Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 335
1.
Cell Death Dis ; 15(4): 252, 2024 Apr 08.
Article En | MEDLINE | ID: mdl-38589352

Cutaneous squamous carcinoma is the second most common epithelial malignancy, associated with significant morbidity, mortality, and economic burden. However, the mechanisms underlying cSCC remain poorly understood. In this study, we identified TGM3 as a novel cSCC tumor suppressor that acts via the PI3K-AKT axis. RT-qPCR, IHC and western blotting were employed to assess TGM3 levels. TGM3-overexpression/knockdown cSCC cell lines were utilized to detect TGM3's impact on epithelial differentiation as well as tumor cell proliferation, migration, and invasion in vitro. Additionally, subcutaneous xenograft tumor models were employed to examine the effect of TGM3 knockdown on tumor growth in vivo. Finally, molecular and biochemical approaches were employed to gain insight into the tumor-suppressing mechanisms of TGM3. TGM3 expression was increased in well-differentiated cSCC tumors, whereas it was decreased in poor-differentiated cSCC tumors. Loss of TGM3 is associated with poor differentiation and a high recurrence rate in patients with cSCC. TGM3 exhibited tumor-suppressing activity by regulating cell proliferation, migration, and invasion both in vitro and in vivo. As a novel cSCC tumor differentiation marker, TGM3 expression was positively correlated with cell differentiation. In addition, our results demonstrated an interaction between TGM3 and KRT14 that aids in the degradation of KRT14. TGM3 deficiency disrupts keratinocytes differentiation, and ultimately leads to tumorigenesis. Furthermore, RNA-sequence analysis revealed that loss of TGM3 enhanced EMT via the PI3K-AKT signaling pathway. Deguelin, a PI3K-AKT inhibitor, blocked cSCC tumor growth induced by TGM3 knockdown in vivo. Taken together, TGM3 inhibits cSCC tumor growth via PI3K-AKT signaling, which could also serve as a tumor differentiation marker and a potential therapeutic target for cSCC. Proposed model depicted the mechanism by which TGM3 suppress cSCC development. TGM3 reduces the phosphorylation level of AKT and degrades KRT14. In the epithelial cell layer, TGM3 exhibits a characteristic pattern of increasing expression from bottom to top, while KRT14 and pAKT are the opposite. Loss of TGM3 leads to reduced degradation of KRT14 and activation of pAKT, disrupting keratinocyte differentiation, and eventually resulting in the occurrence of low-differentiated cSCC.


Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Keratin-14/genetics , Keratin-14/metabolism , Carcinoma, Squamous Cell/metabolism , Signal Transduction , Cell Proliferation/genetics , Cell Differentiation , Antigens, Differentiation , Transglutaminases/genetics , Transglutaminases/metabolism , Cell Line, Tumor
2.
Pathol Res Pract ; 257: 155280, 2024 May.
Article En | MEDLINE | ID: mdl-38608372

In breast conservative surgery, it is sometimes difficult to decide whether the cauterised tissue at the inked margin represents normal / hyperplastic or neoplastic tissue. We retrospectively assessed the value of ER, PR, CK5 and CK14 IHC in clarifying the nature of cauterised tissues at the margins concerning 34 lesions of 23 patients. 27 cases belonged to lesions that could not be adequately classified on the basis of the HE stains. Two thirds of them could be classified as non-neoplastic or neoplastic and two thirds of the remaining could be favourised as neoplastic or non-neoplastic, with 3/27 cases remaining uncertain. All 4 IHC reactions were helpful in classifying the lesions in almost half of the cases. However, 3 or 4 immunostains were supportive of the classification in 19/27. The most useful stains were the keratins, generally demonstrating a matching pattern of cell labelling with CK5 and CK14. ER and PR were somewhat less useful in classifying uncertain lesions. Considering all the 27 questionable lesions, IHC with ER, PR, CK5 and CK14 clarified the lesions at the cauterised margins in 23 cases. Taken all these considerations into account, CK5, CK14, PR and ER IHC may help in distinguishing between cautery damaged neoplastic and non-neoplastic tissues. All four IHC may yield the best support for decision making, but CK5 and/or CK14 may be sufficient in their own. The essential approach is that the results must be interpreted with caution, in the context of the given patient's disease, to avoid misinterpretations.


