Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 137(7): 1501-1511, 2017 07.
Article in English | MEDLINE | ID: mdl-28274739

ABSTRACT

TRAF3IP2 is a candidate psoriasis susceptibility gene encoding Act1, an adaptor protein with ubiquitin ligase activity that couples the IL-17 receptor to downstream signaling pathways. We investigated the role of Act1 in keratinocyte responses to IL-17 using a tetracycline inducible short hairpin RNA targeting TRAF3IP2. Tetracycline exposure for 7 days effectively silenced TRAF3IP2 mRNA and Act1 protein, resulting in 761 genes with significant changes in expression (495 down, 266 up; >1.5-fold, P < 0.05). Gene ontology analysis showed that genes affected by TRAF3IP2 silencing are involved in epidermal differentiation, with early differentiation genes (KRT1, KRT10, DSC1, DSG1) being down-regulated and late differentiation genes (SPRR2, SPRR3, LCE3) being up-regulated. AP1 binding sites were enriched upstream of genes up-regulated by TRAF3IP2 silencing. Correspondingly, nuclear expression of FosB and Fra1 was increased in TRAF3IP2-silenced cells. Many genes involved in host defense were induced by IL-17 in a TRAF3IP2-dependent fashion. Inflammatory differentiation conditions (serum addition for 4 days postconfluence) markedly amplified these IL-17 responses and increased basal levels and TRAF3IP2 silencing-dependent up-regulation of multiple late differentiation genes. These findings suggest that TRAF3IP2 may alter both epidermal homeostasis and keratinocyte defense responses to influence psoriasis risk.


Subject(s)
Connexin 43/metabolism , Gene Expression Regulation , Interleukin-17/metabolism , Keratinocytes/metabolism , Peptide Fragments/metabolism , Psoriasis/genetics , RNA/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Adaptor Proteins, Signal Transducing , Cell Differentiation , Cells, Cultured , Humans , Keratinocytes/pathology , Polymerase Chain Reaction , Psoriasis/metabolism , Psoriasis/pathology , Signal Transduction , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/biosynthesis
2.
Genomics ; 108(5-6): 201-208, 2016 12.
Article in English | MEDLINE | ID: mdl-27829169

ABSTRACT

SLC44A2 was discovered as the target of an antibody that causes hearing loss. Knockout mice develop age related hearing loss, loss of sensory cells and spiral ganglion neurons. SLC44A2 has polymorphic sites implicated in human disease. Transfusion related acute lung injury (TRALI) is linked to rs2288904 and genome wide association studies link rs2288904 and rs9797861 to venous thromboembolism (VTE), coronary artery disease and stroke. Here we report linkage disequilibrium of rs2288904 with rs3087969 and the association of these SLC44A2 SNPs with Meniere's disease severity. Tissue-specific isoform expression differences suggest that the N-terminal domain is linked to different functions in different cell types. Heterozygosity at rs2288904 CGA/CAA and rs3087969 GAT/GAC showed a trend for association with intractable Meniere's disease compared to less severe disease and to controls. The association of SLC44A2 SNPs with VTE suggests that thrombi affecting cochlear vessels could be a factor in Meniere's disease.


Subject(s)
Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Meniere Disease/genetics , Polymorphism, Single Nucleotide , Adult , Case-Control Studies , Cells, Cultured , Ear, Inner/metabolism , Female , Heterozygote , Humans , Linkage Disequilibrium , Male , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Meniere Disease/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism
3.
J Invest Dermatol ; 136(2): 444-452, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26802239

ABSTRACT

The epidermal growth factor receptor (EGFR) and its ligands are essential regulators of epithelial biology, which are often amplified in cancer cells. We have previously shown that shRNA-mediated silencing of one of these ligands, amphiregulin (AREG), results in keratinocyte growth arrest that cannot be rescued by soluble extracellular EGFR ligands. To further explore the functional importance of specific AREG domains, we stably transduced keratinocytes expressing tetracycline-inducible AREG-targeted shRNA with lentiviruses expressing silencing-proof, membrane-tethered AREG cytoplasmic and extracellular domains (AREG-CTD and AREG-ECD), as well as full-length AREG precursor (proAREG). Here we show that growth arrest of AREG-silenced keratinocytes occurs in G2/M and is significantly restored by proAREG and AREG-CTD but not by AREG-ECD. Moreover, the AREG-CTD was sufficient to normalize cell cycle distribution profiles and expression of mitosis-related genes. Our findings uncover an important role of the AREG-CTD in regulating cell division, which may be relevant to tumor resistance to EGFR-directed therapies.


