Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters











Publication year range
2.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830368

ABSTRACT

Skin aging is a complex process influenced by intrinsic and extrinsic factors. Together, these factors affect the structure and function of the epidermis and dermis. Histologically, aging skin typically shows epidermal atrophy due to decreased cell numbers. The dermis of aged skin shows decreased numbers of mast cells and fibroblasts. Fibroblast senescence contributes to skin aging by secreting a senescence-associated secretory phenotype, which decreases proliferation by impairing the release of essential growth factors and enhancing degradation of the extracellular matrix through activation of matrix metalloproteinases (MMPs). Several molecular mechanisms affect skin aging including telomere shortening, oxidative stress and MMP, cytokines, autophagic control, microRNAs, and the microbiome. Accumulating evidence on the molecular mechanisms of skin aging has provided clinicians with a wide range of therapeutic targets for treating aging skin.


Subject(s)
Atrophy/genetics , Cellular Senescence/genetics , Skin Aging/genetics , Atrophy/pathology , Cell Proliferation/genetics , Epidermal Cells/metabolism , Epidermal Cells/pathology , Fibroblasts/pathology , Humans , Mast Cells/pathology , Matrix Metalloproteinases/genetics , Telomere Shortening/genetics
3.
J Burn Care Res ; 42(6): 1162-1167, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34387313

ABSTRACT

Burn experts are only 77% accurate when subjectively assessing burn depth, leaving almost a quarter of patients to undergo unnecessary surgery or conversely suffer a delay in treatment. To aid clinicians in burn depth assessment (BDA), new technologies are being studied with machine learning algorithms calibrated to histologic standards. Our group has iteratively created a theoretical burn biopsy algorithm (BBA) based on histologic analysis, and subsequently informed it with the largest burn wound biopsy repository in the literature. Here, we sought to report that process. This was an IRB-approved, prospective, multicenter study. A BBA was created a priori and refined in an iterative manner. Patients with burn wounds assessed by burn experts as requiring excision and autograft underwent 4 mm biopsies procured every 25 cm2. Serial still photos were obtained at enrollment and at excision intraoperatively. Burn biopsies were histologically assessed for presence/absence of epidermis, papillary dermis, reticular dermis, and proportion of necrotic adnexal structures by a dermatopathologist using H&E with whole slide scanning. First degree and superficial second degree were considered to be burn wounds likely to have healed without surgery, while deep second- and third-degree burns were considered unlikely to heal by 21 days. Biopsy pathology results were correlated with still photos by five burn experts for consensus of final burn depth diagnosis. Sixty-six subjects were enrolled with 117 wounds and 816 biopsies. The BBA was used to categorize subjects' wounds into four categories: 7% of burns were categorized as first degree, 13% as superficial second degree, 43% as deep second degree, and 37% as third degree. Therefore, 20% of burn wounds were incorrectly judged as needing excision and grafting by the clinical team as per the BBA. As H&E is unable to assess the viability of papillary and reticular dermis, with time our team came to appreciate the greater importance of adnexal structure necrosis over dermal appearance in assessing healing potential. Our study demonstrates that a BBA with objective histologic criteria can be used to categorize BDA with clinical misclassification rates consistent with past literature. This study serves as the largest analysis of burn biopsies by modern day burn experts and the first to define histologic parameters for BDA.


Subject(s)
Algorithms , Burns/pathology , Epidermal Cells/pathology , Burns/classification , Burns/physiopathology , Female , Humans , Male , Prospective Studies , Skin/pathology , Wound Healing
4.
Int J Mol Sci ; 22(15)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34360653