Biomarkers, Tumor , Breast Neoplasms , Immunohistochemistry , Keratin-14 , Keratin-5 , Margins of Excision , Receptors, Estrogen , Receptors, Progesterone , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Breast Neoplasms/metabolism , Receptors, Progesterone/metabolism , Keratin-5/metabolism , Keratin-5/analysis , Receptors, Estrogen/metabolism , Retrospective Studies , Middle Aged , Keratin-14/metabolism , Keratin-14/analysis , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Mastectomy, Segmental , Aged , Adult , Cell Proliferation
3.
Genesis ; 62(2): e23596, 2024 Apr.
Article En | MEDLINE | ID: mdl-38665067

The vomeronasal organ (VNO) is a part of the accessory olfactory system, which detects pheromones and chemical factors that trigger a spectrum of sexual and social behaviors. The vomeronasal epithelium (VNE) shares several features with the epithelium of the main olfactory epithelium (MOE). However, it is a distinct neuroepithelium populated by chemosensory neurons that differ from the olfactory sensory neurons in cellular structure, receptor expression, and connectivity. The vomeronasal organ of rodents comprises a sensory epithelium (SE) and a thin non-sensory epithelium (NSE) that morphologically resembles the respiratory epithelium. Sox2-positive cells have been previously identified as the stem cell population that gives rise to neuronal progenitors in MOE and VNE. In addition, the MOE also comprises p63 positive horizontal basal cells, a second pool of quiescent stem cells that become active in response to injury. Immunolabeling against the transcription factor p63, Keratin-5 (Krt5), Krt14, NrCAM, and Krt5Cre tracing experiments highlighted the existence of horizontal basal cells distributed along the basal lamina of SE of the VNO. Single cell sequencing and genetic lineage tracing suggest that the vomeronasal horizontal basal cells arise from basal progenitors at the boundary between the SE and NSE proximal to the marginal zones. Moreover, our experiments revealed that the NSE of rodents is, like the respiratory epithelium, a stratified epithelium where the p63/Krt5+ basal progenitor cells self-replicate and give rise to the apical columnar cells facing the lumen of the VNO.


Vomeronasal Organ , Vomeronasal Organ/metabolism , Vomeronasal Organ/cytology , Animals , Mice , Olfactory Mucosa/metabolism , Olfactory Mucosa/cytology , Keratin-15/metabolism , Keratin-15/genetics , Keratin-5/metabolism , Keratin-5/genetics , Keratin-14/metabolism , Keratin-14/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
4.
Dev Biol ; 504: 120-127, 2023 12.
Article En | MEDLINE | ID: mdl-37813160

The current gold-standard for genetic lineage tracing in transgenic mice is based on cell-type specific expression of Cre recombinase. As an alternative, we developed a cell-type specific CRISPR/spCas9 system for lineage tracing. This method relies on RNA polymerase II promoter driven self-cleaving guide RNAs (scgRNA) to achieve tissue-specificity. To demonstrate proof-of-principle for this approach a transgenic mouse was generated harbouring a knock-in of a scgRNA into the Cytokeratin 14 (Krt14) locus. Krt14 expression marks the stem cells of squamous epithelium in the skin and oral mucosa. The scgRNA targets a Stop cassette preceding a fluorescent reporter in the Ai9-tdtomato mouse. Ai9-tdtomato reporter mice harbouring this allele along with a spCas9 transgene demonstrated precise marking of the Krt14 lineage. We conclude that RNA polymerase II promoter driven scgRNAs enable the use of CRISPR/spCas9 for genetic lineage tracing.


CRISPR-Cas Systems , RNA Polymerase II , Animals , Mice , CRISPR-Cas Systems/genetics , Integrases/genetics , Keratin-14/genetics , Keratin-14/metabolism , Mice, Transgenic , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
5.
Int J Mol Sci ; 24(14)2023 Jul 21.
Article En | MEDLINE | ID: mdl-37511501

The Wnt/ß-catenin signaling pathway plays important roles in the multi-phases of wound healing: homeostasis, inflammation, proliferative, and remodeling phases. However, there are no clinically available therapeutic agents targeting the Wnt/ß-catenin pathway. In this study, we tested the effect of 5, 6-dichloroindirubin-3'-methoxime (KY19382), a small molecule that activates the Wnt/ß-catenin pathway via interference with the function of the negative feedback regulator CXXC5, on cutaneous wound healing. KY19382 significantly enhanced cell migration of human keratinocytes and dermal fibroblasts with increased levels of ß-catenin, phalloidin, Keratin 14, proliferating cell nuclear antigen (PCNA), Collagen I, and alpha-smooth muscle actin (α-SMA) by activating the Wnt/ß-catenin signaling pathway without causing significant cytotoxicity. In addition, levels of Collagen I, Keratin 14, PCNA, and stem cell markers were significantly increased by KY19382 in a cutaneous murine wound healing model. Moreover, KY19382 treatment accelerated re-epithelialization and neo-epidermis formation with collagen deposition and stem cell activation at an early stage of cutaneous wound healing. Overall, KY19382 accelerates wound healing via activating the Wnt/ß-catenin pathway, and may have the potential to be used for the development of a new wound healing agent.


