical application, hair plucking, or skin wound healing. Using PPARalpha, beta, and gamma mutant mice, we demonstrate that PPARalpha and beta are important for the rapid epithelialization of a skin wound and that each of them plays a specific role in this process. PPARalpha is mainly involved in the early inflammation phase of the healing, whereas PPARbeta is implicated in the control of keratinocyte proliferation. In addition and very interestingly, PPARbeta mutant primary keratinocytes show impaired adhesion and migration properties. Thus, the findings presented here reveal unpredicted roles for PPARalpha and beta in adult mouse epidermal repair.
Collapse
Affiliation(s)
- L Michalik
- Institut de Biologie Animale, Université de Lausanne, Bâtiment de Biologie, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wojcik SM, Longley MA, Roop DR. Discovery of a novel murine keratin 6 (K6) isoform explains the absence of hair and nail defects in mice deficient for K6a and K6b. J Cell Biol 2001; 154:619-30. [PMID: 11489919 PMCID: PMC2196416 DOI: 10.1083/jcb.200102079] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The murine genome is known to have two keratin 6 (K6) genes, mouse K6 (MK6)a and MK6b. These genes display a complex expression pattern with constitutive expression in the epithelia of oral mucosa, hair follicles, and nail beds. We generated mice deficient for both genes through embryonic stem cell technology. The majority of MK6a/b-/- mice die of starvation within the first two weeks of life. This is due to a localized disintegration of the dorsal tongue epithelium, which results in the build up of a plaque of cell debris that severely impairs feeding. However, approximately 25% of MK6a/b-/- mice survive to adulthood. Remarkably, the surviving MK6a/b-/- mice have normal hair and nails. To our surprise, we discovered MK6 staining both in the hair follicle and the nail bed of MK6a/b-/- mice, indicating the presence of a third MK6 gene. We cloned this previously unknown murine keratin gene and found it to be highly homologous to human K6hf, which is expressed in hair follicles. We therefore termed this gene MK6 hair follicle (MK6hf). The presence of MK6hf in the MK6a/b-/- follicles and nails offers an explanation for the absence of hair and nail defects in MK6a/b-/- animals.
Collapse
Affiliation(s)
- S M Wojcik
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | |
Collapse
|
48
|
Rangarajan A, Talora C, Okuyama R, Nicolas M, Mammucari C, Oh H, Aster JC, Krishna S, Metzger D, Chambon P, Miele L, Aguet M, Radtke F, Dotto G. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J 2001; 20:3427-36. [PMID: 11432830 PMCID: PMC125257 DOI: 10.1093/emboj/20.13.3427] [Citation(s) in RCA: 670] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The role of Notch signaling in growth/differentiation control of mammalian epithelial cells is still poorly defined. We show that keratinocyte-specific deletion of the Notch1 gene results in marked epidermal hyperplasia and deregulated expression of multiple differentiation markers. In differentiating primary keratinocytes in vitro endogenous Notch1 is required for induction of p21WAF1/Cip1 expression, and activated Notch1 causes growth suppression by inducing p21WAF1/Cip1 expression. Activated Notch1 also induces expression of 'early' differentiation markers, while suppressing the late markers. Induction of p21WAF1/Cip1 expression and early differentiation markers occur through two different mechanisms. The RBP-Jkappa protein binds directly to the endogenous p21 promoter and p21 expression is induced specifically by activated Notch1 through RBP-Jkappa-dependent transcription. Expression of early differentiation markers is RBP-Jkappa-independent and can be induced by both activated Notch1 and Notch2, as well as the highly conserved ankyrin repeat domain of the Notch1 cytoplasmic region. Thus, Notch signaling triggers two distinct pathways leading to keratinocyte growth arrest and differentiation.
Collapse
Affiliation(s)
- Annapoorni Rangarajan
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Claudio Talora
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Ryuhei Okuyama
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Michael Nicolas
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Cristina Mammucari
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Heysun Oh
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Jon C. Aster
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Sudhir Krishna
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Daniel Metzger
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Pierre Chambon
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Lucio Miele
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Michel Aguet
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - Freddy Radtke
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| | - G.Paolo Dotto
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, Pathology Department, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, Cardinal Bernardin Cancer Center, Loyola University, Chicago, IL, USA, National Centre for Biological Sciences, TIFR, Bangalore, India, Swiss Institute of Experimental Cancer Research, Epalinges, Ludwig Institute Cancer Research, Epalinges, Switzerland and Institut de Génétique, Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France Corresponding authors e-mail: or A.Rangarajan, C.Talora, R.Okuyama and M.Nicolas contributed equally to this work
| |
Collapse
|
49
|
Peters B, Kirfel J, Büssow H, Vidal M, Magin TM. Complete cytolysis and neonatal lethality in keratin 5 knockout mice reveal its fundamental role in skin integrity and in epidermolysis bullosa simplex. Mol Biol Cell 2001; 12:1775-89. [PMID: 11408584 PMCID: PMC37340 DOI: 10.1091/mbc.12.6.1775] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
In human patients, a wide range of mutations in keratin (K) 5 or K14 lead to the blistering skin disorder epidermolysis bullosa simplex. Given that K14 deficiency does not lead to the ablation of a basal cell cytoskeleton because of a compensatory role of K15, we have investigated the requirement for the keratin cytoskeleton in basal cells by inactivating the K5 gene in mice. We report that the K5(-/-) mice die shortly after birth, lack keratin filaments in the basal epidermis, and are more severely affected than K14(-/-) mice. In contrast to the K14(-/-) mice, we detected a strong induction of the wound-healing keratin K6 in the suprabasal epidermis of cytolyzed areas of postnatal K5(-/-) mice. In addition, K5 and K14 mice differed with respect to tongue lesions. Moreover, we show that in the absence of K5 and other type II keratins, residual K14 and K15 aggregated along hemidesmosomes, demonstrating that individual keratins without a partner are stable in vivo. Our data indicate that K5 may be the natural partner of K15 and K17. We suggest that K5 null mutations may be lethal in human epidermolysis bullosa simplex patients.
Collapse
Affiliation(s)
- B Peters
- Institut fuer Genetik, Abteilung Molekulargenetik, Rheinische Friedrich-Wilhelms-Universitaet, 53117 Bonn, Germany
| | | | | | | | | |
Collapse
|
50
|
Angel P, Szabowski A, Schorpp-Kistner M. Function and regulation of AP-1 subunits in skin physiology and pathology. Oncogene 2001; 20:2413-23. [PMID: 11402337 DOI: 10.1038/sj.onc.1204380] [Citation(s) in RCA: 317] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mouse skin has become the model of choice to study the regulation and function of AP-1 subunits in many physiological and pathological processes in vivo and in vitro. Genetically modified mice, in vitro reconstituted skin equivalents and epidermal cell lines were established, in which AP-1-regulated genetic programs of cell proliferation, differentiation and tumorigenesis can be analysed. Since the epidermis, as our interface with the environment, is subjected to radiation and injury, signal transduction pathways and critical AP-1 members regulating the mammalian stress response could be identified. Regulated expression of important components of the cytokine network, cell surface receptors and proteases, which orchestrate the process of wound healing has been found to rely on AP-1 activity. Here we review our current knowledge on the function of AP-1 subunits and AP-1 target genes in these fascinating fields of skin physiology and pathology.
Collapse
Affiliation(s)
- P Angel
- Deutsches Krebsforschungszentrum, Division of Signal Transduction and Growth Control, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | | | | |
Collapse
|