Subject(s)
Cell Proliferation/genetics , EGF Family of Proteins/genetics , Gene Expression Regulation , Gene Silencing , Keratinocytes/cytology , Amphiregulin/genetics , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Knockout , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction/methods , Signal Transduction/genetics
4.
Exp Dermatol ; 25(3): 187-93, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26519132

ABSTRACT

To explore the role of amphiregulin in inflammatory epidermal hyperplasia, we overexpressed human AREG (hAREG) in FVB/N mice using a bovine K5 promoter. A construct containing AREG coding sequences flanked by 5' and 3' untranslated region sequences (AREG-UTR) led to a >10-fold increase in hAREG expression compared to an otherwise-identical construct containing only the coding region (AREG-CDR). AREG-UTR mice developed tousled, greasy fur as well as elongated nails and thickened, erythematous tail skin. No such phenotype was evident in AREG-CDR mice. Histologically, AREG-UTR mice presented with marked epidermal hyperplasia of tail skin (2.1-fold increase in epidermal thickness with a 9.5-fold increase in Ki-67(+) cells) accompanied by significantly increased CD4+ T-cell infiltration. Dorsal skin of AREG-UTR mice manifested lesser but still significant increases in epidermal thickness and keratinocyte hyperplasia. AREG-UTR mice also developed marked and significant sebaceous gland enlargement, with corresponding increases in Ki-67(+) cells. To determine the response of AREG-UTR animals to a pro-inflammatory skin challenge, topical imiquimod (IMQ) or vehicle cream was applied to dorsal and tail skin. IMQ increased dorsal skin thickness similarly in both AREG-UTR and wild type mice (1.7- and 2.2-fold vs vehicle, P < 0.001 each), but had no such effect on tail skin. These results confirm that keratinocyte expression of hAREG elicits inflammatory epidermal hyperplasia, and are consistent with prior reports of tail epidermal hyperplasia and increased sebaceous gland size in mice expressing human epigen.


Subject(s)
Amphiregulin/genetics , Amphiregulin/metabolism , Skin/metabolism , 3' Untranslated Regions , 5' Untranslated Regions , Animals , Cattle , Epidermis/pathology , ErbB Receptors/metabolism , Homeostasis , Humans , Hyperplasia/metabolism , Inflammation , Ligands , Mice , Mice, Transgenic , Phenotype , Promoter Regions, Genetic , Sebaceous Glands/metabolism
5.
Nat Commun ; 6: 7001, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25939698

ABSTRACT

Psoriasis is a chronic autoimmune disease with complex genetic architecture. Previous genome-wide association studies (GWAS) and a recent meta-analysis using Immunochip data have uncovered 36 susceptibility loci. Here, we extend our previous meta-analysis of European ancestry by refined genotype calling and imputation and by the addition of 5,033 cases and 5,707 controls. The combined analysis, consisting of over 15,000 cases and 27,000 controls, identifies five new psoriasis susceptibility loci at genome-wide significance (P<5 × 10(-8)). The newly identified signals include two that reside in intergenic regions (1q31.1 and 5p13.1) and three residing near PLCL2 (3p24.3), NFKBIZ (3q12.3) and CAMK2G (10q22.2). We further demonstrate that NFKBIZ is a TRAF3IP2-dependent target of IL-17 signalling in human skin keratinocytes, thereby functionally linking two strong candidate genes. These results further integrate the genetics and immunology of psoriasis, suggesting new avenues for functional analysis and improved therapies.


Subject(s)
Genetic Loci , Genetic Predisposition to Disease , Psoriasis/genetics , Adaptor Proteins, Signal Transducing , Genome-Wide Association Study , Humans , I-kappa B Proteins/genetics , Interleukin-17/metabolism , Keratinocytes/metabolism , Nuclear Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
6.
J Immunol ; 192(12): 6053-61, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24829417