Wnt Signaling Pathway , beta Catenin , Mice , Humans , Animals , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Keratin-14/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Wnt Proteins/metabolism , Wound Healing , Collagen/pharmacology , DNA-Binding Proteins/metabolism , Transcription Factors
6.
FASEB J ; 36(12): e22631, 2022 12.
Article En | MEDLINE | ID: mdl-36342387

Macroautophagy/autophagy is critically involved in the process of salivary gland (SG) diseases such as xerostomia, which has a serious impact on quality of life. KRT14+ progenitor cells are found to be the main progenitors for maintaining the ductal homeostasis of the submandibular SGs. In this study, we investigated the role of ATG5 in SG KRT14+ cells in mice and humans. Human labial salivary glands (LSG) from primary Sjogren's syndrome (pSS) and non-pSS patients (normal), and submandibular glands (SMG) from Atg5flox/flox ; Krt14-Cre (cKO) mice were used. ATG5+ KRT14+ and p62+ KRT14+ cells were detected by immunofluorescence staining in LSG. TUNEL, immunofluorescence, immunohistochemistry, and western blot were performed to detect cell death in SMG. Saliva was collected in 12-week-old (12 W) and 32-week-old (32 W) mice, then the concentration of calcium and buffering capacity were detected to analyze the function of SG. We found that LSG from pSS patients showed increased p62 and decreased ATG5 in KRT14+ cells. We further revealed that in 32 W, (1) the function of salivary glands was significantly impaired in cKO mice, (2) cell death increased in cKO mice, but cl-Caspase 3 was not significantly changed, and (3) cleaved gasdermin D increased and was highly expressed in KRT14+ cells of cKO mice. After applying a pyroptosis inhibitor to 32 W mice, the reduced saliva flow rate was rescued. In addition, pyroptosis was also found in KRT14+ cells of pSS patients. Collectively, our results indicate that Atg5 deficiency would induce pyroptosis in mice SG, which could lead to functional impairments of SG.


Sjogren's Syndrome , Humans , Mice , Animals , Sjogren's Syndrome/metabolism , Pyroptosis , Quality of Life , Salivary Glands/metabolism , Salivary Glands, Minor/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Keratin-14/metabolism
7.
Int J Oral Sci ; 14(1): 49, 2022 Oct 10.
Article En | MEDLINE | ID: mdl-36216809

Stem/progenitor cells are important for salivary gland development, homeostasis maintenance, and regeneration following injury. Keratin-14+ (K14+) cells have been recognized as bona fide salivary gland stem/progenitor cells. However, K14 is also expressed in terminally differentiated myoepithelial cells; therefore, more accurate molecular markers for identifying salivary stem/progenitor cells are required. The intraflagellar transport (IFT) protein IFT140 is a core component of the IFT system that functions in signaling transduction through the primary cilia. It is reportedly expressed in mesenchymal stem cells and plays a role in bone formation. In this study, we demonstrated that IFT140 was intensively expressed in K14+ stem/progenitor cells during the developmental period and early regeneration stage following ligation-induced injuries in murine submandibular glands. In addition, we demonstrated that IFT140+/ K14+ could self-renew and differentiate into granular duct cells at the developmental stage in vivo. The conditional deletion of Ift140 from K14+ cells caused abnormal epithelial structure and function during salivary gland development and inhibited regeneration. IFT140 partly coordinated the function of K14+ stem/progenitor cells by modulating ciliary membrane trafficking. Our investigation identified a combined marker, IFT140+/K14+, for salivary gland stem/progenitor cells and elucidated the essential role of IFT140 and cilia in regulating salivary stem/progenitor cell differentiation and gland regeneration.


