Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892279

ABSTRACT

Although not completely understood, the role of the Hedgehog-GLI (HH-GLI) signaling pathway in melanoma and epithelial skin tumors has been reported before. In this study, we confirmed in various melanoma cell line models that keratin 16 (KRT16) and S100 Calcium-Binding Protein A7 (S100A7) are transcriptional targets of GLI Family Zinc Finger (GLI) proteins. Besides their important role in protecting and maintaining the epidermal barrier, keratins are somehow tightly connected with the S100 family of proteins. We found that stronger expression of KRT16 indeed corresponds to stronger expression of S100A7 in our clinical melanoma samples. We also report a trend regarding staining of GLI1, which corresponds to stronger staining of GLI3, KRT16, and S100A7 proteins. The most interesting of our findings is that all the proteins are detected specifically in the epidermis overlying the tumor, but rarely in the tumor itself. The examined proteins were also not detected in the healthy epidermis at the edges of the sample, suggesting that the staining is specific to the epidermis overlaying the tumor mass. Of all proteins, only S100A7 demonstrated a statistically significant trend regarding tumor staging and staining intensity. Results from our clinical samples prove that immune infiltration is an important feature of melanoma. Pigmentophages and tumor-infiltrating lymphocytes (TIL) demonstrate a significant association with tumor stage, while mononuclear cells are equally present in all stages. For S100A7, we found an association between the number of TILs and staining intensity. Considering these new findings presented in our study, we suggest a more detailed examination of the possible role of the S100A7 protein as a biomarker in melanoma.


Subject(s)
Epidermis , Gene Expression Regulation, Neoplastic , Keratin-16 , Melanoma , S100 Calcium Binding Protein A7 , Skin Neoplasms , Zinc Finger Protein GLI1 , Humans , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , S100 Calcium Binding Protein A7/metabolism , S100 Calcium Binding Protein A7/genetics , Epidermis/metabolism , Epidermis/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Cell Line, Tumor , Keratin-16/metabolism , Keratin-16/genetics , Up-Regulation , Male , Female , Middle Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Aged
2.
Arch Dermatol Res ; 316(6): 236, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38795158

ABSTRACT

Accumulating evidence indicates that microRNAs (miRNAs) have a vital effect on the pathogenesis of psoriasis. This study is conducted to investigate the potential involvement of miR-181a-5p and miR-181b-5p in the proliferation of HaCaT keratinocytes. Cell viability and proliferation were evaluated respectively in this study using the CCK-8 and the 5-ethynyl-2'-deoxyuridine (EdU) assays. The expression of Maternal Embryonic Leucine Zipper Kinase (MELK) and Keratin 16 (KRT16) mRNA and protein in tissues and cells was assessed using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. The Luciferase reporter system analyzes the connection between miR-181a-5p/miR-181b-5p and MELK. The results showed that miR-181a/b-5p expression was downregulated in the psoriasis lesions and negatively regulated the proliferation of keratinocytes. MELK was directly targeted by miR-181a-5p/miR-181b-5p. In addition, HaCaT keratinocytes proliferation was inhibited by knockdown of MELK while promoted dramatically by MELK overexpression. Notably, miR-181a/b-5p mimics could attenuate the effects of MELK in keratinocytes. In conclusion, our research findings suggested miR-181a-5p and miR-181b-5p negatively regulate keratinocyte proliferation by targeting MELK, providing potential diagnostic biomarkers and therapeutic targets for psoriasis.


Subject(s)
Cell Proliferation , HaCaT Cells , Keratinocytes , MicroRNAs , Protein Serine-Threonine Kinases , Psoriasis , Humans , MicroRNAs/metabolism , MicroRNAs/genetics , Keratinocytes/metabolism , Cell Proliferation/genetics , Psoriasis/pathology , Psoriasis/genetics , Psoriasis/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Keratin-16/metabolism , Keratin-16/genetics , Down-Regulation , Cell Survival , Cell Line
3.
Eur J Histochem ; 65(1)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33666385

ABSTRACT

This pilot study was aimed at comparing TLR7/TLR9 expression, cytoskeletal arrangement, and cell proliferation by indirect immunofluorescence in parallel lesional and non lesional skin samples of guttate psoriasis (PG) and psoriasis vulgaris (PV) in five male patients for each group (n=10). TLR7 expression was detected throughout all the epidermal compartment in PV samples, while in PG skin was restricted to the granular layer. TLR9 was present in the granular layer of non lesional skin and in the suprabasal layers of PV/PG lesional skin. Cell proliferation was localized in all the epidermal layers in lesional PG and PV, consistently with the immunopositivity for the "psoriatic keratin" K16. In the suprabasal layers of lesional PG and PV skin, a similar K17 expression was detected and K10 exhibited a patchy distribution. The present results suggest that TLR7 expression can be considered an intrinsic and differential histomorphological feature of PV.