ABSTRACT

The IL-1 family members IL-36α (IL-1F6), IL-36ß (IL-1F8), and IL-36γ (IL-1F9) and the receptor antagonist IL-36Ra (IL-1F5) constitute a novel signaling system that is poorly understood. We now show that these cytokines have profound effects on the skin immune system. Treatment of human keratinocytes with IL-36 cytokines significantly increased the expression of CXCL1, CXCL8, CCL3, CCL5, and CCL20, potent chemotactic agents for activated leukocytes, and IL-36α injected intradermally resulted in chemokine expression, leukocyte infiltration, and acanthosis of mouse skin. Blood monocytes, myeloid dendritic cells (mDC), and monocyte-derived DC (MO-DC) expressed IL-36R and responded to IL-36. In contrast, no direct effects of IL-36 on resting or activated human CD4(+) or CD8(+) T cells, or blood neutrophils, could be demonstrated. Monocytes expressed IL-1A, IL-1B, and IL-6 mRNA and IL-1ß and IL-6 protein, and mDC upregulated surface expression of CD83, CD86, and HLA-DR and secretion of IL-1ß and IL-6 after treatment with IL-36. Furthermore, IL-36α-treated MO-DC enhanced allogeneic CD4(+) T cell proliferation, demonstrating that IL-36 can stimulate the maturation and function of DC and drive T cell proliferation. These data indicate that IL-36 cytokines actively propagate skin inflammation via the activation of keratinocytes, APC, and, indirectly, T cells.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Dermatitis/immunology , Interleukin-1/immunology , Keratinocytes/immunology , Monocytes/immunology , Skin/immunology , Allografts , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line , Cell Proliferation , Cytokines/immunology , Dendritic Cells/pathology , Dermatitis/pathology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interleukin-1/pharmacology , Keratinocytes/pathology , Mice , Monocytes/pathology , Skin/pathology , Skin Transplantation
7.
J Invest Dermatol ; 132(9): 2148-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22592159

ABSTRACT

We have shown that autocrine proliferation of human keratinocytes (KCs) is strongly dependent upon amphiregulin (AREG), whereas blockade of heparin-binding EGF-like growth factor (HB-EGF) inhibits KC migration in scratch wound assays. Here we demonstrate that expression of soluble HB-EGF (sHB-EGF) or full-length transmembrane HB-EGF (proHB-EGF), but not proAREG, results in profound increases in KC migration and invasiveness in monolayer culture. Coincident with these changes, HB-EGF significantly decreases mRNA expression of several epithelial markers including keratins 1, 5, 10, and 14 while increasing expression of markers of cellular motility including SNAI1, ZEB1, COX-2, and MMP1. Immunostaining revealed HB-EGF-induced expression of the mesenchymal protein vimentin and decreased expression of E-cadherin, as well as nuclear translocation of ß-catenin. Suggestive of a trade-off between KC motility and proliferation, overexpression of HB-EGF also reduced KC growth by >90%. We also show that HB-EGF is strongly induced in regenerating epidermis after partial-thickness wounding of human skin. Taken together, our data suggest that expression of HB-EGF in human KCs triggers a migratory and invasive phenotype with many features of epithelial-mesenchymal transition (EMT), which may be beneficial in the context of cutaneous wound healing.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Intercellular Signaling Peptides and Proteins/biosynthesis , Keratinocytes/physiology , Amphiregulin , Cadherins/biosynthesis , Cell Line , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Proliferation , EGF Family of Proteins , Glycoproteins/biosynthesis , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Keratinocytes/pathology , Keratins/biosynthesis , Protein Transport , Vimentin/biosynthesis , Wound Healing , beta Catenin/biosynthesis
8.
J Invest Dermatol ; 131(2): 329-37, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20962853

ABSTRACT

Ligands of the EGF family regulate autocrine keratinocyte proliferation, and IL-1 family cytokines orchestrate epithelial defense responses. Although members of both families are overexpressed in wound healing and psoriasis, their roles in regulating the innate immune functions of keratinocytes remain incompletely explored. Using sensitive assays, we found significant increases of heparin-binding EGF-like growth factor, transforming growth factor-α, and amphiregulin mRNA and protein in lesional psoriasis compared with uninvolved or control skin. In normal human keratinocyte (NHK) monolayers, EGFR ligands were ineffective in inducing DEFB4, S100A7, and CCL20 mRNAs and human ß-defensin (hBD)-2 peptide. Combined with IL-1α, however, EGFR ligands provoked 250 × more DEFB4 and CCL20 and a 9-fold rise in S100A7 mRNA relative to the EGFR ligand alone. This synergy was also reflected in secreted hBD-2 protein, both from NHK and reconstituted human epidermis. Keratinocyte differentiation was critical for these responses, as postconfluent NHK yielded mRNA and protein levels an order of magnitude greater than subconfluent cells. Differentiation also influenced signal transduction, with subconfluent cells using NF-κB and postconfluent cells using EGFR, MEK1/2, and p38. We propose that EGFR ligands are important modifiers of IL-1 activity, synergizing with IL-1 to stimulate epidermal production of hBD-2, S100A7, and CCL20, three of the most upregulated transcripts in psoriatic plaques.