Salivary Glands , Stem Cells , Animals , Carrier Proteins/metabolism , Cell Differentiation , Keratin-14/metabolism , Mice , Osteogenesis , Salivary Glands/metabolism
8.
Molecules ; 27(20)2022 Oct 21.
Article En | MEDLINE | ID: mdl-36296727

The activation of the Wnt/ß-catenin signaling pathway plays a key role in the wound-healing process through tissue regeneration. The extract of Euodia daniellii Hemsl. (E. daniellii), a member of the Rutaceae family, activates the Wnt/ß-catenin signaling pathway. However, the function of E. daniellii in wound healing has not yet been elucidated. We performed a migration assay to determine the wound-healing effect of E. daniellii extract in vitro using human keratinocytes and dermal fibroblast. In addition, a mouse acute wound model was used to investigate the cutaneous wound-healing effect of E. daniellii extract in vivo and confirm the potential mechanism. E. daniellii extract enhanced the migration of human keratinocytes and dermal fibroblasts via the activation of the Wnt/ß-catenin pathway. Moreover, the E. daniellii extract increased the levels of keratin 14, PCNA, collagen I, and α-SMA, with nuclei accumulation of ß-catenin in vitro. E. daniellii extract also efficiently accelerated re-epithelialization and stimulated wound healing in vivo. Furthermore, we confirmed that hesperidin, one of the components of E. daniellii, efficiently accelerated the migration of human keratinocytes and dermal fibroblasts, as well as wound healing in vivo via the activation of the Wnt/ß-catenin pathway. Overall, E. daniellii extract and its active component, hesperidin, have potential to be used as therapeutic agents for wound healing.


Evodia , Hesperidin , Mice , Animals , Humans , Wnt Signaling Pathway , beta Catenin/metabolism , Keratin-14/metabolism , Hesperidin/pharmacology , Evodia/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Wound Healing , Collagen/metabolism , Fibroblasts/metabolism
9.
Sci Rep ; 12(1): 17820, 2022 10 24.
Article En | MEDLINE | ID: mdl-36280775

Nestin is an intermediate filament protein transiently expressed in neural stem/progenitor cells. We previously demonstrated that outer root sheath (ORS) keratinocytes of adult hair follicles (HFs) in mice descend from nestin-expressing cells, despite being an epithelial cell lineage. This study determined the exact stage when nestin-expressing ORS stem/precursor cells or their descendants appear during HF morphogenesis, and whether they are present in adult HFs. Using Nes-Cre/CAG-CAT-EGFP mice, in which enhanced green fluorescent protein (EGFP) is expressed following Cre-based recombination driven by the nestin promoter, we found that EGFP+ cells appeared in the epithelial layer of embryonic HFs as early as the peg stage. EGFP+ cells in hair pegs were positive for keratin 14 (K14) and K5, but not vimentin, SOX2, SOX10, or S100 alpha 6. Tracing of tamoxifen-induced EGFP+ cells in postnatal Nes-CreERT2/CAG-CAT-EGFP mice revealed labeling of some isthmus HF epithelial cells in the first anagen stage. EGFP+ cells in adult HFs were not immunolabeled for K15, an HF multipotent stem cell marker. However, when hairs were depilated in Nes-CreERT2/CAG-CAT-EGFP mice to induce the anagen stage after tamoxifen injection, the majority of ORS keratinocytes in depilation-induced anagen HFs were labeled for EGFP. Our findings indicate that nestin-expressing unipotent progenitor cells capable of differentiating into ORS keratinocytes are present in HF primordia and adult HFs.


Epithelial Cells , Hair Follicle , Nestin , Animals , Mice , Biomarkers/metabolism , Epithelial Cells/metabolism , Hair Follicle/metabolism , Keratin-14/genetics , Keratin-14/metabolism , Mice, Transgenic , Nestin/genetics , Nestin/metabolism , Tamoxifen/metabolism
10.
Dev Biol ; 491: 1-12, 2022 11.
Article En | MEDLINE | ID: mdl-36049534

Mammalian corneal development is a multistep process, including formation of the corneal epithelium (CE), endothelium and stroma during embryogenesis, followed by postnatal stratification of the epithelial layers and continuous renewal of the epithelium to replace the outermost corneal cells. Here, we employed the Cre-loxP system to conditionally deplete Pax6 proteins in two domains of ocular cells, i.e., the ocular surface epithelium (cornea, limbus and conjunctiva) (OSE) or postnatal CE via K14-cre or Aldh3-cre, respectively. Earlier and broader inactivation of Pax6 in the OSE resulted in thickened OSE with CE and limbal cells adopting the conjunctival keratin expression pattern. More restricted depletion of Pax6 in postnatal CE resulted in an abnormal cornea marked by reduced epithelial thickness despite increased epithelial cell proliferation. Immunofluorescence studies revealed loss of intermediate filament Cytokeratin 12 and diffused expression of adherens junction components, together with reduced tight junction protein, Zonula occludens-1. Furthermore, the expression of Cytokeratin 14, a basal cell marker in apical layers, indicates impaired differentiation of CE cells. Collectively, our data demonstrate that Pax6 is essential for maintaining proper differentiation and strong intercellular adhesion in postnatal CE cells, whereas limbal Pax6 is required to prevent the outgrowth of conjunctival cells to the cornea.