Subject(s)
Cell Proliferation/physiology , Cytoskeleton/metabolism , Psoriasis/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Biomarkers/metabolism , Fluorescent Antibody Technique , Humans , Keratin-10/metabolism , Keratin-16/metabolism , Keratin-17/metabolism , Keratinocytes/metabolism , Male , Pilot Projects , Psoriasis/classification , Psoriasis/pathology , Skin/pathology
4.
Exp Dermatol ; 29(10): 993-1003, 2020 10.
Article in English | MEDLINE | ID: mdl-32737987

ABSTRACT

Skin models mimicking features of psoriasis-related inflammation are needed to support the development of new drugs in dermatology. Reconstructed skin models lack tissue complexity, including a fully competent skin barrier, and presence and/or diversity of immune cells. Here, we describe InflammaSkin®, a novel human Th17-driven ex vivo skin inflammation model. In this model, skin-resident T cells are in situ activated by intradermal injection of anti-CD3 and anti-CD28 antibodies and Th17 cell polarization is sustained by culture in a chemically defined medium supplemented with IL-1ß, IL-23 and TGF-ß for seven days. The acquired Th17 signature is demonstrated by the sustained secretion of IL-17A, IL-17AF, IL-17F, IL-22, IFN-γ, and to some degree IL-15 and TNF-α observed in the activated ex vivo skin inflammation model compared with the non-activated skin model control. Furthermore, expression of S100A7 and Keratin-16 by keratinocytes and loss of epidermal structure integrity occur subsequently to in situ Th17cell activation, demonstrating cellular crosstalk between Th17 cells and keratinocytes. Finally, we demonstrate the use of this model to investigate the modulation of the IL-23/IL-17 immune axis by topically applied anti-inflammatory compounds. Taken together, we show that by in situ activation of skin-resident Th17 cells, the InflammaSkin® model reproduces aspects of inflammatory responses observed in psoriatic lesions and could be used as a translational tool to assess efficacy of test compounds.


Subject(s)
Dermatitis/immunology , Lymphocyte Activation , Models, Biological , Th17 Cells/immunology , Anti-Inflammatory Agents/therapeutic use , Antibodies , Betamethasone/analogs & derivatives , Betamethasone/therapeutic use , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Communication , Culture Media , Dermatitis/drug therapy , Humans , Interferon-gamma/metabolism , Interleukin-15/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Keratin-16/metabolism , Keratinocytes/metabolism , Phosphodiesterase 4 Inhibitors/therapeutic use , S100 Calcium Binding Protein A7/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
5.
Mol Biol Cell ; 31(11): 1103-1111, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32213122

ABSTRACT

Mitochondria fulfill essential roles in ATP production, metabolic regulation, calcium signaling, generation of reactive oxygen species (ROS), and additional determinants of cellular health. Recent studies have highlighted a role for mitochondria during cell differentiation, including in skin epidermis. The observation of oxidative stress in keratinocytes from Krt16 null mouse skin, a model for pachyonychia congenita (PC)-associated palmoplantar keratoderma, prompted us to examine the role of Keratin (K) 16 protein and its partner K6 in regulating the structure and function of mitochondria. Electron microscopy revealed major anomalies in mitochondrial ultrastructure in late stage, E18.5, Krt6a/Krt6b null embryonic mouse skin. Follow-up studies utilizing biochemical, metabolic, and live imaging readouts showed that, relative to controls, skin keratinocytes null for Krt6a/Krt6b or Krt16 exhibit elevated ROS, reduced mitochondrial respiration, intracellular distribution differences, and altered movement of mitochondria within the cell. These findings highlight a novel role for K6 and K16 in regulating mitochondrial morphology, dynamics, and function and shed new light on the causes of oxidative stress observed in PC and related keratin-based skin disorders.


