Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Allergy Clin Immunol ; 149(2): 640-649.e5, 2022 02.
Article in English | MEDLINE | ID: mdl-34343561

ABSTRACT

BACKGROUND: A major issue with the current management of psoriasis is our inability to predict treatment response. OBJECTIVE: Our aim was to evaluate the ability to use baseline molecular expression profiling to assess treatment outcome for patients with psoriasis. METHODS: We conducted a longitudinal study of 46 patients with chronic plaque psoriasis treated with anti-TNF agent etanercept, and molecular profiles were assessed in more than 200 RNA-seq samples. RESULTS: We demonstrated correlation between clinical response and molecular changes during the course of the treatment, particularly for genes responding to IL-17A/TNF in keratinocytes. Intriguingly, baseline gene expressions in nonlesional, but not lesional, skin were the best marker of treatment response at week 12. We identified USP18, a known regulator of IFN responses, as positively correlated with Psoriasis Area and Severity Index (PASI) improvement (P = 9.8 × 10-4) and demonstrate its role in regulating IFN/TNF responses in keratinocytes. Consistently, cytokine gene signatures enriched in baseline nonlesional skin expression profiles had strong correlations with PASI improvement. Using this information, we developed a statistical model for predicting PASI75 (ie, 75% of PASI improvement) at week 12, achieving area under the receiver-operating characteristic curve value of 0.75 and up to 80% accurate PASI75 prediction among the top predicted responders. CONCLUSIONS: Our results illustrate feasibility of assessing drug response in psoriasis using nonlesional skin and implicate involvement of IFN regulators in anti-TNF responses.


Subject(s)
Cytokines/biosynthesis , Psoriasis/drug therapy , Skin/immunology , Tumor Necrosis Factor Inhibitors/therapeutic use , Cytokines/genetics , Humans , Longitudinal Studies , Psoriasis/immunology , RNA-Seq , Severity of Illness Index , Transcriptome
2.
Exp Dermatol ; 31(7): 1036-1047, 2022 07.
Article in English | MEDLINE | ID: mdl-35213752

ABSTRACT

Psoriasis vulgaris is an inflammatory skin disease that affects 2%-3% of the population worldwide. One of the major challenges in discovering novel therapies is the poor translatability of animal models to human disease. Therefore, it is imperative to develop human preclinical models of psoriasis that are amenable to pharmacological intervention. Here, we report a 3-D reconstituted human epidermis (RHE) culture system treated with cytokines commonly associated with psoriasis (TNFα, IL-17A and IL-22) that reproduced some key features of the human disease. The effects on epidermal morphology, gene transcription and cytokine production, which are dysregulated in psoriasis were assessed. Certain morphological features of psoriatic epidermis were evident in cytokine-stimulated RHEs, including hypogranulosis and parakeratosis. In addition, RHEs responded to a cytokine mix in a dose-dependent manner by expressing genes and proteins associated with impaired keratinocyte differentiation (keratin 10/K10, loricrin), innate immune responses (S100A7, DEFB4, elafin) and inflammation (IL-1α, IL-6, IL-8, IL-10, IL-12/23p40, IL-36γ, GM-CSF and IFNγ) typical of psoriasis. These disease-relevant changes in morphology, gene transcription and cytokine production were robustly attenuated by pharmacologically blocking TNFα/IL-17A-induced NF-κB activation with IKK-2 inhibitor IV. Conversely, inhibition of IL-22-induced JAK1 signalling with ABT-317 strongly attenuated morphological features of the disease but had no effect on NFκB-dependent cytokine production, suggesting distinct mechanisms of action by the cytokines driving psoriasis. These data support the use of cytokine-induced RHE models for identifying and targeting keratinocyte signalling pathways important for disease progression and may provide translational insights into novel keratinocyte mechanisms for novel psoriasis therapies.