Cornea , Epithelium, Corneal , Animals , Cornea/metabolism , Epithelium, Corneal/metabolism , Keratin-12/metabolism , Keratin-14/metabolism , Keratins/metabolism , Mammals/metabolism , Tight Junction Proteins/metabolism
11.
J Invest Dermatol ; 142(12): 3282-3293, 2022 12.
Article En | MEDLINE | ID: mdl-35691363

Epidermolysis bullosa simplex (EBS) is a severe and potentially life-threatening disorder for which no adequate therapy exists. Most cases are caused by dominant sequence variations in keratin genes K5 or K14, leading to the formation of cytoplasmic keratin aggregates, profound keratinocyte fragility, and cytolysis. We hypothesized that pharmacological reduction of keratin aggregates, which compromise keratinocyte integrity, represents a viable strategy for the treatment of EBS. In this study, we show that the multikinase inhibitor PKC412, which is currently in clinical use for acute myeloid leukemia and advanced systemic mastocytosis, reduced keratin aggregation by 40% in patient-derived K14.R125C EBS-associated keratinocytes. Using a combination of epithelial shear stress assay and real-time impedance spectroscopy, we show that PKC412 restored intercellular adhesion. Molecularly, global phosphoproteomic analysis together with immunoblots using phosphoepitope-specific antibodies revealed that PKC412 treatment altered phosphorylated sites on keratins and desmoplakin. Thus, our data provide a proof of concept to repurpose existing drugs for the targeted treatment of EBS and showcase how one broad-range kinase inhibitor reduced keratin filament aggregation in patient-derived EBS keratinocytes and the fragility of EBS cell monolayers. Our study paves the way for a clinical trial using PKC412 for systemic or local application in patients with EBS.


Epidermolysis Bullosa Simplex , Humans , Epidermolysis Bullosa Simplex/genetics , Epidermolysis Bullosa Simplex/metabolism , Keratins/metabolism , Staurosporine/metabolism , Cytoskeleton/metabolism , Cytoskeletal Proteins/genetics , Keratin-14/genetics , Keratin-14/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Mutation
12.
J Invest Dermatol ; 142(10): 2695-2705.e11, 2022 10.
Article En | MEDLINE | ID: mdl-35490743

Epidermolysis bullosa simplex (EBS), an autosomal dominant skin disorder, is characterized by skin fragility. Genetically, the majority of cases are related to missense sequence variations in two keratin genes K5 or K14, leading to cytolysis of basal keratinocytes (KCs) and intraepidermal blistering. Progress toward the identification of treatments has been hampered by an incomplete understanding of the mechanisms underlying this disease and availability of relevant and reliable in vitro models recapitulating the physiopathological mechanisms. Recent advances in stem cell field have fueled the prospect that these limitations could be overcome, thanks to the availability of disease-specific human induced pluripotent stem cells (hiPSCs). In this study, we generated hiPSC-derived KCs from patients carrying keratin gene K5-dominant sequence variations and compared them with nonaffected hiPSC-derived KCs as well as their primary counterparts. Our results showed that EBS hiPSC-derived KCs displayed proliferative defects, increased capacity to migrate, alteration of extracellular signal‒regulated kinase signaling pathway, and cytoplasmic keratin filament aggregates as observed in primary EBS KCs. Of interest, EBS hiPSC-derived KCs exhibited downregulation of hemidesmosomal proteins, revealing the different effects of keratin gene K5 sequence variations on keratin cytoskeletal organization. With a combination of culture miniaturization and treatment with the chaperone molecule 4-phenylbutyric acid, our results showed that hiPSC-derived KCs represent a suitable model for identifying novel therapies for EBS.