Subject(s)
Keratins/metabolism , Mitochondria/metabolism , Skin/metabolism , Animals , Cytoskeletal Proteins , Epidermis , Female , Keratin-16/genetics , Keratin-16/metabolism , Keratin-6/genetics , Keratin-6/metabolism , Keratinocytes/metabolism , Keratinocytes/physiology , Keratins/physiology , Keratoderma, Palmoplantar , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/physiology , Mutation , Pachyonychia Congenita
6.
Br J Dermatol ; 182(3): 564-573, 2020 03.
Article in English | MEDLINE | ID: mdl-31021398

ABSTRACT

BACKGROUND: Pachyonychia congenita (PC), a rare genodermatosis, primarily affects ectoderm-derived epithelial appendages and typically includes oral leukokeratosis, nail dystrophy and very painful palmoplantar keratoderma (PPK). PC dramatically impacts quality of life although it does not affect lifespan. PC can arise from mutations in any of the wound-repair-associated keratin genes KRT6A, KRT6B, KRT6C, KRT16 or KRT17. There is no cure for this condition, and current treatment options for PC symptoms are limited and palliative in nature. OBJECTIVES: This review focuses on recent progress made towards understanding the pathophysiology of PPK lesions, the most prevalent and debilitating of all PC symptoms. METHODS: We reviewed the relevant literature with a particular focus on the Krt16 null mouse, which spontaneously develops footpad lesions that mimic several aspects of PC-associated PPK. RESULTS: There are three main stages of progression of PPK-like lesions in Krt16 null mice. Ahead of lesion onset, keratinocytes in the palmoplantar (footpad) skin exhibit specific defects in terminal differentiation, including loss of Krt9 expression. At the time of PPK onset, there is elevated oxidative stress and hypoactive Keap1-Nrf2 signalling. During active PPK, there is a profound defect in the ability of the epidermis to maintain or return to normal homeostasis. CONCLUSIONS: The progress made suggests new avenues to explore for the treatment of PC-based PPK and deepens our understanding of the mechanisms controlling skin tissue homeostasis. What's already known about this topic? Pachyonychia congenita (PC) is a rare genodermatosis caused by mutations in KRT6A, KRT6B, KRT6C, KRT16 and KRT17, which are normally expressed in skin appendages and induced following injury. Individuals with PC present with multiple clinical symptoms that usually include thickened and dystrophic nails, palmoplantar keratoderma (PPK), glandular cysts and oral leukokeratosis. The study of PC pathophysiology is made challenging because of its low incidence and high complexity. There is no cure or effective treatment for PC. What does this study add? This text reviews recent progress made when studying the pathophysiology of PPK associated with PC. This recent progress points to new possibilities for devising effective therapeutics that may complement current palliative strategies.


Subject(s)
Keratoderma, Palmoplantar , Pachyonychia Congenita , Animals , Homeostasis , Kelch-Like ECH-Associated Protein 1 , Keratin-16/genetics , Keratin-16/metabolism , Keratoderma, Palmoplantar/genetics , Mice , Mutation/genetics , NF-E2-Related Factor 2/metabolism , Pachyonychia Congenita/genetics , Quality of Life
7.
Br J Ophthalmol ; 104(7): 999-1004, 2020 07.
Article in English | MEDLINE | ID: mdl-31585964

ABSTRACT

BACKGROUND/AIMS: Meibomian gland dysfunction (MGD) is the most common form of evaporative dry eye disease, but its pathogenesis is poorly understood. This study examined the histopathological features of meibomian gland (MG) tissue from cadaver donors to identify potential pathogenic processes that underlie MGD in humans. METHODS: Histological analyses was performed on the MGs in the tarsal plates dissected from four cadaver donors, two young and two old adults, including a 36-year-old female (36F) and three males aged 30, 63 and 64 years (30M, 63M and 64M). RESULTS: The MGs of 36F displayed normal anatomy and structure, whereas the MGs of 30M showed severe ductal obstruction with mild distortion. The obstruction was caused by increased cytokeratin levels in association with hyperproliferation, but not hyperkeratinisation. In two older males, moderate to severe MG atrophy was noted. Cell proliferation was significantly reduced in the MG acini of the two older donors as measured by Ki67 labelling index (6.0%±3.4% and 7.9%±2.8% in 63M and 64M, respectively) when compared with that of the two younger donors (23.2%±5.5% and 16.9%±4.8% in 30M and 36F, respectively) (p<0.001). The expression patterns of meibocyte differentiation biomarkers were similar in the older and younger donors. CONCLUSION: Our histopathological study, based on a small sample size, suggests potentially distinct pathogenic mechanisms in MGD. In the young male adult, hyperproliferation and aberrant differentiation of the central ductal epithelia may lead to the obstruction by overproduced cytokeratins. In contrast, in older adults, decreased cell proliferation in acinar basal epithelia could be a contributing factor leading to MG glandular atrophy.