ABSTRACT

Mucopolysaccharidosis (MPS) type I and II are two closely related lysosomal storage diseases associated with disrupted glycosaminoglycan catabolism. In MPS II, the first step of degradation of heparan sulfate (HS) and dermatan sulfate (DS) is blocked by a deficiency in the lysosomal enzyme iduronate 2-sulfatase (IDS), while, in MPS I, blockage of the second step is caused by a deficiency in iduronidase (IDUA). The subsequent accumulation of HS and DS causes lysosomal hypertrophy and an increase in the number of lysosomes in cells, and impacts cellular functions, like cell adhesion, endocytosis, intracellular trafficking of different molecules, intracellular ionic balance, and inflammation. Characteristic phenotypical manifestations of both MPS I and II include skeletal disease, reflected in short stature, inguinal and umbilical hernias, hydrocephalus, hearing loss, coarse facial features, protruded abdomen with hepatosplenomegaly, and neurological involvement with varying functional concerns. However, a few manifestations are disease-specific, including corneal clouding in MPS I, epidermal manifestations in MPS II, and differences in the severity and nature of behavioral concerns. These phenotypic differences appear to be related to different ratios between DS and HS, and their sulfation levels. MPS I is characterized by higher DS/HS levels and lower sulfation levels, while HS levels dominate over DS levels in MPS II and sulfation levels are higher. The high presence of DS in the cornea and its involvement in the arrangement of collagen fibrils potentially causes corneal clouding to be prevalent in MPS I, but not in MPS II. The differences in neurological involvement may be due to the increased HS levels in MPS II, because of the involvement of HS in neuronal development. Current treatment options for patients with MPS II are often restricted to enzyme replacement therapy (ERT). While ERT has beneficial effects on respiratory and cardiopulmonary function and extends the lifespan of the patients, it does not significantly affect CNS manifestations, probably because the enzyme cannot pass the blood-brain barrier at sufficient levels. Many experimental therapies, therefore, aim at delivery of IDS to the CNS in an attempt to prevent neurocognitive decline in the patients.


Subject(s)
Corneal Diseases/complications , Epidermal Cells/pathology , Mucopolysaccharidosis II/pathology , Mucopolysaccharidosis I/pathology , Nervous System Diseases/complications , Animals , Humans , Mucopolysaccharidosis I/etiology , Mucopolysaccharidosis II/etiology
5.
Cell Host Microbe ; 29(8): 1235-1248.e8, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34214492

ABSTRACT

The epidermis forms a barrier that defends the body from desiccation and entry of harmful substances, while also sensing and integrating environmental signals. The tightly orchestrated cellular changes needed for the formation and maintenance of this epidermal barrier occur in the context of the skin microbiome. Using germ-free mice, we demonstrate the microbiota is necessary for proper differentiation and repair of the epidermal barrier. These effects are mediated by microbiota signaling through the aryl hydrocarbon receptor (AHR) in keratinocytes, a xenobiotic receptor also implicated in epidermal differentiation. Mice lacking keratinocyte AHR are more susceptible to barrier damage and infection, during steady-state and epicutaneous sensitization. Colonization with a defined consortium of human skin isolates restored barrier competence in an AHR-dependent manner. We reveal a fundamental mechanism whereby the microbiota regulates skin barrier formation and repair, which has far-reaching implications for the numerous skin disorders characterized by epidermal barrier dysfunction.


Subject(s)
Microbiota/physiology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Skin/microbiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cell Line , Epidermal Cells/metabolism , Epidermal Cells/pathology , Epidermis/metabolism , Female , Humans , Keratinocytes , Male , Mice , Mice, Inbred C57BL , Skin/pathology , Skin Diseases/microbiology
6.
J Invest Dermatol ; 141(9): 2178-2188.e6, 2021 09.
Article in English | MEDLINE | ID: mdl-33984347

ABSTRACT

Maintaining tissue homeostasis depends on a balance between cell proliferation, differentiation, and apoptosis. Within the epidermis, the levels of the polyamines putrescine, spermidine, and spermine are altered in many different skin conditions, yet their role in epidermal tissue homeostasis is poorly understood. We identify the polyamine regulator, Adenosylmethionine decarboxylase 1 (AMD1), as a crucial regulator of keratinocyte (KC) differentiation. AMD1 protein is upregulated on differentiation and is highly expressed in the suprabasal layers of the human epidermis. During KC differentiation, elevated AMD1 promotes decreased putrescine and increased spermine levels. Knockdown or inhibition of AMD1 results in reduced spermine levels and inhibition of KC differentiation. Supplementing AMD1-knockdown KCs with exogenous spermidine or spermine rescued aberrant differentiation. We show that the polyamine shift is critical for the regulation of key transcription factors and signaling proteins that drive KC differentiation, including KLF4 and ZNF750. These findings show that human KCs use controlled changes in polyamine levels to modulate gene expression to drive cellular behavior changes. Modulation of polyamine levels during epidermal differentiation could impact skin barrier formation or can be used in the treatment of hyperproliferative skin disorders.