Subject(s)
Interleukin-17 , Psoriasis , Animals , Humans , Interleukin-17/metabolism , Keratinocytes/metabolism , NF-kappa B/metabolism , Psoriasis/metabolism , Skin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
3.
FASEB J ; 34(1): 1652-1664, 2020 01.
Article in English | MEDLINE | ID: mdl-31914670

ABSTRACT

Conditions such as asthma and inflammatory bowel disease are characterized by aberrant smooth muscle contraction. It has proven difficult to develop human cell-based models that mimic acute muscle contraction in 2D in vitro cultures due to the nonphysiological chemical and mechanical properties of lab plastics that do not allow for muscle cell contraction. To enhance the relevance of in vitro models for human disease, we describe how functional 3D smooth muscle tissue that exhibits physiological and pharmacologically relevant acute contraction and relaxation responses can be reproducibly fabricated using a unique microfluidic 3D bioprinting technology. Primary human airway and intestinal smooth muscle cells were printed into rings of muscle tissue at high density and viability. Printed tissues contracted to physiological concentrations of histamine (0.01-100 µM) and relaxed to salbutamol, a pharmacological compound used to relieve asthmatic exacerbations. The addition of TGFß to airway muscle rings induced an increase in unstimulated muscle shortening and a decreased response to salbutamol, a phenomenon which also occurs in chronic lung diseases. Results indicate that the 3D bioprinted smooth muscle is a physiologically relevant in vitro model that can be utilized to study disease pathways and the effects of novel therapeutics on acute contraction and chronic tissue stenosis.


Subject(s)
Bioprinting/methods , Microfluidics/methods , Muscle, Smooth/cytology , Myocytes, Smooth Muscle/cytology , Respiratory System/cytology , Albuterol/pharmacology , Asthma/drug therapy , Asthma/pathology , Cells, Cultured , Humans , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Printing, Three-Dimensional , Respiratory System/drug effects , Tissue Engineering/methods
4.
Biol Reprod ; 103(3): 487-496, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32614039

ABSTRACT

To investigate genomic pathways that may influence physiology and infectivity during the menstrual cycle, RNA sequence analysis was performed on patient-matched engineered ectocervical tissue after follicular and luteal phase (LP) hormone treatments. We developed distinct cellular, molecular, and biological profiles in ectocervical epithelium dependent on the menstrual cycle phase. Follicular phase hormones were associated with proliferation, transcription, and cell adhesion, while LP samples expressed genes involved in immune cell recruitment, inflammation, and protein modifications. Additionally, our analysis revealed mucins not previously reported in ectocervical tissue, which could play an important role in fertility and disease prevention. This study provides insight into the phenomenon of increased LP vulnerability to infection and identifies potential targets for future research.


Subject(s)
Cervix Uteri/metabolism , Follicular Phase/physiology , Gene Expression Regulation/genetics , Luteal Phase/physiology , Menstrual Cycle/physiology , Tissue Engineering , Adult , Cell Adhesion , Cell Proliferation , Cervix Uteri/cytology , Cluster Analysis , Epithelium/metabolism , Female , High-Throughput Nucleotide Sequencing , Hormones/pharmacology , Humans , Models, Anatomic , Mucins/physiology
5.
Biol Reprod ; 103(3): 497-507, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32401296

ABSTRACT

There is a shortage of research models that adequately represent the unique mucosal environment of human ectocervix, limiting development of new therapies for treating infertility, infection, or cancer. We developed three microphysiologic human ectocervix models to study hormone action during homeostasis. First, we reconstructed ectocervix using decellularized extracellular matrix scaffolds, which supported cell integration and could be clinically useful. Secondly, we generated organotypic systems consisting of ectocervical explants co-cultured with murine ovaries or cycling exogenous hormones, which mimicked human menstrual cycles. Finally, we engineered ectocervix tissue consisting of tissue-specific stromal-equivalents and fully-differentiated epithelium that mimicked in vivo physiology, including squamous maturation, hormone response, and mucin production, and remained viable for 28 days in vitro. The localization of differentiation-dependent mucins in native and engineered tissue was identified for the first time, which will allow increased efficiency in mucin targeting for drug delivery. In summary, we developed and characterized three microphysiologic human ectocervical tissue models that will be useful for a variety of research applications, including preventative and therapeutic treatments, drug and toxicology studies, and fundamental research on hormone action in a historically understudied tissue that is critical for women's health.