Subject(s)
Biomarkers/metabolism , Dry Eye Syndromes/pathology , Keratin-16/metabolism , Keratin-17/metabolism , Keratin-6/metabolism , Meibomian Gland Dysfunction/pathology , Meibomian Glands/pathology , Adult , Cell Proliferation , Dry Eye Syndromes/metabolism , Female , Humans , Immunoenzyme Techniques , Ki-67 Antigen/metabolism , Male , Meibomian Gland Dysfunction/metabolism , Meibomian Glands/metabolism , Middle Aged , Tissue Donors
8.
Cells ; 8(8)2019 08 01.
Article in English | MEDLINE | ID: mdl-31374826

ABSTRACT

Located at the skin surface, keratinocytes (KCs) are constantly exposed to external stimuli and are the first responders to invading pathogens and injury. Upon skin injury, activated KCs secrete an array of alarmin molecules, providing a rapid and specific innate immune response against danger signals. However, dysregulation of the innate immune response of KCs may lead to uncontrolled inflammation and psoriasis pathogenesis. Keratins (KRT) are the major structural intermediate filament proteins in KCs and are expressed in a highly specific pattern at different differentiation stages of KCs. While KRT14-KRT5 is restricted to basal proliferative KCs, and KRT10-KRT1 is restricted to suprabasal differentiated KCs in normal skin epidermis, the wound proximal KCs downregulate KRT10-K1 and upregulate KRT16/KRT17-KRT6 upon skin injury. Recent studies have recognized KRT6/16/17 as key early barrier alarmins and upregulation of these keratins alters proliferation, cell adhesion, migration and inflammatory features of KCs, contributing to hyperproliferation and innate immune activation of KCs in response to an epidermal barrier breach, followed by the autoimmune activation of T cells that drives psoriasis. Here, we have reviewed how keratins are dysregulated during skin injury, their roles in wound repairs and in initiating the innate immune system and the subsequent autoimmune amplification that arises in psoriasis.


Subject(s)
Keratin-16/metabolism , Keratin-17/metabolism , Keratin-6/metabolism , Psoriasis/metabolism , Wound Healing , Alarmins/metabolism , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Proliferation , Gene Expression Regulation , Humans , Immunity, Innate
9.
Int J Biol Sci ; 15(7): 1419-1428, 2019.
Article in English | MEDLINE | ID: mdl-31337972

ABSTRACT

Objectives: keratin 16 (KRT16) is a type I cytokeratin that overexpressed in many kinds of cancers, but unlike other keratins, KRT16 was poorly studied, so the aim of current study was to determine the biological role of KRT16 in lung adenocarcinoma (LUAD). Materials and Methods: by utilizing open access data, we determined KRT16 expression in LUAD. After that we evaluated the biological role of KRT16 in-vitro and in-vivo. We also explored the reason for KRT16 overexpression. Last, we explored the clinical significance of KRT16 in LUAD. Results: we found KRT16 is overexpressed in LUAD and positively correlated with lymph node metastasis. Knockdown of KRT16 significantly influenced the LUAD cells' migration, invasion, proliferation and epithelial-mesenchymal transition (EMT). Moreover, TFAP2A could transcriptionally overexpress KRT16, which contributed to the TFAP2A tumorigenicity. Last, we determined that high level of KRT16 predicts poor prognosis of LUAD patients. Conclusions: our data indicate that, TFAP2A induced KRT16 overexpression promotes tumorigenicity in LUAD via EMT, and KRT16 expression could serve as an independent prognosis marker.


Subject(s)
Adenocarcinoma/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Keratin-16/metabolism , Lung Neoplasms/metabolism , Transcription Factor AP-2/metabolism , Adenocarcinoma/genetics , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Profiling , Humans , Lung Neoplasms/genetics , Lymphatic Metastasis , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Oncogenes , Prognosis , Tissue Array Analysis , Wound Healing
10.
Hum Mol Genet ; 28(13): 2255-2270, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31220272