Subject(s)
Adenosylmethionine Decarboxylase/metabolism , Epidermal Cells/metabolism , Spermine/metabolism , Adenosylmethionine Decarboxylase/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Epidermal Cells/pathology , Gene Knockdown Techniques , Humans , Kruppel-Like Factor 4/metabolism , Mice , Polyamines/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation
7.
Mol Immunol ; 135: 304-311, 2021 07.
Article in English | MEDLINE | ID: mdl-33964631

ABSTRACT

The RIP family plays a key role in mediating cell inflammation, oxidative stress and death. Among them, RIPK1, as an important regulatory factor in the upstream of the NF-κB pathway, is involved in multiple pathways of cell inflammation and death. Epidermal cells constitute the outermost barrier of the human body. Radiation can induce epidermal cell death, inflammation and oxidative stress to cause damage. Therefore, this paper selected HaCaT cell and used CRISPR/Cas technology to construct a cell model of stable knockout of RIPK1 gene, to analyze the effect and regulation of RIPK1 knockout on the function and death of HaCaT cells induced by UVB or TNF-α. The results showed that knockout of RIPK1 had no significant effect on the morphology of HaCaT cells at rest, but it led to slowing cell proliferation and blocking the G2M phase of cell cycle. Compared with HaCaTWT, HaCaTRIP1KO was abnormally sensitive to TNF-α-induced cell death and apoptosis, and may be associated with inhibition of NF-κB pathway. Knocking out RIPK1 led to a more significant inhibition of cell growth by UVB, and up-regulation of the expression of the inflammatory factor IL-1α. P38 MAPK and NF-κB pathways may be involved this process. This study further found that RIPK1 in epidermal cell has a regulatory function on pro-survival signals.


Subject(s)
Apoptosis/physiology , Epidermal Cells/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ultraviolet Rays/adverse effects , CRISPR-Cas Systems/genetics , Cell Line, Transformed , Cell Proliferation/physiology , Cell Survival/physiology , G2 Phase Cell Cycle Checkpoints/physiology , Gene Knockout Techniques , HaCaT Cells , Humans , Interleukin-1alpha/metabolism , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/physiology , Skin/injuries , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Invest Dermatol ; 141(10): 2521-2529.e4, 2021 10.
Article in English | MEDLINE | ID: mdl-33839145

ABSTRACT

Keloid disease is a benign skin disease that does not have an effective therapy. More and more research shows that epidermal abnormalities are involved in keloid pathogenesis. Little is known about the relationship between the abnormal epidermal immunophenotype and clinical outcome. Nine-color flow cytometry with computational analysis was performed to detect the altered cellular subpopulation distribution in keloid lesions. Receiver operating characteristic curves were drawn to compare predictive ability between the alteration of cell subgroup frequency and the Vancouver Scar Scale. The frequency of CD49fhi/CD29+/TLR7+ cellular subsets increased in the keloid epidermis compared with that in the healthy control. CD49fmid-hi/CD29+/TLR7+/CD24+ cellular subpopulation level was increased significantly in keloids, whereas CD49flo-mid/CD29‒/TLR7‒/CD24‒ cellular subpopulation frequency was decreased. The CD49flo/CD29‒/TLR7‒/CD24+/CD117+ cellular subpopulation showed an increased frequency during recurrence with a sensitivity of 66.7% and specificity of 91.7%. The area under the curve was 0.806 for cellular subpopulation analysis, which was higher than the area under the curve for the Vancouver Scar Scale (0.583). The alteration of keloid epidermal subpopulation frequency is related to recurrence, which will provide an optional predictive marker for keloid recurrence and a potential target subset for investigating the generation of keloid.