Subject(s)
Cell Differentiation/physiology , ErbB Receptors/physiology , Immunity, Innate/physiology , Interleukin-1/physiology , Keratinocytes/immunology , Signal Transduction/physiology , Adolescent , Adult , Aged , Biopsy , Calcium/metabolism , Cell Count , Cells, Cultured , Chemokine CCL20/metabolism , Humans , Keratinocytes/cytology , Keratins/metabolism , Middle Aged , Psoriasis/immunology , Psoriasis/pathology , S100 Calcium Binding Protein A7 , S100 Proteins/metabolism , Young Adult , beta-Defensins/metabolism
9.
Nat Genet ; 42(11): 991-5, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20953188

ABSTRACT

Psoriasis is a multifactorial skin disease characterized by epidermal hyperproliferation and chronic inflammation, the most common form of which is psoriasis vulgaris (PsV). We present a genome-wide association analysis of 2,339,118 SNPs in 472 PsV cases and 1,146 controls from Germany, with follow-up of the 147 most significant SNPs in 2,746 PsV cases and 4,140 controls from three independent replication panels. We identified an association at TRAF3IP2 on 6q21 and genotyped two SNPs at this locus in two additional replication panels (the combined discovery and replication panels consisted of 6,487 cases and 8,037 controls; combined P = 2.36 × 10⁻¹° for rs13210247 and combined P = 1.24 × 10⁻¹6 for rs33980500). About 15% of psoriasis cases develop psoriatic arthritis (PsA). A stratified analysis of our datasets including only PsA cases (1,922 cases compared to 8,037 controls, P = 4.57 × 10⁻¹² for rs33980500) suggested that TRAF3IP2 represents a shared susceptibility for PsV and PsA. TRAF3IP2 encodes a protein involved in IL-17 signaling and which interacts with members of the Rel/NF-κB transcription factor family.


Subject(s)
Genome-Wide Association Study/methods , Psoriasis/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Adaptor Proteins, Signal Transducing , Arthritis, Psoriatic/etiology , Arthritis, Psoriatic/genetics , Chromosomes, Human, Pair 6 , Diseases in Twins/genetics , Genetic Predisposition to Disease , Genotype , Germany/epidemiology , HLA-C Antigens/genetics , Humans , Interleukins/genetics , Polymorphism, Single Nucleotide , Psoriasis/complications , Psoriasis/epidemiology , Siblings , White People/genetics
10.
J Invest Dermatol ; 130(7): 1849-59, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20376066

ABSTRACT

The Wnt gene family encodes a set of highly conserved secreted signaling proteins that have major roles in embryogenesis and tissue homeostasis. Yet the expression of this family of important mediators in psoriasis, a disease characterized by marked changes in keratinocyte growth and differentiation, is incompletely understood. We subjected 58 paired biopsies from lesional and uninvolved psoriatic skin and 64 biopsies from normal skin to global gene expression profiling. WNT5A transcripts were upregulated fivefold in lesional skin, accompanied by increased Wnt-5a protein levels. Notably, WNT5A mRNA was markedly induced by IL-1alpha, tumor necrosis factor-alpha, IFN-gamma, and transforming growth factor-alpha in cultured keratinocytes. Frizzled 2 (FZD2) and FZD5, which encode receptors for Wnt5A, were also increased in lesional psoriatic skin. In contrast, expression of WIF1 mRNA, encoding a secreted antagonist of the Wnt proteins, was downregulated >10-fold in lesional skin, along with decreased WNT inhibitory factor (WIF)-1 immunostaining. Interestingly, pathway analysis along with reduced AXIN2 expression and lack of nuclear translocation of beta-catenin indicated a suppression of canonical Wnt signaling in lesional skin. The results of our study suggest a shift away from canonical Wnt signaling toward noncanonical pathways driven by interactions between Wnt-5a and its cognate receptors in psoriasis, accompanied by impaired homeostatic inhibition of Wnt signaling by WIF-1 and dickkopf.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Keratinocytes/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Psoriasis/physiopathology , Repressor Proteins/metabolism , Signal Transduction/physiology , Wnt Proteins/genetics , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Biopsy , Cells, Cultured , Cytokines/metabolism , Down-Regulation/physiology , Frizzled Receptors/genetics , Gene Expression Profiling , Homeostasis/physiology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Keratinocytes/cytology , Psoriasis/metabolism , Psoriasis/pathology , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/physiology , Wnt-5a Protein
11.
J Invest Dermatol ; 130(8): 2031-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20428186