ABSTRACT

The type I intermediate filament keratin 16 (KRT16 gene; K16 protein) is constitutively expressed in ectoderm-derived appendages and in palmar/plantar epidermis and is robustly induced when the epidermis experiences chemical, mechanical or environmental stress. Missense mutations at the KRT16 locus can cause pachyonychia congenita (PC, OMIM:167200) or focal non-epidermolytic palmoplantar keratoderma (FNEPPK, OMIM:613000), which each entail painful calluses on palmar and plantar skin. Krt16-null mice develop footpad lesions that mimic PC-associated PPK, providing an opportunity to decipher its pathophysiology, and develop therapies. We report on insight gained from a genome-wide analysis of gene expression in PPK-like lesions of Krt16-null mice. Comparison of this data set with publicly available microarray data of PPK lesions from individuals with PC revealed significant synergies in gene expression profiles. Keratin 9 (Krt9/K9), the most robustly expressed gene in differentiating volar keratinocytes, is markedly downregulated in Krt16-null paw skin, well-ahead of lesion onset, and is paralleled by pleiotropic defects in terminal differentiation. Effective prevention of PPK-like lesions in Krt16-null paw skin (via topical delivery of the Nrf2 inducer sulforaphane) involves the stimulation of Krt9 expression. These findings highlight a role for defective terminal differentiation and loss of Krt9/K9 expression as additional drivers of PC-associated PPK and highlight restoration of KRT9 expression as a worthy target for therapy. Further, we report on the novel observation that keratin 16 can localize to the nucleus of epithelial cells, implying a potential nuclear function that may be relevant to PC and FNEPPK.


Subject(s)
Keratin-16/genetics , Keratin-9/metabolism , Keratinocytes/cytology , Keratoderma, Palmoplantar/genetics , Animals , Cell Differentiation , Dermis/drug effects , Dermis/physiopathology , HeLa Cells , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Isothiocyanates/therapeutic use , Kelch-Like ECH-Associated Protein 1/metabolism , Keratin-16/metabolism , Keratin-9/genetics , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratins/metabolism , Keratoderma, Palmoplantar/drug therapy , Keratoderma, Palmoplantar/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Mutation, Missense , NF-E2-Related Factor 2/metabolism , Signal Transduction , Sulfoxides , Tissue Array Analysis
11.
Kaohsiung J Med Sci ; 35(5): 284-296, 2019 May.
Article in English | MEDLINE | ID: mdl-30942529

ABSTRACT

Psoriasis is a multisystem disease affecting about 2% of the population, while keratin16 (KRT16) has been reported to participate in psoriasis. However, the specific mechanism of KRT16 in psoriasis was inadequately investigated. The objective of the study was to elucidate the mechanism by which siRNA-mediated silencing of KRT16 affects keratinocyte proliferation and vascular endothelial growth factor (VEGF) secretion in psoriasis through the extracellular signal-related kinase (ERK) signaling pathway. Psoriasis-related core gene KRT16 was screened out. Then, the expression of KRT16, VEGF, and ERK signaling pathway-related genes was detected in psoriatic patients. To further investigate the mechanism of KRT16, keratinocytes in psoriatic patients were treated with KRT16 siRNA or/and ERK inhibitor (PD98059) to detect the changes in related gene expression and cell survival. KRT16 was involved in psoriasis development. The expression levels of KRT16, p-ERK1/2, and VEGF in lesion tissues are significantly elevated. Keratinocytes treated with KRT16-siRNA and KRT16-siRNA + PD98059 exhibited reduced KRT16, p-ERK1/2, and VEGF expression. The cell survival rate in cells treated with KRT16-siRNA, PD98059, and KRT16-siRNA + PD98059 reduced significantly. These findings indicate that silencing KRT16 inhibits keratinocyte proliferation and VEGF secretion in psoriasis via inhibition of ERK signaling pathway, which provides a basic theory in the treatment of psoriasis.


Subject(s)
Keratin-16/genetics , Keratinocytes/metabolism , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Psoriasis/genetics , Vascular Endothelial Growth Factor A/genetics , Adult , Aged , Case-Control Studies , Cell Proliferation , Female , Flavonoids/pharmacology , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Keratin-16/antagonists & inhibitors , Keratin-16/metabolism , Keratinocytes/drug effects , Keratinocytes/pathology , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Psoriasis/metabolism , Psoriasis/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
13.
Int Immunopharmacol ; 65: 84-95, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30293051

ABSTRACT

Gallic acid (GA), a natural small molecule found in Radix Paeoniae Rubra, has a variety of favorable biological activities. However, the anti-psoriasis effect of GA has never been explored up to now. This study evaluates the protective effect of GA on psoriasis-like skin disease both in vitro and in vivo and explores the underlying mechanism. The results show that GA significantly decreases the mRNA and protein expression of keratin 16 and keratin 17 which are the markers of psoriasis. Additionally, GA obviously ameliorates psoriasis area and severity index scores and decreases the epidermal hyperplasia of psoriasis-like disease mice. The activity of Nrf2 which targets keratin 16 and keratin 17 is significantly downregulated by GA. Furthermore, the downregulation of keratin 16 and keratin 17 induced by GA was abolished by the Nrf2-overexpression in vitro. This study initially elucidates the anti-psoriasis effect and mechanism of GA which hints that GA would be a potential candidate for the treatment of psoriasis.