Subject(s)
Epidermal Cells/pathology , Flow Cytometry/methods , Keloid/pathology , Epidermal Cells/classification , Epidermal Cells/immunology , Female , Humans , Immunophenotyping , Integrin alpha6/analysis , Integrin beta1/analysis , Keloid/immunology , Male , Recurrence , Toll-Like Receptor 7/analysis
9.
Arch Toxicol ; 95(6): 2201-2221, 2021 06.
Article in English | MEDLINE | ID: mdl-33890134

ABSTRACT

Deoxynivalenol (vomitoxin, DON) is a secondary metabolite produced by Fusarium spp. fungi and it is one of the most prevalent mycotoxins worldwide. Crop infestation results not only in food and feed contamination, but also in direct dermal exposure, especially during harvest and food processing. To investigate the potential dermotoxicity of DON, epidermoid squamous cell carcinoma cells A431 were compared to primary human neonatal keratinocytes (HEKn) cells via proteome/phosphoproteome profiling. In A431 cells, 10 µM DON significantly down-regulated ribosomal proteins, as well as mitochondrial respiratory chain elements (OXPHOS regulation) and transport proteins (TOMM22; TOMM40; TOMM70A). Mitochondrial impairment was reflected in altered metabolic competence, apparently combined with interference of the lipid biosynthesis machinery. Functional effects on the cell membrane were confirmed by live cell imaging and membrane fluidity assays (0.1-10 µM DON). Moreover, a common denominator for both A431 and HEKn cells was a significant downregulation of the squalene synthase (FDFT1). In sum, proteome alterations could be traced back to the transcription factor Klf4, a crucial regulator of skin barrier function. Overall, these results describe decisive molecular events sustaining the capability of DON to impair skin barrier function. Proteome data generated in the study are fully accessible via ProteomeXchange with the accession numbers PXD011474 and PXD013613.


Subject(s)
Epidermal Cells/drug effects , Keratinocytes/drug effects , Lipids/biosynthesis , Trichothecenes/toxicity , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Epidermal Cells/pathology , Fusarium/metabolism , Humans , Keratinocytes/pathology , Mitochondria/drug effects , Mitochondria/pathology , Proteomics , Secondary Metabolism , Trichothecenes/administration & dosage , Trichothecenes/isolation & purification
10.
Am J Forensic Med Pathol ; 42(1): 16-22, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-32925207

ABSTRACT

ABSTRACT: The study was undertaken to differentiate antemortem electrical (AME) and postmortem electrical (PME) burn marks with the help of histopathology. The electrical burn mark was produced on 25 dead bodies. Alongside 25 cases of electrocution deaths were included for comparison. Slides were prepared and stained with hematoxylin-eosin stains. Intraepidermal and subepidermal separation; coagulative necrosis of the epidermis; nuclear elongation and hyperchromasia of epidermal cells; homogenization of the dermis; nuclear elongation and hyperchromasia of hair follicles, sweat glands, sebaceous glands, and blood vessel endothelium were studied for histopathological changes and graded. The findings of the study suggest that the histopathological changes in electrical burn marks are due to the physical effect of heat produced by the electric current. The classical histopathological features of electrical burn mark cannot differentiate between AME and PME burn marks. However, careful evaluation of grading of the dermal changes can be helpful in differentiating AME and PME burn marks. Highest grade of dermal thickness homogenization and highest grade of nuclear elongation of dermal appendages were significantly more in the antemortem electrical burn marks than PME burn marks.


Subject(s)
Burns, Electric/pathology , Postmortem Changes , Adolescent , Adult , Aged , Aged, 80 and over , Blood Vessels/pathology , Cadaver , Cell Nucleus Shape , Child , Dermis/pathology , Endothelium, Vascular/pathology , Epidermal Cells/pathology , Epidermis/pathology , Female , Forensic Pathology , Hair Follicle/pathology , Humans , Male , Middle Aged , Sebaceous Glands/pathology , Sweat Glands/pathology , Young Adult
11.
J Allergy Clin Immunol ; 147(6): 2202-2212.e8, 2021 06.
Article in English | MEDLINE | ID: mdl-33338537