ABSTRACT

The EGFR ligand amphiregulin (AREG) has been implicated as an important autocrine growth factor in several epithelial malignancies and in psoriasis, a hyperproliferative skin disorder. To characterize the mechanisms by which AREG regulates autocrine epithelial cell growth, we transduced human keratinocytes (KCs) with lentiviral constructs expressing tetracycline (TET)-inducible small hairpin RNA (shRNA). TET-induced expression of AREG shRNA markedly reduced autocrine extracellular signal-regulated kinase phosphorylation, strongly inhibited autocrine KC growth with an efficiency similar to metalloproteinase and EGFR inhibitors, and induced several markers of KC differentiation, including keratins 1 and 10. Addition of various concentrations of exogenous EGFR ligands to KC cultures reversed the growth inhibition in response to AREG-blocking antibodies but not to shRNA-mediated AREG knockdown. Lentivirus-mediated expression of the full-length AREG transmembrane (TM) precursor, but not of the AREG extracellular domain, markedly reversed the shRNA-mediated growth inhibition and morphological changes, and strongly reduced the induction of multiple markers of KC differentiation. Taken together, our data show that autocrine human KC growth is highly dependent on the AREG TM precursor protein and strongly suggest a previously unreported function of the metalloproteinase-processed carboxy (C)-terminal domain of AREG.


Subject(s)
Autocrine Communication/physiology , Glycoproteins/chemistry , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Amphiregulin , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , EGF Family of Proteins , ErbB Receptors/metabolism , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lentivirus/genetics , Ligands , Phosphorylation/physiology , Protein Precursors/metabolism , Protein Structure, Tertiary , RNA, Small Interfering , Signal Transduction/physiology , Transduction, Genetic
12.
J Invest Dermatol ; 130(1): 295-304, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19609315

ABSTRACT

Human keratinocytes (KCs) express multiple EGF receptor (EGFR) ligands; however, their functions in specific cellular contexts remain largely undefined. To address this issue, first we measured mRNA and protein levels for multiple EGFR ligands in KCs and skin. Amphiregulin (AREG) was by far the most abundant EGFR ligand in cultured KCs, with >19 times more mRNA and >7.5 times more shed protein than any other family member. EGFR ligand expression in normal skin was low (<8 per thousand of RPLP0/36B4); however, HB-EGF and AREG mRNAs were strongly induced in human skin organ culture. KC migration in scratch wound assays was highly metalloproteinase (MP)- and EGFR dependent, and was markedly inhibited by EGFR ligand antibodies. However, lentivirus-mediated expression of soluble HB-EGF, but not soluble AREG, strongly enhanced KC migration, even in the presence of MP inhibitors. Lysophosphatidic acid (LPA)-induced ERK phosphorylation was also strongly EGFR and MP dependent and markedly inhibited by neutralization of HB-EGF. In contrast, autocrine KC proliferation and ERK phosphorylation were selectively blocked by neutralization of AREG. These data show that distinct EGFR ligands stimulate KC behavior in different cellular contexts, and in an MP-dependent fashion.


Subject(s)
Cell Movement/physiology , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Keratinocytes/metabolism , Metalloproteases/metabolism , Amphiregulin , Autocrine Communication/physiology , Cell Division/physiology , Cells, Cultured , EGF Family of Proteins , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycoproteins/genetics , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Keratinocytes/cytology , Lentivirus/genetics , Ligands , Lipopolysaccharides/pharmacology , Metalloproteases/antagonists & inhibitors , Organ Culture Techniques , Phosphorylation/drug effects , Phosphorylation/physiology , RNA, Messenger/metabolism , Signal Transduction/physiology , Skin/cytology
13.
Aging Cell ; 8(6): 738-51, 2009 12.
Article in English | MEDLINE | ID: mdl-20050020

ABSTRACT

Skin hair follicles (HF) contain bulge stem cells (SC) that regenerate HFs during hair cycles, and repair skin epithelia following injury. As natural aging is associated with decreased skin repair capacity in humans, we have investigated the impact of age on human scalp HF bulge cell number and function. Here, we isolated human bulge cells, characterized as CD200+/KRT15+/KRT19+ cells of the HF, by dissection-combined CD200 selection in young and aged human skin. Targeted transcriptional profiling indicates that KRT15, KRT19, Dkk3, Dkk4, Tcf3, S100A4, Gas1, EGFR and CTGF/CCN2 are also preferentially expressed by human bulge cells, compared to differentiated HF keratinocytes (KC). Our results demonstrate that aging does not alter expression or localization of these HF SC markers. In addition, we could not detect significant differences in HF density or bulge cell number between young and aged human scalp skin. Interestingly, hedgehog (Hh) signaling is activated in human bulge cells in vivo, and down-regulated in differentiated HF KCs, both in young and aged skin. In addition, activation of Hh signaling by lentivirus-mediated overexpression of transcription factor Gli1 induces transcription of HF SC markers KRT15, KRT19, and Gas1, in cultured KCs. Together with previously reported knock-out mouse results, these data suggest a role for Hh signaling in maintaining bulge cell phenotype in young and aged human skin.


Subject(s)
Aging , Hair Follicle/cytology , Hedgehog Proteins/metabolism , Stem Cells/metabolism , Adult , Aged , Antigens, CD/metabolism , Cell Cycle Proteins/metabolism , Cell Separation , GPI-Linked Proteins , Hair Follicle/metabolism , Humans , Keratin-15/metabolism , Keratinocytes/metabolism , Membrane Proteins/metabolism
14.
Arch Dermatol Res ; 301(3): 205-17, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18773213

ABSTRACT

S100A2 is a homodimeric protein that undergoes oxidative cross-linking and translocation from the nucleus to the cytosol in the context of oxidative stress. Suggestive of a role for S100A2 in the cutaneous response to ultraviolet light, we found altered S100A2 immunostaining in photodamaged human skin, and crosslinking of S100A2 after ultraviolet A (UVA) irradiation of normal human keratinocytes (NHK). Skin from mice, rats, and rabbits did not contain S100A2 protein, whereas skin samples from pigs, frogs and humans were strongly positive. Survival after UVA irradiation was significantly greater in NHK compared to mouse keratinocytes, suggesting a protective role for S100A2. To test this hypothesis in vivo, we expressed S100A2 in SKH2/J hairless mice under the control of a bovine keratin 5 promoter, and compared responses of TG and WT mice from 1 to 7 days after a single dose (0.5-1 MED) of solar-simulated radiation (SSR) from UVA-340 bulbs. WT and TG mice manifested a similarly robust response to SSR, characterized by epidermal hyperplasia, marked induction of p21(WAF), and a twofold increase in p53. Thymine dimers (TD) were markedly increased in the epidermis and the dermis, but while over 95% of the epidermal TD were removed by 5-6 days, elevated dermal TD persisted nearly unchanged for 7 days. Global transcriptional profiling of WT and TG mice revealed strong induction of multiple transcripts, including keratins K6 and K16, defensin beta 3, S100A8, S100A9, Sprr2i and Sprr2f. However, the only S100A2-dependent difference we observed was an induction of Cxcl13 transcripts in TG, but not WT mice (4.4-fold vs. 0.7-fold, n = 3, P = 0.022). This finding was confirmed in an independent set of mice analyzed by quantitative RT-PCR (8.8-fold vs. 1.2-fold, n = 4, P = 0.001). The finding of persistent dermal DNA damage after suberythemal doses of SSR merits further study.


Subject(s)
Chemokine CXCL13/metabolism , Chemotactic Factors/metabolism , S100 Proteins/metabolism , Skin/metabolism , Skin/radiation effects , Ultraviolet Rays , Animals , Cells, Cultured , Chemokine CXCL13/genetics , Chemotactic Factors/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Radiation , Gene Expression Regulation/radiation effects , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Mice , Mice, Hairless , Mice, Transgenic , Models, Animal , Psoriasis/metabolism , Psoriasis/pathology , RNA, Messenger/metabolism , S100 Proteins/genetics , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
15.
Am J Pathol ; 170(6): 2089-99, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17525275

ABSTRACT

In this study, we examined ErbB1 signaling in human basal and squamous cell carcinomas (BCC and SCC) of the skin in vivo. We used enzyme-linked immunosorbent assay, laser capture microdissection-coupled real-time reverse transcriptase-polymerase chain reaction, and immunohistochemistry to assess expression and activation levels of ErbB1 protein, ligands, and potential downstream effectors, in BCC and SCC tumors, stroma, and adjacent epidermis. Although total ErbB1 protein and mRNA were similar in cancerous and normal skin, we found that ErbB1 activation (phospho-Tyr(1068)) was greater in bulk SCC versus BCC or normal skin. In addition, three ErbB1 ligand transcripts (amphiregulin, heparin-binding epidermal growth factor-like growth factor, and transforming growth factor-alpha) were up-regulated in tumor cells of SCC but not BCC. Expression of these ligands was also increased in asymptomatic epidermis adjacent to both SCC and BCC, relative to normal skin. Interestingly, betacellulin transcript levels were inversely regulated compared with the other ligands. Consistently, downstream ErbB1 effectors (Erk1/2 and Akt) were activated in tumor cells of SCC but not of BCC and in adjacent epidermis of both BCC and SCC. These results demonstrate that ErbB1 signaling is hyperactive in tumor cells of SCC but not of BCC and in nearby asymptomatic epidermis of both tumor types. Our results suggest that targeting ErbB1 signaling might be of benefit in the treatment of SCC.


Subject(s)
Carcinoma, Basal Cell/metabolism , Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Signal Transduction/physiology , Skin Neoplasms/metabolism , Amphiregulin , Animals , Betacellulin , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , EGF Family of Proteins , Enzyme Activation , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Epiregulin , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Skin/cytology , Skin/metabolism , Skin/pathology , Skin Neoplasms/pathology , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
16.
Mol Pharmacol ; 67(4): 1145-57, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15615697

ABSTRACT

c-Src potentiates proliferation, survival, and invasiveness in response to epidermal growth factor (EGF) in human mammary carcinoma cells. Tyrosine (Tyr) 845 of ErbB1 is phosphorylated by Src and has been implicated in control of malignant behavior. Although several lines of evidence also suggest important interactions of ErbB and Src family kinase signaling in normal epithelial cells, little is known about the mechanism of this interaction. Studying normal human keratinocytes (NHKs), here we demonstrate strong expression of the Src family kinases Src, Yes, and Fyn; Src family kinase-dependent stimulation of Tyr 845 by EGF; and potent inhibition of NHK proliferation and migration by two Src family kinase inhibitors PP1 and PD173952. EGF-stimulated extracellular signal-regulated kinase (ERK) phosphorylation occurred at much lower concentrations of EGF than required to phosphorylate Tyr 845. Moreover, the effect of Src family kinase inhibitors on EGF-stimulated ERK phosphorylation was transient, prompting a search for other targets of Src family kinase action. By enzyme-linked immunosorbent assay analysis, we found that three different Src family kinase inhibitors [6-(2,6-dichlorophenyl)-8-methyl-2-(4-morpholin-4-ylphenylamino)-8H-pyrido[2,3-d]pyrimidin-7-one (PD173952), 4-amino-5-(4-methylphenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP1), and 2-oxo-3-(4,5,6,7-tetrahydro-1H-indol-2-ylmethylene)-2,3-dihydro-1H-indole-5-sulfonic acid dimethylamide (SU6656)] markedly inhibited elaboration of soluble amphiregulin by NHKs. The ErbB inhibitor PD158780 and the mitogen-activated protein kinase kinase inhibitor U0126 also markedly inhibited NHK proliferation, migration, and amphiregulin production. Together, these observations demonstrate that one or more Src family kinases act upstream as well as downstream of ErbB1 to promote amphiregulin-dependent autocrine stimulation of NHKs and suggest that autocrine NHK proliferation is more dependent upon ERK activation than upon Tyr 845 phosphorylation.


Subject(s)
ErbB Receptors/physiology , Glycoproteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/pharmacology , Keratinocytes/metabolism , Signal Transduction , src-Family Kinases/antagonists & inhibitors , Amphiregulin , Cells, Cultured , EGF Family of Proteins , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Phosphorylation , Pyrimidines/pharmacology , src-Family Kinases/physiology
17.
Mol Biol Cell ; 15(9): 4299-309, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15254267

ABSTRACT

ErbB signaling through extracellular signal-regulated kinase (ERK) has been implicated in regulating the expression of ErbB ligands in hyperproliferative skin disorders and wound healing. Here, we characterize the process of autocrine ERK activation in cultured normal human keratinocytes (NHKs) subjected to growth factor (GF) deprivation. Basal ERK phosphorylation was lower after 48 h than after 24 h of GF deprivation, and lowest at 30-60 min after an additional medium change. ERK phosphorylation was markedly increased by low concentrations of epidermal growth factor (EGF) (0.2-1 ng/ml) that provoked only a limited increase in ErbB1 tyrosine phosphorylation and internalization. Basal ErbB tyrosine phosphorylation and ERK phosphorylation were inhibited by two different ErbB receptor tyrosine kinase inhibitors, by the ErbB1-specific neutralizing monoclonal antibody 225 IgG, by two different metalloproteinase inhibitors, and by neutralizing antibodies against amphiregulin (AR). In contrast, these responses were unaffected by neutralizing antibodies against other ErbB1 ligands or the ErbB2 inhibitors geldanamycin and AG825. The time course of autocrine ERK phosphorylation correlated with the appearance of soluble AR, and two different metalloproteinase inhibitors blocked AR release. These results define an amphiregulin- and ErbB1-dependent mechanism by which autocrine ERK activation is maintained in NHKs, even when ErbB1 autophosphorylation and internalization are limited.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Keratinocytes/metabolism , Metalloproteases/metabolism , Amphiregulin , Antibodies, Monoclonal , Cells, Cultured , Culture Media, Serum-Free , EGF Family of Proteins , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Glycoproteins/antagonists & inhibitors , Glycoproteins/immunology , Humans , Intercellular Signaling Peptides and Proteins/immunology , MAP Kinase Signaling System , Neutralization Tests , Phosphorylation
18.
Wound Repair Regen ; 11(5): 346-53, 2003.
Article in English | MEDLINE | ID: mdl-12950638