Subject(s)
Gallic Acid/pharmacology , Keratin-16/metabolism , Keratin-17/metabolism , NF-E2-Related Factor 2/metabolism , Psoriasis/chemically induced , Animals , Cell Line , Cell Survival , Gallic Acid/chemistry , Gene Expression Regulation/drug effects , Humans , Interleukin-17/toxicity , Keratin-16/antagonists & inhibitors , Keratin-16/genetics , Keratin-17/antagonists & inhibitors , Keratin-17/genetics , Male , Mice , Mice, Inbred BALB C , Molecular Structure , NF-E2-Related Factor 2/genetics , Psoriasis/drug therapy , Psoriasis/metabolism
14.
J Dermatol Sci ; 87(3): 236-245, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28673488

ABSTRACT

BACKGROUND: Psoriasis is an inflammatory skin disease with aberrant keratinocyte proliferation, presumably as a result of immune cell activation. Th17 cytokines like IL-17A and IL-22 are critically implicated in epidermal thickening, altered keratinocyte differentiation and production of innate factors such as antimicrobial peptides. Psoriasis treatment options include modern targeted therapies using anti-cytokine antibodies and traditional non-targeted treatments like anthralin (dithranol). While the mode of action of anti-cytokine antibodies is defined, the effects of topical anthralin on psoriatic skin are not fully understood. OBJECTIVE: This study aims to unravel the direct effects of anthralin on keratinocyte proliferation, differentiation and production of psoriasis-associated factors. METHODS: We tested the effects of anthralin on cell proliferation, cytokeratin expression and changes in the expression of antimicrobial peptides using primary keratinocytes and 3D psoriasis tissue models with and without stimulation of the psoriasis-promoting cytokines IL-17A and IL-22. Moreover, we compared the findings derived from monolayer and multilayer cultures to data derived from lesional skin of patients with psoriasis before and under treatment with anthralin. RESULTS: Our study shows that anthralin directly induces cell apoptosis in vitro in monolayer cultures but not in 3D psoriasis tissue models treated with IL-17A and IL-22. Yet, keratinocyte proliferation as determined by Ki-67 staining is impaired by anthralin in vivo. In lesional skin but not in 3D psoriasis tissue models anthralin rapidly normalizes cytokeratin (CK)16 expression. Furthermore, anthralin directly inhibits DEFB4 expression in vitro and in vivo, while other antimicrobial peptides and cytokines studied like IL-6 and IL-8 are regulated differently in vitro and in vivo. CONCLUSIONS: Our results show that anthralin directly regulates DEFB4A expression. However, its beneficial effects on psoriasis cannot be explained by direct effects on keratinocyte differentiation or cytokine expression.


Subject(s)
Anthralin/pharmacology , Dermatologic Agents/pharmacology , Keratin-16/metabolism , Keratinocytes/drug effects , Psoriasis/drug therapy , beta-Defensins/metabolism , Administration, Cutaneous , Anthralin/therapeutic use , Apoptosis/drug effects , Biopsy , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dermatologic Agents/therapeutic use , Fluorescent Antibody Technique , Humans , Interleukin-17/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Interleukins/metabolism , Keratin-10/metabolism , Keratin-5/metabolism , Keratinocytes/metabolism , Ki-67 Antigen/metabolism , Psoriasis/pathology , Skin/cytology , Skin/drug effects , Skin/pathology , Tissue Culture Techniques/methods , Interleukin-22
15.
Nat Commun ; 8: 14174, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28128203

ABSTRACT

Keratin 16 (K16) is a cytoskeletal scaffolding protein highly expressed at pressure-bearing sites of the mammalian footpad. It can be induced in hyperproliferative states such as wound healing, inflammation and cancer. Here we show that the inactive rhomboid protease RHBDF2 (iRHOM2) regulates thickening of the footpad epidermis through its interaction with K16. K16 expression is absent in the thinned footpads of irhom2-/- mice compared with irhom2+/+mice, due to reduced keratinocyte proliferation. Gain-of-function mutations in iRHOM2 underlie Tylosis with oesophageal cancer (TOC), characterized by palmoplantar thickening, upregulate K16 with robust downregulation of its type II keratin binding partner, K6. By orchestrating the remodelling and turnover of K16, and uncoupling it from K6, iRHOM2 regulates the epithelial response to physical stress. These findings contribute to our understanding of the molecular mechanisms underlying hyperproliferation of the palmoplantar epidermis in both physiological and disease states, and how this 'stress' keratin is regulated.


Subject(s)
Carrier Proteins/metabolism , Epidermis/physiology , Keratin-16/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Cell Proliferation/physiology , Cytoskeleton/physiology , Down-Regulation , Epidermal Cells , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Fibroblasts , Gain of Function Mutation , Humans , Intracellular Signaling Peptides and Proteins , Keratin-6/metabolism , Keratinocytes/physiology , Keratoderma, Palmoplantar/genetics , Keratoderma, Palmoplantar/pathology , Male , Mice , Mice, Knockout , Pressure , RNA, Small Interfering/metabolism , Stress, Physiological/physiology , Tissue Culture Techniques , Up-Regulation , Wound Healing/physiology
16.
J Am Acad Dermatol ; 76(3): 449-453.e1, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27889290

ABSTRACT

BACKGROUND: Epidermolysis bullosa simplex is a skin-blistering disorder caused by mutations in keratin (K)14 or K5. Treatment with nuclear factor (erythroid-derived 2)-like 2 inducer sulforaphane ameliorated skin blistering in Krt14-null mice, correlating with induction of K17. To be therapeutically useful for epidermolysis bullosa simplex, topical broccoli sprout extract (BSE), enriched for sulforaphane, would ideally induce the expression of homologous keratins (eg, K6, K17, K16) in the basal layer of human epidermis without impacting expression of defective keratins (K5/K14). OBJECTIVE: The purpose of this 1-week, randomized, split-body, single-blinded, placebo-controlled trial was to assess the impact of BSE on keratin expression. METHODS: Five subjects (34-71 years old) applied BSE (500 nmol of sulforaphane/mL) or vehicle alone to the inner aspect of the arm daily. Expression of keratin, nuclear factor (erythroid-derived 2)-like 2, and other markers was assessed using reverse transcription-polymerase chain reaction and indirect immunofluorescence. RESULTS: One subject (age 71 years) was excluded a posteriori because of poor tissue quality. Topical BSE activated nuclear factor (erythroid-derived 2)-like 2 and up-regulated K17 in the epidermis of all subjects, had variable effects on K16 and K6 expression, and did not alter expression of K14 or K5. LIMITATIONS: Small sample size is a limitation. CONCLUSION: BSE represents an attractive therapeutic candidate for K14-associated epidermolysis bullosa simplex.


Subject(s)
Brassica , Epidermolysis Bullosa Simplex/drug therapy , Epidermolysis Bullosa Simplex/metabolism , Keratins/metabolism , Phytotherapy , Plant Extracts/therapeutic use , Administration, Cutaneous , Adult , Aged , Humans , Keratin-14/genetics , Keratin-14/metabolism , Keratin-16/genetics , Keratin-16/metabolism , Keratin-17/genetics , Keratin-17/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Keratin-6/genetics , Keratin-6/metabolism , Middle Aged , NF-E2-Related Factor 2/metabolism , Plant Extracts/administration & dosage , RNA, Messenger/metabolism , Seedlings , Single-Blind Method , Up-Regulation/drug effects
18.
J Invest Dermatol ; 137(3): 603-613, 2017 03.
Article in English | MEDLINE | ID: mdl-27825969

ABSTRACT

Current atopic dermatitis (AD) models link epidermal abnormalities in lesional skin to cytokine activation. However, there is evolving evidence of systemic immune activation and detectable abnormalities in nonlesional skin. Because some of the best single correlations with severity (Scoring of AD, or SCORAD) are detected not only in lesional but also nonlesional skin and blood, more complex biomarker models of AD are needed. We thus performed extensive biomarker measures in these compartments using univariate and multivariate approaches to correlate disease biomarkers with SCORAD and with a combined hyperplasia score [thickness and keratin 16 (K16) mRNA] at baseline and after cyclosporine A treatment in 25 moderate to severe AD patients. Increases in serum cytokines and chemokines (IL-13, IL-22, CCL17) were found in AD versus healthy individuals and were reduced with treatment. SCORAD correlated with immune (IL-13, IL-22) and epidermal (thickness, K16) measures in lesional and, even more strongly, in nonlesional AD. Serum cytokines also had higher correlations with nonlesional markers at baseline and with treatment. Multivariate U statistics improved baseline and treatment-response SCORAD correlations. Nonlesional models showed the strongest correlations, with further improvement upon integration of serum markers. Even better correlations were obtained between biomarkers and the hyperplasia score. Larger cohorts are needed to confirm these preliminary data.


Subject(s)
Biomarkers/blood , Dermatitis, Atopic/blood , Dermatitis, Atopic/immunology , Adolescent , Adult , Aged , Chemokine CCL17/metabolism , Cohort Studies , Cyclosporine/therapeutic use , Epidermis/metabolism , Female , Humans , Interleukin-13/metabolism , Interleukins/metabolism , Keratin-16/metabolism , Male , Middle Aged , Multivariate Analysis , Th2 Cells/immunology , Young Adult , Interleukin-22
19.
J Clin Invest ; 126(6): 2356-66, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183391

ABSTRACT

Palmoplantar keratoderma (PPK) are debilitating lesions that arise in individuals with pachyonychia congenita (PC) and feature upregulation of danger-associated molecular patterns and skin barrier regulators. The defining features of PC-associated PPK are reproduced in mice null for keratin 16 (Krt16), which is commonly mutated in PC patients. Here, we have shown that PPK onset is preceded by oxidative stress in footpad skin of Krt16-/- mice and correlates with an inability of keratinocytes to sustain nuclear factor erythroid-derived 2 related factor 2-dependent (NRF2-dependent) synthesis of the cellular antioxidant glutathione (GSH). Additionally, examination of plantar skin biopsies from individuals with PC confirmed the presence of high levels of hypophosphorylated NRF2 in lesional tissue. In Krt16-/- mice, genetic ablation of Nrf2 worsened spontaneous skin lesions and accelerated PPK development in footpad skin. Hypoactivity of NRF2 in Krt16-/- footpad skin correlated with decreased levels or activity of upstream NRF2 activators, including PKCδ, receptor for activated C kinase 1 (RACK1), and p21. Topical application of the NRF2 activator sulforaphane to the footpad of Krt16-/- mice prevented the development of PPK and normalized redox balance via regeneration of GSH from existing cellular pools. Together, these findings point to oxidative stress and dysfunctional NRF2 as contributors to PPK pathogenesis, identify K16 as a regulator of NRF2 activation, and suggest that pharmacological activation of NRF2 should be further explored for PC treatment.


Subject(s)
NF-E2-Related Factor 2/metabolism , Pachyonychia Congenita/metabolism , Animals , Disease Models, Animal , Glutathione/biosynthesis , Humans , Isothiocyanates/pharmacology , Keratin-16/genetics , Keratin-16/metabolism , Keratoderma, Palmoplantar/genetics , Keratoderma, Palmoplantar/metabolism , Keratoderma, Palmoplantar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/chemistry , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Oxidative Stress , Pachyonychia Congenita/genetics , Pachyonychia Congenita/pathology , Phenotype , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sulfoxides
20.
PLoS One ; 11(3): e0150635, 2016.
Article in English | MEDLINE | ID: mdl-26938103

ABSTRACT

Transcription factors are key molecules that finely tune gene expression in response to injury. We focused on the role of a transcription factor, Foxn1, whose expression is limited to the skin and thymus epithelium. Our previous studies showed that Foxn1 inactivity in nude mice creates a pro-regenerative environment during skin wound healing. To explore the mechanistic role of Foxn1 in the skin wound healing process, we analyzed post-injured skin tissues from Foxn1::Egfp transgenic and C57BL/6 mice with Western Blotting, qRT-PCR, immunofluorescence and flow cytometric assays. Foxn1 expression in non-injured skin localized to the epidermis and hair follicles. Post-injured skin tissues showed an intense Foxn1-eGFP signal at the wound margin and in leading epithelial tongue, where it co-localized with keratin 16, a marker of activated keratinocytes. This data support the concept that suprabasal keratinocytes, expressing Foxn1, are key cells in the process of re-epithelialization. The occurrence of an epithelial-mesenchymal transition (EMT) was confirmed by high levels of Snail1 and Mmp-9 expression as well as through co-localization of vimentin/E-cadherin-positive cells in dermis tissue at four days post-wounding. Involvement of Foxn1 in the EMT process was verified by co-localization of Foxn1-eGFP cells with Snail1 in histological sections. Flow cytometric analysis showed the increase of double positive E-cadherin/N-cadherin cells within Foxn1-eGFP population of post-wounded skin cells isolates, which corroborated histological and gene expression analyses. Together, our findings indicate that Foxn1 acts as regulator of the skin wound healing process through engagement in re-epithelization and possible involvement in scar formation due to Foxn1 activity during the EMT process.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Forkhead Transcription Factors/genetics , Skin/metabolism , Wound Healing/genetics , Wounds, Penetrating/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Keratin-16/genetics , Keratin-16/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Skin/injuries , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Vimentin/genetics , Vimentin/metabolism , Wounds, Penetrating/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...