ABSTRACT

BACKGROUND: Janus kinase (JAK) inhibitors are a new class of therapeutic compounds for dermatological diseases. In atopic dermatitis (AD), data of clinical phase III trials show rapid improvement of pruritus and significant reduction of inflammation within the first weeks with a favorable safety profile. However, their mode of action in AD is not fully understood. OBJECTIVES: In our study, we investigate the effect of different JAK inhibitors on cell differentiation, phenotype, and function of inflammatory dendritic epidermal cells (IDECs). METHODS: We analyzed the JAK expression in IDEC from ex vivo skin and in vitro generated IDECs using flow cytometry and PCR. Further, we studied in vitro the effect of different JAK inhibitors on IDEC cell differentiation, phenotype, and maturation. RESULTS: IDECs express JAK1 and JAK2 ex vivo and in vitro. We found that JAK1 and JAK2 were upregulated during the differentiation from monocytes to IDECs. Conversely, JAK2 inhibition by ruxolitinib (JAK1/2 inhibitor) or BMS-911543 (JAK2 inhibitor) abrogated the differentiation from monocytes into IDECs. Differentiated IDECs can redifferentiate into a more monocyte-like phenotype in the presence of ruxolitinib or BMS-911543. Furthermore, we showed that concomitant inhibition of JAK1/2 rather than blocking JAK1 or JAK2 alone, impaired maturation and the release of proinflammatory cytokines on lipopolysaccharide stimulation. CONCLUSIONS: Our results suggest that inhibition of JAK1/2 impairs IDEC differentiation and function. We provide new insight into the mode of action of JAK inhibitors in AD and highlight the role of JAK1/2 inhibitors for the treatment of patients with AD.


Subject(s)
Dermatitis, Atopic/etiology , Dermatitis, Atopic/metabolism , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Differentiation , Cytokines/metabolism , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Epidermal Cells/pathology , Gene Expression , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Protein Kinase Inhibitors/therapeutic use
12.
STAR Protoc ; 1(3): 100115, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377011

ABSTRACT

Psoriasis is an incurable chronic inflammatory skin disorder. The imiquimod (IMQ)-induced mouse model of psoriasis is the most widely used model for drug discovery and pre-clinical studies of psoriasis. The inflamed and thickened skin frequently compromises the quality of single-cell suspensions generated from IMQ-induced skin lesions, which has an impact on subsequent analyses by flow cytometry. This protocol details the complete procedure for the establishment of a mouse model of psoriasis and flow cytometric detection of immune cells in the inflamed epidermis and dermis. For complete details on the use and execution of this protocol, please refer to Lou et al. (2020).


Subject(s)
Flow Cytometry/methods , Psoriasis/immunology , Psoriasis/pathology , Aminoquinolines/adverse effects , Animals , Cytokines/adverse effects , Dermis/pathology , Disease Models, Animal , Epidermal Cells/pathology , Epidermis/pathology , Imiquimod/adverse effects , Inflammation/pathology , Leukemic Infiltration/immunology , Mice , Psoriasis/chemically induced , Skin/immunology , Skin/pathology
13.
Int J Mol Sci ; 21(23)2020 Dec 05.
Article in English | MEDLINE | ID: mdl-33291515

ABSTRACT

Basal cell carcinoma (BCC) originate from Hedgehog/Patched signaling-activated epidermal stem cells. However, the chemically induced tumorigenesis of mice with a CD4Cre-mediated biallelic loss of the Hedgehog signaling repressor Patched also induces BCC formation. Here, we identified the cellular origin of CD4Cre-targeted BCC progenitors as rare Keratin 5+ epidermal cells and show that wildtype Patched offspring of these cells spread over the hair follicle/skin complex with increasing mouse age. Intriguingly, Patched mutant counterparts are undetectable in age-matched untreated skin but are getting traceable upon applying the chemical tumorigenesis protocol. Together, our data show that biallelic Patched depletion in rare Keratin 5+ epidermal cells is not sufficient to drive BCC development, because the spread of these cells is physiologically suppressed. However, bypassing the repression of Patched mutant cells, e.g., by exogenous stimuli, leads to an accumulation of BCC precursor cells and, finally, to tumor development.