Subject(s)
Cervix Uteri/physiology , Endocrine System/physiology , Models, Biological , Paracrine Communication/physiology , Animals , Drug Delivery Systems , Extracellular Matrix , Female , Hormones/physiology , Humans , Menstruation/physiology , Mice , Mucins/biosynthesis , Mucous Membrane/physiology , Pregnancy , RNA/biosynthesis , RNA/genetics , Tissue Engineering
6.
J Cell Sci ; 130(1): 111-118, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27815408

ABSTRACT

EphA2 is a receptor tyrosine kinase that helps to maintain epidermal tissue homeostasis. A proximity-dependent biotin identification (BioID) approach was used to identify proteins in close proximity to EphA2 within primary human keratinocytes and three-dimensional (3D) reconstituted human epidermis (RHE) cultures to map a putative protein interaction network for this membrane receptor that exhibits a polarized distribution in stratified epithelia. Although a subset of known EphA2 interactors were identified in the BioID screen, >97% were uniquely detected in keratinocytes with over 50% of these vicinal proteins only present in 3D human epidermal culture. Afadin (AFDN), a cytoskeletal and junction-associated protein, was present in 2D and 3D keratinocyte cultures, and validated as a so-far-unknown EphA2-interacting protein. Loss of EphA2 protein disrupted the subcellular distribution of afadin and occludin in differentiated keratinocytes, leading to impairment of tight junctions. Collectively, these studies illustrate the use of the BioID approach in order to map receptor interaction networks in 3D human epithelial cultures, and reveal a positive regulatory role for EphA2 in the organization of afadin and epidermal tight junctions.


Subject(s)
Epidermis/metabolism , Keratinocytes/metabolism , Microfilament Proteins/metabolism , Proteomics , Receptor, EphA2/metabolism , Tight Junctions/metabolism , Carbon-Nitrogen Ligases/metabolism , Cells, Cultured , Escherichia coli Proteins/metabolism , Humans , Infant, Newborn , Male , Protein Binding , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Reproducibility of Results
7.
Ann Rheum Dis ; 77(11): 1653-1664, 2018 11.
Article in English | MEDLINE | ID: mdl-30021804

ABSTRACT

OBJECTIVE: Skin inflammation and photosensitivity are common in patients with cutaneous lupus erythematosus (CLE) and systemic lupus erythematosus (SLE), yet little is known about the mechanisms that regulate these traits. Here we investigate the role of interferon kappa (IFN-κ) in regulation of type I interferon (IFN) and photosensitive responses and examine its dysregulation in lupus skin. METHODS: mRNA expression of type I IFN genes was analysed from microarray data of CLE lesions and healthy control skin. Similar expression in cultured primary keratinocytes, fibroblasts and endothelial cells was analysed via RNA-seq. IFNK knock-out (KO) keratinocytes were generated using CRISPR/Cas9. Keratinocytes stably overexpressing IFN-κ were created via G418 selection of transfected cells. IFN responses were assessed via phosphorylation of STAT1 and STAT2 and qRT-PCR for IFN-regulated genes. Ultraviolet B-mediated apoptosis was analysed via TUNEL staining. In vivo protein expression was assessed via immunofluorescent staining of normal and CLE lesional skin. RESULTS: IFNK is one of two type I IFNs significantly increased (1.5-fold change, false discovery rate (FDR) q<0.001) in lesional CLE skin. Gene ontology (GO) analysis showed that type I IFN responses were enriched (FDR=6.8×10-04) in keratinocytes not in fibroblast and endothelial cells, and this epithelial-derived IFN-κ is responsible for maintaining baseline type I IFN responses in healthy skin. Increased levels of IFN-κ, such as seen in SLE, amplify and accelerate responsiveness of epithelia to IFN-α and increase keratinocyte sensitivity to UV irradiation. Notably, KO of IFN-κ or inhibition of IFN signalling with baricitinib abrogates UVB-induced apoptosis. CONCLUSION: Collectively, our data identify IFN-κ as a critical IFN in CLE pathology via promotion of enhanced IFN responses and photosensitivity. IFN-κ is a potential novel target for UVB prophylaxis and CLE-directed therapy.


Subject(s)
Epidermis/immunology , Interferon Type I/biosynthesis , Lupus Erythematosus, Cutaneous/complications , Photosensitivity Disorders/etiology , Adult , Cells, Cultured , Dendritic Cells/immunology , Female , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Keratinocytes/immunology , Lupus Erythematosus, Cutaneous/immunology , Male , Middle Aged , Photosensitivity Disorders/immunology , RNA, Messenger/genetics , Skin/immunology , TYK2 Kinase/immunology , Up-Regulation/immunology
8.
Exp Cell Res ; 358(1): 58-64, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28322822

ABSTRACT

During development, cells of seemingly homogenous character sort themselves out into distinct compartments in order to generate cell types with specialized features that support tissue morphogenesis and function. This process is often driven by receptors at the cell membrane that probe the extracellular microenvironment for specific ligands and alter downstream signaling pathways impacting transcription, cytoskeletal organization, and cell adhesion to regulate cell sorting and subsequent boundary formation. This review will focus on two of these receptor families, Eph and Notch, both of which are intrinsically non-adhesive and are activated by a unique set of ligands that are asymmetrically distributed from their receptor on neighboring cells. Understanding the requirement of asymmetric ligand-receptor signaling at the membrane under homeostatic conditions gives insight into how misregulation of these pathways contributes to boundary disruption in diseases like cancer.


Subject(s)
Cell Adhesion/physiology , Cell Differentiation/physiology , Ephrins/metabolism , Morphogenesis/physiology , Receptors, Eph Family/metabolism , Signal Transduction/physiology , Animals , Humans
9.
Biol Reprod ; 96(5): 971-981, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28449068

ABSTRACT

Three-dimensional (3D) in vitro models have been established to study the physiology and pathophysiology of the endometrium. With emerging evidence that the native extracellular matrix (ECM) provides appropriate cues and growth factors essential for tissue homeostasis, we describe, a novel 3D endometrium in vitro model developed from decellularized human endometrial tissue repopulated with primary endometrial cells. Analysis of the decellularized endometrium using mass spectrometry revealed an enrichment of cell adhesion molecules, cytoskeletal proteins, and ECM proteins such as collagen IV and laminin. Primary endometrial cells within the recellularized scaffolds proliferated and remained viable for an extended period of time in vitro. In order to evaluate the hormonal response of cells within the scaffolds, the recellularized scaffolds were treated with a modified 28-day hormone regimen to mimic the human menstrual cycle. At the end of 28 days, the cells within the endometrial scaffold expressed both estrogen and progesterone receptors. In addition, decidualization markers, IGFBP-1 and prolactin, were secreted upon addition of dibutyryl cyclic AMP indicative of a decidualization response. This 3D model of the endometrium provides a new experimental tool to study endometrial biology and drug testing.


Subject(s)
Endometrium/drug effects , Hormones/pharmacology , Adolescent , Adult , Cell Adhesion Molecules/metabolism , Collagen Type IV/metabolism , Cytoskeletal Proteins/metabolism , Endometrium/cytology , Extracellular Matrix/metabolism , Female , Humans , In Vitro Techniques , Laminin/metabolism , Menstrual Cycle/physiology , Primary Cell Culture , Proteomics , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Young Adult
10.
Hum Mol Genet ; 23(15): 4064-76, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24643277

ABSTRACT

iRHOM2 is a highly conserved, catalytically inactive member of the Rhomboid family, which has recently been shown to regulate the maturation of the multi-substrate ectodomain sheddase enzyme ADAM17 (TACE) in macrophages. Dominant iRHOM2 mutations are the cause of the inherited cutaneous and oesophageal cancer-susceptibility syndrome tylosis with oesophageal cancer (TOC), suggesting a role for this protein in epithelial cells. Here, using tissues derived from TOC patients, we demonstrate that TOC-associated mutations in iRHOM2 cause an increase in the maturation and activity of ADAM17 in epidermal keratinocytes, resulting in significantly upregulated shedding of ADAM17 substrates, including EGF-family growth factors and pro-inflammatory cytokines. This activity is accompanied by increased EGFR activity, increased desmosome processing and the presence of immature epidermal desmosomes, upregulated epidermal transglutaminase activity and heightened resistance to Staphylococcal infection in TOC keratinocytes. Many of these features are consistent with the presence of a constitutive wound-healing-like phenotype in TOC epidermis, which may shed light on a novel pathway in skin repair, regeneration and inflammation.


Subject(s)
ADAM Proteins/genetics , Carrier Proteins/genetics , Epidermis/metabolism , Esophageal Neoplasms/genetics , Keratinocytes/metabolism , Keratoderma, Palmoplantar/genetics , Staphylococcal Skin Infections/genetics , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , ADAM17 Protein , Carrier Proteins/metabolism , Cytokines/biosynthesis , Desmosomes/metabolism , Desmosomes/pathology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Epidermis/microbiology , Epidermis/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/microbiology , Esophageal Neoplasms/pathology , Female , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins , Keratinocytes/microbiology , Keratinocytes/pathology , Keratoderma, Palmoplantar/metabolism , Keratoderma, Palmoplantar/microbiology , Keratoderma, Palmoplantar/pathology , Male , Mutation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/physiology , Transglutaminases/genetics , Transglutaminases/metabolism
11.
Stem Cells ; 33(5): 1642-56, 2015 May.
Article in English | MEDLINE | ID: mdl-25639731

ABSTRACT

The stem cell niche is thought to affect cell cycle quiescence, proliferative capacity, and communication between stem cells and their neighbors. How these activities are controlled is not completely understood. Here we define a microRNA family (miRs-103/107) preferentially expressed in the stem cell-enriched limbal epithelium that regulates and integrates these stem cell characteristics. miRs-103/107 target the ribosomal kinase p90RSK2, thereby arresting cells in G0/G1 and contributing to a slow-cycling phenotype. Furthermore, miRs-103/107 increase the proliferative capacity of keratinocytes by targeting Wnt3a, which enhances Sox9 and YAP1 levels and thus promotes a stem cell phenotype. This miRNA family also regulates keratinocyte cell-cell communication by targeting: (a) the scaffolding protein NEDD9, preserving E-cadherin-mediated cell adhesion; and (b) the tyrosine phosphatase PTPRM, which negatively regulates connexin 43-based gap junctions. We propose that such regulation of cell communication and adhesion molecules maintains the integrity of the stem cell niche ultimately preserving self-renewal, a hallmark of epithelial stem cells.


Subject(s)
Epithelial Cells/metabolism , MicroRNAs/metabolism , Stem Cells/metabolism , Animals , Cadherins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Gap Junctions/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Limbus Corneae/cytology , Male , Mice, Inbred BALB C , MicroRNAs/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Stem Cells/drug effects , Wnt3A Protein/pharmacology
12.
Hum Mol Genet ; 22(3): 531-43, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23108156

ABSTRACT

Ankyloblepharon, ectodermal defects, cleft lip/palate (AEC) syndrome is a rare autosomal dominant disorder caused by mutations in the p63 gene, essential for embryonic development of stratified epithelia. The most severe cutaneous manifestation of this disorder is the long-lasting skin fragility associated with severe skin erosions after birth. Using a knock-in mouse model for AEC syndrome, we found that skin fragility was associated with microscopic blistering between the basal and suprabasal compartments of the epidermis and reduced desmosomal contacts. Expression of desmosomal cadherins and desmoplakin was strongly reduced in AEC mutant keratinocytes and in newborn epidermis. A similar impairment in desmosome gene expression was observed in human keratinocytes isolated from AEC patients, in p63-depleted keratinocytes and in p63 null embryonic skin, indicating that p63 mutations causative of AEC syndrome have a dominant-negative effect on the wild-type p63 protein. Among the desmosomal components, desmocollin 3, desmoplakin and desmoglein 1 were the most significantly reduced by mutant p63 both at the RNA and protein levels. Chromatin immunoprecipitation experiments and transactivation assays revealed that p63 controls these genes at the transcriptional level. Consistent with reduced desmosome function, AEC mutant and p63-deficient keratinocytes had an impaired ability to withstand mechanical stress, which was alleviated by epidermal growth factor receptor inhibitors known to stabilize desmosomes. Our study reveals that p63 is a crucial regulator of a subset of desmosomal genes and that this function is impaired in AEC syndrome. Reduced mechanical strength resulting from p63 mutations can be alleviated pharmacologically by increasing desmosome adhesion with possible therapeutic implications.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Desmosomes/metabolism , Ectodermal Dysplasia/genetics , Eye Abnormalities/genetics , Membrane Proteins/metabolism , Animals , Cell Adhesion , Cells, Cultured , Chromatin Immunoprecipitation , Cleft Lip/pathology , Cleft Palate/pathology , Cloning, Molecular , Desmosomes/genetics , Ectodermal Dysplasia/pathology , Epidermis/metabolism , Epidermis/physiopathology , Epithelium/metabolism , Epithelium/physiopathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Eye Abnormalities/pathology , Eyelids/abnormalities , Eyelids/pathology , Female , Gene Expression Regulation , Humans , Keratinocytes/metabolism , Luciferases/analysis , Luciferases/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Real-Time Polymerase Chain Reaction , Skin/metabolism , Skin/physiopathology
13.
Am J Pathol ; 184(12): 3262-71, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25455687

ABSTRACT

Factor inhibiting hypoxia-inducible factor 1 (FIH-1; official symbol HIF1AN) is a hydroxylase that negatively regulates hypoxia-inducible factor 1α but also targets other ankyrin repeat domain-containing proteins such as Notch receptor to limit epithelial differentiation. We show that FIH-1 null mutant mice exhibit delayed wound healing. Importantly, in vitro scratch wound assays demonstrate that the positive role of FIH-1 in migration is independent of Notch signaling, suggesting that this hydroxylase targets another ankyrin repeat domain-containing protein to positively regulate motogenic signaling pathways. Accordingly, FIH-1 increases epidermal growth factor receptor (EGFR) signaling, which in turn enhances keratinocyte migration via mitogen-activated protein kinase pathway, leading to extracellular signal-regulated kinase 1/2 activation. Our studies identify leucine-rich repeat kinase 1 (LRRK1), a key regulator of the EGFR endosomal trafficking and signaling, as an FIH-1 binding partner. Such an interaction prevents the formation of an EGFR/LRRK1 complex, necessary for proper EGFR turnover. The identification of LRRK1 as a novel target for FIH-1 provides new insight into how FIH-1 functions as a positive regulator of epithelial migration.


Subject(s)
ErbB Receptors/metabolism , Keratinocytes/cytology , Mixed Function Oxygenases/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Movement , Cells, Cultured , Epithelium/embryology , Humans , Immunohistochemistry , Keratinocytes/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Mixed Function Oxygenases/genetics , Protein Binding , Signal Transduction , Wound Healing
14.
Cell Tissue Res ; 360(3): 501-12, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25693896

ABSTRACT

Desmosomes are cell-cell adhesive organelles with a well-known role in forming strong intercellular adhesion during embryogenesis and in adult tissues subject to mechanical stress, such as the heart and skin. More recently, desmosome components have also emerged as cell signaling regulators. Loss of expression or interference with the function of desmosome molecules results in diseases of the heart and skin and contributes to cancer progression. However, the underlying molecular mechanisms that result in inherited and acquired disorders remain poorly understood. To address this question, researchers are directing their studies towards determining the functions that occur inside and outside of the junctions and the extent to which functions are adhesion-dependent or independent. This review focuses on recent discoveries that provide insights into the role of desmosomes and desmosome components in cell signaling and disease; wherever possible, we address molecular functions within and outside of the adhesive structure.


Subject(s)
Desmosomes/metabolism , Disease , Signal Transduction , Animals , Humans , Proteins/metabolism
15.
Proc Natl Acad Sci U S A ; 109(35): 14030-4, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22891326

ABSTRACT

Notch plays a critical role in the transition from proliferation to differentiation in the epidermis and corneal epithelium. Furthermore, aberrant Notch signaling is a feature of diseases like psoriasis, eczema, nonmelanoma skin cancer, and melanoma where differentiation and proliferation are impaired. Whereas much is known about the downstream events following Notch signaling, factors responsible for negatively regulating Notch receptor signaling after ligand activation are incompletely understood. Notch can undergo hydroxylation by factor-inhibiting hypoxia-inducible factor 1 (FIH-1); however, the biological significance of this phenomenon is unclear. Here we show that FIH-1 expression is up-regulated in diseased epidermis and corneal epithelium. Elevating FIH-1 levels in primary human epidermal keratinocytes (HEKs) and human corneal epithelial keratinocytes (HCEKs) impairs differentiation in submerged cultures and in a "three-dimensional" organotypic raft model of human epidermis, in part, via a coordinate decrease in Notch signaling. Knockdown of FIH-1 enhances keratinocyte differentiation. Loss of FIH-1 in vivo increased Notch activity in the limbal epithelium, resulting in a more differentiated phenotype. microRNA-31 (miR-31) is an endogenous negative regulator of FIH-1 expression that results in keratinocyte differentiation, mediated by Notch activation. Ectopically expressing miR-31 in an undifferentiated corneal epithelial cell line promotes differentiation and recapitulates a corneal epithelium in a three-dimensional raft culture model. Our results define a previously unknown mechanism for keratinocyte fate decisions where Notch signaling potential is, in part, controlled through a miR-31/FIH-1 nexus.


Subject(s)
Keratinocytes/cytology , Keratinocytes/physiology , MicroRNAs/metabolism , Mixed Function Oxygenases/metabolism , Psoriasis/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Transformed , Epidermal Cells , Epidermis/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Epithelium, Corneal/cytology , Epithelium, Corneal/physiology , Female , Humans , Mice , Mice, Transgenic , Mixed Function Oxygenases/genetics , Organ Culture Techniques , Phenotype , Psoriasis/genetics , Psoriasis/pathology , Receptors, Notch/metabolism , Repressor Proteins/genetics , Signal Transduction/physiology
16.
Semin Cell Dev Biol ; 23(1): 92-101, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22040910

ABSTRACT

Eph receptor tyrosine kinases mediate cell-cell communication by interacting with ephrin ligands residing on adjacent cell surfaces. In doing so, these juxtamembrane signaling complexes provide important contextual information about the cellular microenvironment that helps orchestrate tissue morphogenesis and maintain homeostasis. Eph/ephrin signaling has been implicated in various aspects of mammalian skin physiology, with several members of this large family of receptor tyrosine kinases and their ligands present in the epidermis, hair follicles, sebaceous glands, and underlying dermis. This review focuses on the emerging role of Eph receptors and ephrins in epidermal keratinocytes where they can modulate proliferation, migration, differentiation, and death. The activation of Eph receptors by ephrins at sites of cell-cell contact also appears to play a key role in the maturation of intercellular junctional complexes as keratinocytes move out of the basal layer and differentiate in the suprabasal layers of this stratified, squamous epithelium. Furthermore, alterations in the epidermal Eph/ephrin axis have been associated with cutaneous malignancy, wound healing defects and inflammatory skin conditions. These collective observations suggest that the Eph/ephrin cell-cell communication pathway may be amenable to therapeutic intervention for the purpose of restoring epidermal tissue homeostasis and integrity in dermatological disorders.


Subject(s)
Cell Differentiation , Ephrins/physiology , Epidermis/metabolism , Receptors, Eph Family/physiology , Signal Transduction , Animals , Cell Adhesion , Cell Proliferation , Ephrins/metabolism , Epidermis/pathology , Humans , Keratinocytes/metabolism , Keratinocytes/physiology , Receptors, Eph Family/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Wound Healing
18.
J Invest Dermatol ; 144(8): 1798-1807.e1, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38520417

ABSTRACT

Intricate signaling systems are required to maintain homeostasis and promote differentiation in the epidermis. Receptor tyrosine kinases are central in orchestrating these systems in epidermal keratinocytes. In particular, EPHA2 and EGFR transduce distinct signals to dictate keratinocyte fate, yet how these cell communication networks are integrated has not been investigated. Our work shows that loss of EPHA2 impairs keratinocyte stratification, differentiation, and barrier function. To determine the mechanism of this dysfunction, we drew from our proteomics data of potential EPHA2 interacting proteins. We identified EGFR as a high-ranking EPHA2 interactor and subsequently validated this interaction. We found that when EPHA2 is reduced, EGFR activation and downstream signaling are intensified and sustained. Evidence indicates that prolonged SRC association contributes to the increase in EGFR signaling. We show that hyperactive EGFR signaling underlies the differentiation defect caused by EPHA2 knockdown because EGFR inhibition restores differentiation in EPHA2-deficient 3-dimensional skin organoids. Our data implicate a mechanism whereby EPHA2 restrains EGFR signaling, allowing for fine tuning in the processes of terminal differentiation and barrier formation. Taken together, we purport that crosstalk between receptor tyrosine kinases EPHA2 and EGFR is critical for epidermal differentiation.


Subject(s)
Cell Differentiation , Epidermis , ErbB Receptors , Keratinocytes , Receptor, EphA2 , Signal Transduction , Receptor, EphA2/metabolism , Receptor, EphA2/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Humans , Keratinocytes/metabolism , Keratinocytes/cytology , Epidermis/metabolism , Cells, Cultured , Mice , Animals , Epidermal Cells/metabolism , Epidermal Cells/cytology , Proteomics/methods
19.
Am J Pathol ; 177(6): 2921-37, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21075858

ABSTRACT

Desmoglein 1 (Dsg1) is a desmosomal cadherin that is essential to epidermal integrity. In the blistering diseases bullous impetigo and staphylococcal scalded-skin syndrome, pathogenesis depends on cleavage of Dsg1 by a bacterial protease, exfoliative toxin A, which removes residues 1 to 381 of the Dsg1 ectodomain. However, the cellular responses to Dsg1 cleavage that precipitate keratinocyte separation to induce blister formation are unknown. Here, we show that ectodomain-deleted Dsg1 (Δ381-Dsg1) mimics the toxin-cleaved cadherin, disrupts desmosomes, and reduces the mechanical integrity of keratinocyte sheets. In addition, we demonstrate that truncated Dsg1 remains associated with its catenin partner, plakoglobin, and causes a reduction in the levels of endogenous desmosomal cadherins in a dose-dependent manner, leading us to hypothesize that plakoglobin sequestration by truncated Dsg1 destabilizes other cadherins. Accordingly, a triple-point mutant of the ectodomain-deleted cadherin, which is uncoupled from plakoglobin, does not impair adhesion, indicating that this interaction is essential to the pathogenic potential of truncated Dsg1. Moreover, we demonstrate that increasing plakoglobin levels rescues cadherin expression, desmosome organization, and functional adhesion in cells expressing Δ381-Dsg1 or treated with exfoliative toxin A. Finally, we report that histone deacetylase inhibition up-regulates desmosomal cadherins and prevents the loss of adhesion induced by Dsg1 truncation. These findings further our understanding of the mechanism of exfoliative toxin-induced pathology and suggest novel strategies to suppress blistering in bulbous impetigo and staphylococcal scalded-skin syndrome.


Subject(s)
Blister/etiology , Dermatitis, Exfoliative/etiology , Desmoglein 1/genetics , Exfoliatins/adverse effects , Protein Interaction Domains and Motifs/genetics , Sequence Deletion/physiology , gamma Catenin/physiology , Blister/genetics , Blister/pathology , Cell Adhesion/genetics , Cells, Cultured , Dermatitis, Exfoliative/genetics , Dermatitis, Exfoliative/pathology , Desmoglein 1/chemistry , Desmoglein 1/metabolism , Desmoglein 1/physiology , Desmosomal Cadherins/chemistry , Desmosomal Cadherins/genetics , Desmosomal Cadherins/metabolism , Desmosomal Cadherins/physiology , Exfoliatins/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/physiology , Protein Interaction Domains and Motifs/physiology , Protein Processing, Post-Translational/drug effects , Skin/metabolism , Skin/pathology , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/pathology , Transduction, Genetic , gamma Catenin/genetics , gamma Catenin/metabolism
20.
FASEB J ; 24(10): 3950-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20530248

ABSTRACT

microRNA-205 (miR-205) and miR-184 coordinately regulate the lipid phosphatase SHIP2 for Akt survival signaling in keratinocytes. As the PI3K-Akt pathway has also been implicated in regulating the actin cytoskeleton and cell motility, we investigated the role that these 2 miRNAs play in keratinocyte migration. We used antagomirs (antago) to reduce the levels of miR-205 and miR-184 in primary human epidermal keratinocytes (HEKs) and corneal epithelial keratinocytes (HCEKs) as well as direct SHIP2 silencing using siRNA oligos. Treatment of HEKs and HCEKs with antago-205 increased SHIP2 levels and impaired the ability of these cells to seal linear scratch wounds compared with untreated or irrelevant-antago treatments. In contrast, AKT signaling was enhanced and wounds sealed faster in HCEKs where miR-184 was suppressed, enabling miR-205 to inhibit SHIP2. Similar increases in migration were observed following direct SHIP2 silencing in HEKs. Furthermore, down-regulation of miR-205 resulted in an increase in Rho-ROCKI activity, phosphorylation of the actin severing protein cofilin, and a corresponding diminution of filamentous actin. The connection among miR-205, RhoA-ROCKI-cofilin inactivation, and the actin cytoskeleton represents a novel post-translational mechanism for the regulation of normal human keratinocyte migration.


Subject(s)
Cell Movement/physiology , Gene Expression Regulation/genetics , Hair/metabolism , Keratinocytes/cytology , MicroRNAs/physiology , Skin/metabolism , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Flow Cytometry , Hair/cytology , In Situ Hybridization , Mice , Skin/cytology
SELECTION OF CITATIONS
SEARCH DETAIL