ABSTRACT

Organ culture of skin is known to recapitulate several early events in the process of wound healing. Here we investigate the function of p38 kinase signaling as a regulator of keratinocyte behavior in human skin organ culture. We first show that skin organ culture recapitulates the transition from migration to proliferation that is known to characterize the reepithelialization process. We next show that inhibition of p38 markedly impairs the formation of keratinocyte outgrowth in human skin explant cultures, as well as the migration of keratinocytes in an in vitro wound assay. In contrast, the marked induction of mRNA encoding the ErbB ligand heparin-binding epidermal growth factor-like growth factor, known to occur after skin wounding, was not blocked by inhibition of p38. As assessed by immunoblotting, phosphorylation of p38 was limited and was not increased between 0 and 7 days of organ culture. Our results show the sensitivity of reepithelialization to inhibition by p38 and suggest that p38 acts primarily during the migration phase of this process. These data also indicate that autocrine heparin-binding epidermal growth factor expression is not regulated by p38.


Subject(s)
Keratinocytes/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/physiology , Amphiregulin , Cell Culture Techniques , Cell Division/drug effects , EGF Family of Proteins , Epidermal Growth Factor/genetics , Gene Expression/drug effects , Glycoproteins/genetics , Heparin-binding EGF-like Growth Factor , Humans , Imidazoles/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Keratinocytes/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Organ Culture Techniques , Phosphorylation/drug effects , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases
19.
Biochem Biophys Res Commun ; 295(5): 1108-17, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12135609

ABSTRACT

ErbB1 and ErbB2 display differential subcellular localization in human skin and cultured keratinocytes. To determine whether ErbB1 and ErbB2 also differ in cytoskeletal binding properties, normal human keratinocytes grown under conditions favoring a basal or differentiated phenotype were repeatedly extracted in a non-ionic detergent buffer. In basaloid keratinocytes, cytoskeletal association of ErbB1 and ErbB2 was limited. ErbB1 ( approximately 5%) was tightly associated with the cytoskeleton, compared to <1% of ErbB2 (p=0.004). After EGF stimulation, activated ErbB1 and ERK associated with the cytoskeleton to a greater extent than did total ErbB1 and total ERK. Association of ErbB2 increased markedly in differentiated keratinocytes, whereas association of ErbB1 was similar in basaloid and differentiated cells. Cytoskeletal association of ErbB2 correlated with plasma membrane localization. These results suggest that ErbB1 and ErbB2 employ different mechanisms of plasma membrane targeting during keratinocyte differentiation, and that cytoskeletal association may facilitate the coupling of activated ErbB1 and ERK.


Subject(s)
Cell Differentiation/physiology , Cytoskeleton/metabolism , ErbB Receptors/metabolism , Keratinocytes/metabolism , Receptor, ErbB-2/metabolism , Calibration , Cells, Cultured , Cytosol/metabolism , Epidermal Growth Factor/metabolism , Humans , Keratinocytes/cytology , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Subcellular Fractions , Tyrosine/metabolism
20.
J Biol Chem ; 277(30): 26839-45, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12016209

ABSTRACT

To investigate the role of ERK signaling in human skin responses to wounding, organ cultures of human skin were maintained for 0.5-24 h in the presence of various inhibitors, followed by measurement of ERK phosphorylation or mRNA levels. The MEK inhibitor PD98059 produced near-complete (97-98%) inhibition of ERK phosphorylation, whereas inhibition of c-Fos, c-Jun, HB-EGF, AR, and VEGF mRNA by this compound was incomplete (41-65%). PD98059 was significantly more effective than either PD158780 or BB2516 as an inhibitor of ERK phosphorylation and of the rapid rise in c-Fos and c-Jun mRNA expression. In contrast, all three compounds inhibited the more delayed rise in HB-EGF mRNA to the same extent. Exogenous epidermal growth factor abrogated the inhibition of ERK phosphorylation caused by BB2516. These data indicate that one or more metalloproteinases activate ErbB signaling in skin organ culture, that ErbB signaling plays an important but not exclusive role in the activation of ERK, and that non-ERK pathways contribute to gene expression in this system. Because metalloproteinase-mediated cleavage of the HB-EGF transmembrane precursor is known to be ERK-dependent, our data suggest that ERK activation resulting from initial trauma leads to metalloproteinase-mediated cleavage of HB-EGF, thereby triggering the ErbB signaling cascade.


Subject(s)
Metalloendopeptidases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Blotting, Western , Cell Division , Cell Membrane/metabolism , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , MAP Kinase Signaling System , Models, Biological , Phosphorylation , Precipitin Tests , Pyrimidines/pharmacology , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...