Subject(s)
Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Cell Transformation, Neoplastic/genetics , Mutation , Patched-1 Receptor/genetics , Age Factors , Animals , Carcinoma, Basal Cell/metabolism , Disease Susceptibility , Epidermal Cells/metabolism , Epidermal Cells/pathology , Fluorescent Antibody Technique , Gene Knock-In Techniques , Genes, Reporter , Hair Follicle/metabolism , Hair Follicle/pathology , Humans , Immunohistochemistry , Immunophenotyping , Mice , Mice, Transgenic , Patched-1 Receptor/metabolism , Skin/metabolism , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Stem Cells/metabolism , Stem Cells/pathology
14.
Exp Mol Med ; 52(9): 1537-1549, 2020 09.
Article in English | MEDLINE | ID: mdl-32873845

ABSTRACT

Psoriasis is one of the most common immune-mediated chronic inflammatory skin diseases. However, little is known about the molecular mechanism underlying the immunological circuits that maintain innate and adaptive immune responses in established psoriasis. In this study, we found that the Pellino1 (Peli1) ubiquitin E3 ligase is activated by innate pattern-recognition receptors (PRRs), such as Toll-like receptors (TLRs), and is highly upregulated in human psoriatic skin lesions and murine psoriasis-like models. Increased Peli1 expression is strongly correlated with the immunopathogenesis of psoriasis by activating hyperproliferation of keratinocytes in the S and G2/M phases of the cell cycle and promoting chronic skin inflammation. Furthermore, Peli1-induced psoriasis-like lesions showed significant changes in the expression levels of several T helper 17 (Th17)-related cytokines, such as IL-17a, IL-21, IL-22, IL-23, and IL-24, indicating that overexpression of Peli1 resulted in the sequential engagement of the Th17 cell response. However, the overexpression of Peli1 in T cells was insufficient to trigger psoriasis, while T cells were indispensable for disease manifestation. In summary, our findings demonstrate that Peli1 is a critical cell cycle activator of innate immunity, which subsequently links Th17 cell immune responses to the psoriatic microenvironment.


Subject(s)
Dermatitis/etiology , Dermatitis/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Nuclear Proteins/genetics , Th17 Cells/immunology , Th17 Cells/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Biomarkers , Cell Cycle Checkpoints , Chronic Disease , Dermatitis/pathology , Disease Models, Animal , Epidermal Cells/metabolism , Epidermal Cells/pathology , Gene Expression , Immunophenotyping , Mice , Models, Biological , Nuclear Proteins/metabolism , Phenotype , Ubiquitin-Protein Ligases/metabolism
15.
Toxicol In Vitro ; 69: 104962, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32781017

ABSTRACT

Skin irritation tests using reconstructed human epidermis (RhE) employ viability as an endpoint, but color interference or borderline results are often problematic. We examined whether the cytology of cells from treated RhE could determine skin irritancy. Six chemicals (three irritants; DnP, 1-B, PH, three non-irritants; DP, APA, HS) were evaluated in a RhE, Keraskin™. DP, HS, and PH were clearly classified with viability, but DnP, 1-B, and APA were often falsely determined, due to borderline values falling near the cutoff, 50%. In histology, the tissues treated with DnP, 1-B, and PH showed erosion of the stratum corneum, vacuolization, and necrosis in the basal layer. DP- and HS-treated tissues showed relatively normal morphology but APA induced necrosis similar to irritants. Cytology revealed that DnP, 1-B or PH depleted cells and induced irregular and abnormal cell shapes. In contrast, relatively regular and normal shapes and clear distinction between the nucleus and cytoplasm was observed for DP, APA and HS. To further confirm it, additional 10 substances, including false positives from OECD TG 439, were tested. Overall (16 substances in total), cytology: total area predicted the skin irritancy of test chemicals with the highest accuracy (87.5%) followed by cytology: cell count (81.3%), histology (75%) and viability (68.8%), confirming the utility of cytology as an alternative endpoint in the skin irritation test using RhE.


Subject(s)
Epidermal Cells/drug effects , Epidermis/drug effects , Irritants/toxicity , Skin Irritancy Tests/methods , Animal Testing Alternatives , Disulfides/toxicity , Epidermal Cells/pathology , Epidermis/pathology , Glycolates/toxicity , Humans , Hydrocarbons, Halogenated/toxicity , Hydroxides/toxicity , Phthalic Acids/toxicity , Potassium Compounds/toxicity , Salicylates/toxicity
16.
Nat Cell Biol ; 22(7): 779-790, 2020 07.
Article in English | MEDLINE | ID: mdl-32451440