Epidermolysis Bullosa Simplex , Induced Pluripotent Stem Cells , Epidermolysis Bullosa Simplex/metabolism , Extracellular Signal-Regulated MAP Kinases , Humans , Keratin-14/genetics , Keratin-14/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Keratinocytes/metabolism , Keratins/genetics , Keratins/metabolism , Mutation , Phenotype
14.
Biochem Biophys Res Commun ; 586: 55-62, 2022 01 01.
Article En | MEDLINE | ID: mdl-34826701

Salivary gland hypofunction due to radiation therapy for head and neck cancer or Sjögren syndrome may cause various oral diseases, which can lead to a decline in the quality of life. Cell therapy using salivary gland stem cells is a promising method for restoring hypofunction. Herein, we show that salivary gland-like cells can be induced from epithelial tissues that were transdifferentiated from mouse embryonic fibroblasts (MEFs). We introduced four genes, Dnp63a, Tfap2a, Grhl2, and Myc (PTMG) that are known to transdifferentiate fibroblasts into oral mucosa-like epithelium in vivo into MEFs. MEFs overexpressing these genes showed epithelial cell characteristics, such as cobblestone appearance and E-cadherin positivity, and formed oral epithelial-like tissue under air-liquid interface culture conditions. The epithelial sheet detached from the culture dish was infected with adenoviruses encoding Sox9 and Foxc1, which we previously identified as essential factors to induce salivary gland formation. The cells detached from the cell sheet formed spheres 10 days after infection and showed a branching morphology. The spheres expressed genes encoding basal/myoepithelial markers, cytokeratin 5, cytokeratin 14, acinar cell marker, aquaporin 5, and the myoepithelial marker α-smooth muscle actin. The dissociated cells of these primary spheres had the ability to form secondary spheres. Taken together, our results provide a new strategy for cell therapy of salivary glands and hold implications in treating patients with dry mouth.


Acinar Cells/metabolism , Fibroblasts/metabolism , Forkhead Transcription Factors/genetics , SOX9 Transcription Factor/genetics , Salivary Glands/metabolism , Spheroids, Cellular/metabolism , Acinar Cells/cytology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Aquaporin 5/genetics , Aquaporin 5/metabolism , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Transdifferentiation/genetics , Cell- and Tissue-Based Therapy/methods , Embryo, Mammalian , Fibroblasts/cytology , Forkhead Transcription Factors/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Keratin-14/genetics , Keratin-14/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Mice , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOX9 Transcription Factor/metabolism , Salivary Glands/cytology , Spheroids, Cellular/cytology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Theranostics ; 11(20): 9918-9936, 2021.
Article En | MEDLINE | ID: mdl-34815795

Background: Although CREB phosphorylation is known to be essential in UVB/cAMP-stimulated melanogenesis, CREB null mice did not show identifiable pigmentation phenotypes. Here, we show that CREB-regulated transcription co-activator 3 (CRTC3) quantitatively regulates and orchestrates melanogenesis by directly targeting microphthalmia-associated transcription factor (MITF) and regulating the expression of most key melanogenesis-related genes. Methods: We analyzed CRTC3-null, KRT14-SCF transgenic, and their crossover mice. The molecular basis of CRTC3 effects on pigmentation was investigated by histology, melanin/tyrosinase assay, immunoblotting, shRNA, promoter assay, qRT-PCR, and subcellular localization. These analyses were carried out in primary cultured melanocytes, mouse cell lines, normal human cells, co-cultures, and ex vivo human skin. CRTC/CREB activity screening was performed to identify candidate agents for the regulation of melanogenesis. Results: The coat and skin color of CRTC3-null mice was paler due to a reduction in melanin deposition. Melanogenesis-related genes were reduced in CRTC3-deficient cultured melanocytes and tail skin of CRTC3-null mice. Notably, basal levels of MITF present in CRTC3-null mice were sufficient for melanocytic differentiation/survival. Thus CRTC3-null mice showed a comparable number of epidermal melanocytes compared to control mice. Stem cell factor (SCF) introduction by crossing with KRT14-SCF mice increased epidermal melanocytes and melanin deposition in control and CRTC3-null mice, but the skin color remained still light on the CRTC3-null background. Furthermore, we identified the therapeutic potential of altiratinib to inhibit melanogenesis in human melanocytes and human skin effectively and safely. Conclusion: CRTC3 appears to be a key sensor for melanogenesis and can be used as a reversible and tunable tool for selectively regulating melanogenesis without affecting melanocyte integrity. Thus, CRTC3 can also serve as a screening tool for the discovery of ideal melanogenesis-modulating small molecules.


Melanoma/genetics , Skin Pigmentation/genetics , Transcription Factors/metabolism , Animals , Cell Line , Epidermis/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Keratin-14/genetics , Keratin-14/metabolism , Male , Melanins/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Phosphorylation , Primary Cell Culture , Skin/metabolism , Skin Pigmentation/physiology , Transcription Factors/genetics
16.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article En | MEDLINE | ID: mdl-34830328

Epidermolysis bullosa simplex (EBS) is a group of inherited keratinopathies that, in most cases, arise due to mutations in keratins and lead to intraepidermal ruptures. The cellular pathology of most EBS subtypes is associated with the fragility of the intermediate filament network, cytolysis of the basal layer of the epidermis, or attenuation of hemidesmosomal/desmosomal components. Mutations in keratins 5/14 or in other genes that encode associated proteins induce structural disarrangements of different strengths depending on their locations in the genes. Keratin aggregates display impaired dynamics of assembly and diminished solubility and appear to be the trigger for endoplasmic reticulum (ER) stress upon being phosphorylated by MAPKs. Global changes in cellular signaling mainly occur in cases of severe dominant EBS mutations. The spectrum of changes initiated by phosphorylation includes the inhibition of proteasome degradation, TNF-α signaling activation, deregulated proliferation, abnormal cell migration, and impaired adherence of keratinocytes. ER stress also leads to the release of proinflammatory danger-associated molecular pattern (DAMP) molecules, which enhance avalanche-like inflammation. Many instances of positive feedback in the course of cellular stress and the development of sterile inflammation led to systemic chronic inflammation in EBS. This highlights the role of keratin in the maintenance of epidermal and immune homeostasis.


Alarmins/genetics , Epidermis/metabolism , Epidermolysis Bullosa Simplex/genetics , Keratin-14/genetics , Keratin-5/genetics , Keratinocytes/metabolism , Alarmins/metabolism , Endoplasmic Reticulum Stress/genetics , Epidermis/pathology , Epidermolysis Bullosa Simplex/metabolism , Epidermolysis Bullosa Simplex/pathology , Gene Expression Regulation , Humans , Inflammation , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , Intermediate Filaments/ultrastructure , Keratin-14/metabolism , Keratin-5/metabolism , Keratinocytes/pathology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Aggregates , Proteolysis , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Biochem Biophys Res Commun ; 579: 97-104, 2021 11 19.
Article En | MEDLINE | ID: mdl-34597998

Psoriasis is a severe skin disease with significant physical and psychological health consequences. As a typical type of immune disease, both innate and adaptive immunity disorders play key roles in the development of psoriasis. Interleukin (IL)-30 was thought as a natural antagonist of gp130-mediated signaling that affects T helper type 1 and 17 cell polarization by inhibiting IL-6 and IL-27 signaling pathways. Here, we found that, in vitro, IL-30 reduced cytokine levels of HaCaT keratinocytes and dendritic cells (DCs), weakened the maturationS of DCs, inhibited DC-mediated T cell proliferation, and blocked the activation of nuclear factor-κB. In vivo, IL-30 inhibited the development of skin disease in two animal models: Krt14-Vegfa and imiquimod (IMQ)-induced psoriasis-like skin disease. Thus, IL-30 may be useful as a therapeutic agent for controlling psoriasis.


Imiquimod , Interleukins/biosynthesis , Keratin-14/metabolism , Psoriasis/metabolism , Vascular Endothelial Growth Factor A , Adaptive Immunity , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Humans , Inflammation , Interleukins/metabolism , Keratinocytes/cytology , Lymphocytes/cytology , Mice , Signal Transduction
18.
J Exp Clin Cancer Res ; 40(1): 276, 2021 Sep 01.
Article En | MEDLINE | ID: mdl-34470672

BACKGROUND: Leader cells are a subset of cancer cells that coordinate the complex cell-cell and cell-matrix interactions required for ovarian cancer migration, invasion, tumour deposition and are negatively associated with progression-free survival and response to therapy. Emerging evidence suggests leader cells may be enriched in response to chemotherapy, underlying disease recurrence following treatment. METHODS: CRISPR was used to insert a bicistronic T2A-GFP cassette under the native KRT14 (leader cell) promoter. 2D and 3D drug screens were completed in the presence of chemotherapies used in ovarian cancer management. Leader cell; proliferative (Ki67); and apoptotic status (Cleaved Caspase 3) were defined by live cell imaging and flow cytometry. Quantitative real-time PCR defined "stemness" profiles. Proliferation was assessed on the xCELLigence real time cell analyser. Statistical Analysis was performed using unpaired non-parametric t-tests or one-way ANOVA and Tukey's multiple comparison post hoc. RESULTS: Leader cells represent a transcriptionally plastic subpopulation of ovarian cancer cells that arise independently of cell division or DNA replication, and exhibit a "stemness" profile that does not correlate with epithelial-to-mesenchymal transition. Chemotherapeutics increased apoptosis-resistant leader cells in vitro, who retained motility and expressed known chemo-resistance markers including ALDH1, Twist and CD44v6. Functional impairment of leader cells restored chemosensitivity, with leader cell-deficient lines failing to recover following chemotherapeutic intervention. CONCLUSIONS: Our data demonstrate that ovarian cancer leader cells are resistant to a diverse array of chemotherapeutic agents, and are likely to play a critical role in the recurrence of chemo-resistant disease as drivers of poor treatment outcomes.


Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Keratin-14/metabolism , Ovarian Neoplasms/drug therapy , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cisplatin/therapeutic use , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Replication , Female , Green Fluorescent Proteins/genetics , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
19.
Sci Rep ; 11(1): 17296, 2021 08 27.
Article En | MEDLINE | ID: mdl-34453089

Hypertrophic scars represent a common complication in burn patients. In addition to cosmetic defects, they may cause serious sensory abnormalities such as pain and itching, severe dysfunction depending on the site, and emotional disorders such as anxiety and depression. The present study aimed to identify the molecular mechanisms underlying the use of extracorporeal shock wave therapy in keratinocytes. Keratinocytes derived from hypertrophic scar tissue were cultured and expression of proliferation markers (keratin 5 and 14), activation markers (keratin 6 and 17), differentiation markers (keratin 1, 10, and involucrin), apoptosis factors (Bax, Bcl2, and Caspase 14), and proliferation/differentiation regulators (p21 and p27) was investigated to compared with that of those in keratinocytes derived from normal skin tissue. Scar-derived keratinocytes were treated with extracorporeal shock waves under 1000 impulses at 0.1, 0.2, and 0.3 mJ/mm2. Shock waves altered the molecular pattern of proliferation, activation, differentiation, and apoptosis, as well as proliferation/ differentiation regulators, including Bax, Bcl2, ASK1, p21, p27, and Notch1. In summary, we show that extracorporeal shock wave therapy regulates the proliferation and differentiation of keratinocytes derived from hypertrophic scar to maintain normal epidermal integrity.


Cicatrix, Hypertrophic/therapy , Extracorporeal Shockwave Therapy/methods , Keratinocytes/cytology , Biomarkers/metabolism , Caspase 14/metabolism , Cell Differentiation , Humans , Keratin-14/metabolism , Keratin-5/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Skin , Treatment Outcome , bcl-2-Associated X Protein/metabolism
20.
Exp Eye Res ; 210: 108696, 2021 09.
Article En | MEDLINE | ID: mdl-34228968

Studies have reported that the incidence of ocular discomfort in people who often wear makeup is higher than that in the normal population. The incidence of ocular discomfort of these people may be also related to the daily ocular exposure to chemical surfactants during cleaning. The objectives of this study were to explore morphological and pathological changes in the murine ocular surface after low-dose repeated exposure to disodium cocoamphodiacetate (DC), a kind of chemical surfactant widely used in personal cleaning products, and to investigate the possible mechanisms. DC was administered in low dose (0.1%) to the ocular surface of C56BL/6 once daily for two weeks. We found that there were an increase of sodium fluorescein staining on the cornea, a significant thinning of corneal epithelial thickness, and increased TUNEL-positive cells in corneal epithelium in vivo. DC treatment also modulated the distribution of K14+ and P63+ epithelia from the limbal to the center on the cornea. In cultured murine corneal epithelial progenitor cell line (TKE2), DC treatment induced cell detachment and decreased the activation of Ak strain transforming protein (AKT), and extracellular signal-regulated kinase (ERK). And DC increased TUNEL-positive cells in vitro with increased expression of cleaved Caspase3 and B-cell lymphoma-2 associated X protein (Bax). Our results indicated that repeated low-dose DC exposure on ocular surface caused significant impairment on the structure and viability of the corneal epithelium by inhibiting epithelial proliferation and inducing apoptosis. It provides the foundations to understand the harmful effects of cleaning products daily exposure on the ocular surface.


Acetates/adverse effects , Corneal Diseases/chemically induced , Epithelium, Corneal/drug effects , Glycine/analogs & derivatives , Limbus Corneae/drug effects , Surface-Active Agents/adverse effects , Acetates/administration & dosage , Administration, Ophthalmic , Animals , Apoptosis , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Corneal Diseases/metabolism , Corneal Diseases/pathology , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Female , Fluorescein/metabolism , Glycine/administration & dosage , Glycine/adverse effects , Keratin-14/metabolism , Limbus Corneae/metabolism , Limbus Corneae/pathology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Ophthalmic Solutions , Slit Lamp Microscopy , Staining and Labeling , Surface-Active Agents/administration & dosage , Trans-Activators/metabolism
...