ABSTRACT

Tissue stem cells are the cell of origin for many malignancies. Metabolites regulate the balance between self-renewal and differentiation, but whether endogenous metabolic pathways or nutrient availability predispose stem cells towards transformation remains unknown. Here, we address this question in epidermal stem cells (EpdSCs), which are a cell of origin for squamous cell carcinoma. We find that oncogenic EpdSCs are serine auxotrophs whose growth and self-renewal require abundant exogenous serine. When extracellular serine is limited, EpdSCs activate de novo serine synthesis, which in turn stimulates α-ketoglutarate-dependent dioxygenases that remove the repressive histone modification H3K27me3 and activate differentiation programmes. Accordingly, serine starvation or enforced α-ketoglutarate production antagonizes squamous cell carcinoma growth. Conversely, blocking serine synthesis or repressing α-ketoglutarate-driven demethylation facilitates malignant progression. Together, these findings reveal that extracellular serine is a critical determinant of EpdSC fate and provide insight into how nutrient availability is integrated with stem cell fate decisions during tumour initiation.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic/pathology , Epidermal Cells/pathology , Ketoglutaric Acids/metabolism , Serine/metabolism , Stem Cells/pathology , Animals , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Epidermal Cells/metabolism , Female , Humans , Male , Mice , Stem Cells/metabolism
17.
Cell ; 181(3): 604-620.e22, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259486

ABSTRACT

During embryonic and postnatal development, organs and tissues grow steadily to achieve their final size at the end of puberty. However, little is known about the cellular dynamics that mediate postnatal growth. By combining in vivo clonal lineage tracing, proliferation kinetics, single-cell transcriptomics, and in vitro micro-pattern experiments, we resolved the cellular dynamics taking place during postnatal skin epidermis expansion. Our data revealed that harmonious growth is engineered by a single population of developmental progenitors presenting a fixed fate imbalance of self-renewing divisions with an ever-decreasing proliferation rate. Single-cell RNA sequencing revealed that epidermal developmental progenitors form a more uniform population compared with adult stem and progenitor cells. Finally, we found that the spatial pattern of cell division orientation is dictated locally by the underlying collagen fiber orientation. Our results uncover a simple design principle of organ growth where progenitors and differentiated cells expand in harmony with their surrounding tissues.


Subject(s)
Epidermal Cells/metabolism , Epidermis/growth & development , Skin/growth & development , Animals , Animals, Outbred Strains , Cell Differentiation/physiology , Cell Division/physiology , Cell Lineage/genetics , Cell Proliferation/physiology , Cells, Cultured , Epidermal Cells/pathology , Epidermis/metabolism , Female , Male , Mice , Mice, Transgenic , Stem Cells/cytology
18.
Biochim Biophys Acta Mol Cell Res ; 1867(8): 118722, 2020 08.
Article in English | MEDLINE | ID: mdl-32302667

ABSTRACT

Dermal fibroblasts seem critical for epidermal maturation and differentiation and recent work demonstrated that diseased fibroblasts may drive pathophysiological processes. Nevertheless, still very little is known about the actual crosstalk between epidermal keratinocytes and dermal fibroblasts and the impact of dermal fibroblasts on epidermal maturation and differentiation. Aiming for a more fundamental understanding of the impact of the cellular crosstalk between keratinocytes and fibroblasts on the skin homeostasis, we generated full-thickness skin equivalents with and without fibroblasts and subsequently analysed them for the expression of skin differentiation markers, their barrier function, skin lipid content and epidermal cell signalling. Skin equivalents without fibroblasts consistently showed an impaired differentiation and dysregulated expression of skin barrier and tight junction proteins, increased skin permeability, and a decreased skin lipid/protein ratio. Most interestingly, impaired Ras/Raf/ERK/MEK signalling was evident in skin equivalents without fibroblasts. Our data clearly indicate that the epidermal-dermal crosstalk between keratinocytes and fibroblasts is critical for adequate skin differentiation and that fibroblasts orchestrate epidermal differentiation processes.


Subject(s)
Epidermal Cells/metabolism , Fibroblasts/metabolism , Homeostasis/physiology , Keratinocytes/metabolism , Skin/metabolism , Cell Differentiation , Epidermal Cells/pathology , Epidermis/metabolism , Homeostasis/genetics , Humans , Keratinocytes/pathology , Permeability , Skin/pathology , Skin Absorption
19.
Chem Biol Interact ; 321: 109025, 2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32135139

ABSTRACT

Epigenetic regulation is one of the driving forces in the process of carcinogenesis. Corosolic acid (CA); triterpenoid abundantly found in Lagerstroemia speciosa L. is known to modulate various cellular process including cellular oxidative stress and signaling kinases in various diseases, including skin cancer. Genetic mutations in early stages of skin cancer are well-documented, the epigenetic alterations remain elusive. In the present study, we identified the transcriptomic gene expression changes with RNAseq and genome-wide DNA CpG methylation changes with DNA methylseq to profile the early stage transcriptomic and epigenomic changes using tumor promoter TPA-mediated mouse epidermal epithelial JB6 P+ cells. JB6 P+ cells were treated with TPA and Corosolic acid by 7.5uM optimized by MTS assay. Differentiated expressed genes (DEGs) and Differentially methylated genes (DMRs) were analyzed by R software. Ingenuity Pathway Analysis (IPA) was employed to understand the differential regulation of specific pathways. Novel TPA induced differentially overexpressed genes like tumor promoter Prl2c2, small prolin rich protein (Sprr2h) was reported which was downregulated by corosolic acid treatment. Several cancer related pathways were identified by Ingenuity Pathways Analysis (IPA) including p53, Erk, TGF beta signaling pathways. Moreover, differentially methylated regions (DMRs) in genes like Dusp22 (Dual specificity protein phosphatase 22), Rassf (tumor suppressor gene family, Ras association domain family) in JB6 P+ cells were uncovered which are altered by TPA and are reversed by CA treatment. Interestingly, genes like CDK1 (Cyclin-dependent kinases 1) and RASSF2 (Ras association domain family member 2) observed to be differentially methylated and expressed which was further modulated by corosolic acid treatment, validated by qPCR. Given study indicated gene expression changes to DNA CpG methylation epigenomic changes modulated various molecular pathways in TPA-induced JB6 cells and revealed that CA can potentially reverse these changes which deciphering novel molecular targets for future prevention of early stages of skin cancer studies in human.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , DNA Methylation/drug effects , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Triterpenes/pharmacology , Animals , Carcinogens/toxicity , Cell Line , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , CpG Islands/drug effects , Epidermal Cells/pathology , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Mice , Phytochemicals/pharmacology , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Tetradecanoylphorbol Acetate/toxicity , Transcriptome/drug effects
20.
J Invest Dermatol ; 140(7): 1364-1370.e1, 2020 07.
Article in English | MEDLINE | ID: mdl-31972247

ABSTRACT

Streptococcus pyogenes tonsillar infection is well known to trigger and exacerbate psoriasis lesions in both guttate and plaque forms of the disease. Although mucosal and cutaneous tissues are closely involved in psoriasis pathology, the interaction between their specific immune responses has not been deeply explored. This work aims to address and characterize the presence of humoral responses against S. pyogenes in patients with psoriasis and its putative association with cytokine responses detected in vitro in our psoriasis ex vivo model, based on the coculture of cutaneous lymphocyte-associated antigen+/- T cells with autologous epidermal cells. Patients with psoriasis presented increased IgA response to S. pyogenes when compared with control subjects. In patients with plaque psoriasis, despite being negative for anti-streptolysin O antibody titer, IgA plasma levels against S. pyogenes correlated with cutaneous lymphocyte-associated antigen+ T-cell-dependent IL-17F response in vitro. No association is observed for IgG levels in plaque psoriasis. Similar association is observed for IgA anti-S. pyogenes extract and IL-17A in patients with guttate psoriasis. We propose S. pyogenes-specific IgA as a potential new perspective for better understanding the role of S. pyogenes in psoriasis development.


Subject(s)
Immunoglobulin A/immunology , Interleukin-17/immunology , Psoriasis/microbiology , Streptococcal Infections/immunology , Streptococcus pyogenes , T-Lymphocytes/immunology , Adult , Cytokines/immunology , Epidermal Cells/pathology , Female , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Male , Middle Aged , Oligosaccharides/immunology , Palatine Tonsil/immunology , Psoriasis/blood , Psoriasis/immunology , Sialyl Lewis X Antigen/analogs & derivatives , Sialyl Lewis X Antigen/immunology , Skin/pathology , Streptococcal Infections/blood , Streptococcal Infections/microbiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL