Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nat Rev Cancer ; 23(7): 430-449, 2023 07.
Article in English | MEDLINE | ID: mdl-37286893

ABSTRACT

Human malignancies arise predominantly in tissues of epithelial origin, where the stepwise transformation from healthy epithelium to premalignant dysplasia to invasive neoplasia involves sequential dysregulation of biological networks that govern essential functions of epithelial homeostasis. Cutaneous squamous cell carcinoma (cSCC) is a prototype epithelial malignancy, often with a high tumour mutational burden. A plethora of risk genes, dominated by UV-induced sun damage, drive disease progression in conjunction with stromal interactions and local immunomodulation, enabling continuous tumour growth. Recent studies have identified subpopulations of SCC cells that specifically interact with the tumour microenvironment. These advances, along with increased knowledge of the impact of germline genetics and somatic mutations on cSCC development, have led to a greater appreciation of the complexity of skin cancer pathogenesis and have enabled progress in neoadjuvant immunotherapy, which has improved pathological complete response rates. Although measures for the prevention and therapeutic management of cSCC are associated with clinical benefit, the prognosis remains poor for advanced disease. Elucidating how the genetic mechanisms that drive cSCC interact with the tumour microenvironment is a current focus in efforts to understand, prevent and treat cSCC.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/therapy , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Prognosis , Disease Progression , Tumor Microenvironment/genetics
2.
J Invest Dermatol ; 143(11): 2177-2192.e13, 2023 11.
Article in English | MEDLINE | ID: mdl-37142187

ABSTRACT

Epidermal homeostasis is governed by a balance between keratinocyte proliferation and differentiation with contributions from cell-cell interactions, but conserved or divergent mechanisms governing this equilibrium across species and how an imbalance contributes to skin disease are largely undefined. To address these questions, human skin single-cell RNA sequencing and spatial transcriptomics data were integrated and compared with mouse skin data. Human skin cell-type annotation was improved using matched spatial transcriptomics data, highlighting the importance of spatial context in cell-type identity, and spatial transcriptomics refined cellular communication inference. In cross-species analyses, we identified a human spinous keratinocyte subpopulation that exhibited proliferative capacity and a heavy metal processing signature, which was absent in mouse and may account for species differences in epidermal thickness. This human subpopulation was expanded in psoriasis and zinc-deficiency dermatitis, attesting to disease relevance and suggesting a paradigm of subpopulation dysfunction as a hallmark of the disease. To assess additional potential subpopulation drivers of skin diseases, we performed cell-of-origin enrichment analysis within genodermatoses, nominating pathogenic cell subpopulations and their communication pathways, which highlighted multiple potential therapeutic targets. This integrated dataset is encompassed in a publicly available web resource to aid mechanistic and translational studies of normal and diseased skin.


Subject(s)
Skin Diseases , Transcriptome , Humans , Animals , Mice , Skin , Keratinocytes/metabolism , Epidermis/pathology , Skin Diseases/pathology , Cell Communication
5.
Small GTPases ; 10(3): 163-168, 2019 05.
Article in English | MEDLINE | ID: mdl-28055293

ABSTRACT

The small GTPase Ras-related C3 botulinum toxin substrate 1 (RAC1) plays a central role in skin homeostasis, including barrier function, wound healing and inflammatory responses. Psoriasis is a common skin disease characterized by deregulation of these functions, and affected skin exhibit keratinocyte hyperproliferation, inflammation and immune cell infiltration. Although psoriasis is often triggered by environmental stimulus, there is a strong genetic association with genes expressed in both immune cells and keratinocytes, of which several are linked to Rac1 signaling. Rac1 is highly active in human psoriatic lesional skin and keratinocytes, and keratinocyte-specific overexpression of an activated mutant of Rac1, Rac1V12, in a transgenic mouse model closely mimics the presentation of human psoriasis. Both Rac1 activation in keratinocytes and immune derived stimulus are required to drive psoriasiform signaling in transgenic mouse and human xenograft models of psoriasis. Therefore, understanding how increased Rac1 activation in psoriatic epidermis is regulated is central to understanding how the abnormal crosstalk between keratinocytes and immune cells is maintained.


Subject(s)
Cell Proliferation , Epidermis/embryology , Keratinocytes/enzymology , Psoriasis/enzymology , rac1 GTP-Binding Protein/metabolism , Animals , Disease Models, Animal , Enzyme Activation/genetics , Epidermis/pathology , Humans , Keratinocytes/pathology , Mice , Mice, Transgenic , Mutation , Psoriasis/genetics , Psoriasis/pathology , rac1 GTP-Binding Protein/genetics
6.
J Invest Dermatol ; 138(4): 864-871, 2018 04.
Article in English | MEDLINE | ID: mdl-29129599

ABSTRACT

The chronic and highly prevalent skin disorder psoriasis vulgaris is characterized by a hyperproliferative epidermis and aberrant immune activity. Many studies have highlighted the role of differentiated T lymphocytes in psoriasis progression. Several biologics are currently available that target proinflammatory cytokines produced by T lymphocytes, but the need for improved therapies persists. The small molecule PRN694 covalently binds ITK and RLK, two Tec kinases activated downstream of T-lymphocyte activation, both of which are up-regulated in psoriatic skin. These Tec kinases are involved in signaling cascades mediating T-lymphocyte proliferation, differentiation, and migration and proinflammatory cytokine production. In vitro analysis showed that PRN694 effectively inhibited IL-17A production from murine T helper type 17-differentiated T lymphocytes. Additionally, PRN694 effectively reduced the psoriasis-like phenotype severity and reduced epidermal proliferation and thickness in both the Rac1V12 and imiquimod mouse models of psoriasis. PRN694 also inhibited CD3+ T-cell and γδ T-cell infiltration into skin regions. Inhibition of ITK and RLK attenuated psoriasis-associated signaling pathways, indicating that PRN694 is an effective psoriasis therapeutic.


Subject(s)
Benzimidazoles/pharmacology , Dermis/pathology , Gene Expression Regulation , Immunity, Cellular , Protein-Tyrosine Kinases/genetics , Psoriasis/genetics , Animals , Cells, Cultured , Dermis/metabolism , Disease Models, Animal , Humans , Mice , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/biosynthesis , Psoriasis/drug therapy , Psoriasis/immunology , RNA, Messenger/genetics , T-Lymphocytes/immunology
7.
Pediatr Dermatol ; 34(3): e140-e141, 2017 May.
Article in English | MEDLINE | ID: mdl-28295514

ABSTRACT

Genetic variants in filaggrin (FLG) involving truncating mutations or intragenic copy number variation are strongly associated with the risk of developing atopic dermatitis (AD) in European and Asian populations. Few loss-of-function mutations have been identified in Africans, although an association between FLG copy number variation and AD severity in a small African American cohort has been proposed. We studied the association between FLG copy number and AD in 132 Ethiopians and found no association between AD severity and FLG copy number, suggesting that other, still unidentified genetic factors are of more importance in predisposing Ethiopians to AD.


Subject(s)
DNA Copy Number Variations/genetics , Dermatitis, Atopic/genetics , Genetic Predisposition to Disease/ethnology , Intermediate Filament Proteins/genetics , Adolescent , Age Factors , Case-Control Studies , Child , Child, Preschool , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/ethnology , Ethiopia/epidemiology , Female , Filaggrin Proteins , Humans , Male , Polymerase Chain Reaction/methods , Prevalence , Risk Assessment , Severity of Illness Index , Sex Factors
8.
J Clin Invest ; 126(7): 2661-77, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27294528

ABSTRACT

Interactions between the epidermis and the immune system govern epidermal tissue homeostasis. These epidermis-immune interactions are altered in the inflammatory disease psoriasis; however, the pathways that underlie this aberrant immune response are not well understood. Here, we determined that Ras-related C3 botulinum toxin substrate 1 (RAC1) is a key mediator of epidermal dysfunction. RAC1 activation was consistently elevated in psoriatic epidermis and primary psoriatic human keratinocytes (PHKCs) exposed to psoriasis-related stimuli, but not in skin from patients with basal or squamous cell carcinoma. Expression of a constitutively active form of RAC1 (RACV12) in mice resulted in the development of lesions similar to those of human psoriasis that required the presence of an intact immune system. RAC1V12-expressing mice and human psoriatic skin showed similar RAC1-dependent signaling as well as transcriptional overlap of differentially expressed epidermal and immune pathways. Coculture of PHKCs with immunocytes resulted in the upregulation of RAC1-dependent proinflammatory cytokines, an effect that was reproduced by overexpressing RAC1 in normal human keratinocytes. In keratinocytes, modulating RAC1 activity altered differentiation, proliferation, and inflammatory pathways, including STAT3, NFκB, and zinc finger protein 750 (ZNF750). Finally, RAC1 inhibition in xenografts composed of human PHKCs and immunocytes abolished psoriasiform hyperplasia and inflammation in vivo. These studies implicate RAC1 as a potential therapeutic target for psoriasis and as a key orchestrator of pathologic epidermis-immune interactions.


Subject(s)
Epidermis/metabolism , Keratinocytes/cytology , Psoriasis/immunology , Psoriasis/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation , Coculture Techniques , Cytokines/metabolism , Humans , Immune System , Inflammation , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , Phenotype , Skin/pathology
9.
Cell Transplant ; 25(7): 1371-80, 2016.
Article in English | MEDLINE | ID: mdl-26132738

ABSTRACT

Compelling evidence suggests that transplantation of neural stem cells (NSCs) from multiple sources ameliorates motor deficits after stroke. However, it is currently unknown to what extent the electrophysiological activity of grafted NSC progeny participates in the improvement of motor deficits and whether excitatory phenotypes of the grafted cells are beneficial or deleterious to sensorimotor performances. To address this question, we used optogenetic tools to drive the excitatory outputs of the grafted NSCs and assess the impact on local circuitry and sensorimotor performance. We genetically engineered NSCs to express the Channelrhodopsin-2 (ChR2), a light-gated cation channel that evokes neuronal depolarization and initiation of action potentials with precise temporal control to light stimulation. To test the function of these cells in a stroke model, rats were subjected to an ischemic stroke and grafted with ChR2-NSCs. The grafted NSCs identified with a human-specific nuclear marker survived in the peri-infarct tissue and coexpressed the ChR2 transgene with the neuronal markers TuJ1 and NeuN. Gene expression analysis in stimulated versus vehicle-treated animals showed a differential upregulation of transcripts involved in neurotransmission, neuronal differentiation, regeneration, axonal guidance, and synaptic plasticity. Interestingly, genes involved in the inflammatory response were significantly downregulated. Behavioral analysis demonstrated that chronic optogenetic stimulation of the ChR2-NSCs enhanced forelimb use on the stroke-affected side and motor activity in an open field test. Together these data suggest that excitatory stimulation of grafted NSCs elicits beneficial effects in experimental stroke model through cell replacement and non-cell replacement, anti-inflammatory/neurotrophic effects.


Subject(s)
Down-Regulation , Neural Stem Cells/transplantation , Optogenetics/methods , Stroke/therapy , Synaptic Transmission , Animals , Cell Separation , Disease Models, Animal , Gene Expression Profiling , Human Embryonic Stem Cells/cytology , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/therapy , Male , Neostriatum/metabolism , Neural Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Rats, Sprague-Dawley , Rhodopsin/genetics , Stroke/complications , Stroke/genetics , Transduction, Genetic , Transgenes
11.
PLoS Genet ; 11(3): e1005076, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25757221

ABSTRACT

Epidemiological studies suggest that allergy risk is preferentially transmitted through mothers. This can be due to genomic imprinting, where the phenotype effect of an allele depends on its parental origin, or due to maternal effects reflecting the maternal genome's influence on the child during prenatal development. Loss-of-function mutations in the filaggrin gene (FLG) cause skin barrier deficiency and strongly predispose to atopic dermatitis (AD). We investigated the 4 most prevalent European FLG mutations (c.2282del4, p.R501X, p.R2447X, and p.S3247X) in two samples including 759 and 450 AD families. We used the multinomial and maximum-likelihood approach implemented in the PREMIM/EMIM tool to model parent-of-origin effects. Beyond the known role of FLG inheritance in AD (R1meta-analysis = 2.4, P = 1.0 x 10-36), we observed a strong maternal FLG genotype effect that was consistent in both independent family sets and for all 4 mutations analysed. Overall, children of FLG-carrier mothers had a 1.5-fold increased AD risk (S1 = 1.50, Pmeta-analysis = 8.4 x 10-8). Our data point to two independent and additive effects of FLG mutations: i) carrying a mutation and ii) having a mutation carrier mother. The maternal genotype effect was independent of mutation inheritance and can be seen as a non-genetic transmission of a genetic effect. The FLG maternal effect was observed only when mothers had allergic sensitization (elevated allergen-specific IgE antibody plasma levels), suggesting that FLG mutation-induced systemic immune responses in the mother may influence AD risk in the child. Notably, the maternal effect reported here was stronger than most common genetic risk factors for AD recently identified through genome-wide association studies (GWAS). Our study highlights the power of family-based studies in the identification of new etiological mechanisms and reveals, for the first time, a direct influence of the maternal genotype on the offspring's susceptibility to a common human disease.


Subject(s)
Dermatitis, Atopic/genetics , Intermediate Filament Proteins/genetics , Female , Filaggrin Proteins , Genome-Wide Association Study , Genomic Imprinting , Humans , Male , Meta-Analysis as Topic , Mutation
12.
Hum Mol Genet ; 22(23): 4841-56, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23886662

ABSTRACT

Atopic dermatitis (AD) is the most common dermatological disease of childhood. Many children with AD have asthma and AD shares regions of genetic linkage with psoriasis, another chronic inflammatory skin disease. We present here a genome-wide association study (GWAS) of childhood-onset AD in 1563 European cases with known asthma status and 4054 European controls. Using Illumina genotyping followed by imputation, we generated 268 034 consensus genotypes and in excess of 2 million single nucleotide polymorphisms (SNPs) for analysis. Association signals were assessed for replication in a second panel of 2286 European cases and 3160 European controls. Four loci achieved genome-wide significance for AD and replicated consistently across all cohorts. These included the epidermal differentiation complex (EDC) on chromosome 1, the genomic region proximal to LRRC32 on chromosome 11, the RAD50/IL13 locus on chromosome 5 and the major histocompatibility complex (MHC) on chromosome 6; reflecting action of classical HLA alleles. We observed variation in the contribution towards co-morbid asthma for these regions of association. We further explored the genetic relationship between AD, asthma and psoriasis by examining previously identified susceptibility SNPs for these diseases. We found considerable overlap between AD and psoriasis together with variable coincidence between allergic rhinitis (AR) and asthma. Our results indicate that the pathogenesis of AD incorporates immune and epidermal barrier defects with combinations of specific and overlapping effects at individual loci.


Subject(s)
Asthma/genetics , Dermatitis, Atopic/genetics , Genome-Wide Association Study/methods , Psoriasis/genetics , Adolescent , Adult , Case-Control Studies , Child , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 5 , Chromosomes, Human, Pair 6 , Genetic Linkage , Genetic Loci , Genetic Predisposition to Disease , Genotype , Humans , Male , Polymorphism, Single Nucleotide , White People/genetics , Young Adult
13.
PLoS One ; 7(11): e49694, 2012.
Article in English | MEDLINE | ID: mdl-23189155

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is a common chronic inflammatory skin disorder where epidermal barrier dysfunction is a major factor in the pathogenesis. The identification of AD susceptibility genes related to barrier dysfunction is therefore of importance. The epidermal transglutaminases (TGM1, TGM3 and TGM5) encodes essential cross-linking enzymes in the epidermis. OBJECTIVE: To determine whether genetic variability in the epidermal transglutaminases contributes to AD susceptibility. METHODS: Forty-seven single nucleotide polymorphisms (SNPs) in the TGM1, TGM3 and TGM5 gene region were tested for genetic association with AD, independently and in relation to FLG genotype, using a pedigree disequilibrium test (PDT) in a Swedish material consisting of 1753 individuals from 539 families. In addition, a German case-control material, consisting of 533 AD cases and 1996 controls, was used for in silico analysis of the epidermal TGM regions. Gene expression of the TGM1, TGM3 and TGM5 gene was investigated by relative quantification with Real Time PCR (qRT-PCR). Immunohistochemical (IHC) analysis was performed to detect TG1, TG3 and TG5 protein expression in the skin of patients and healthy controls. RESULTS: PDT analysis identified a significant association between the TGM1 SNP rs941505 and AD with allergen-specific IgE in the Swedish AD family material. However, the association was not replicated in the German case-control material. No significant association was detected for analyzed SNPs in relation to FLG genotype. TG1, TG3 and TG5 protein expression was detected in AD skin and a significantly increased TGM3 mRNA expression was observed in lesional skin by qRT-PCR. CONCLUSION: Although TGM1 and TGM3 may be differentially expressed in AD skin, the results from the genetic analysis suggest that genetic variation in the epidermal transglutaminases is not an important factor in AD susceptibility.


Subject(s)
Dermatitis, Atopic/genetics , Epidermis/metabolism , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Transglutaminases/genetics , Case-Control Studies , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Epidermis/pathology , Filaggrin Proteins , Germany , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sweden , Transglutaminases/metabolism , White People/genetics
14.
Arch Dermatol Res ; 304(10): 773-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22552355

ABSTRACT

Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases in industrialized countries. To identify candidate genes involved in the pathogenesis of AD, we previously undertook a genome-wide approach using DNA microarrays. A transcript encoding the epidermal growth factor receptor (EGFR) was found to be among the down-regulated transcripts in AD skin. Here, we further investigated the expression pattern of two EGFR family members (EGFR and ErbB2) in AD skin on a protein level. Immunohistochemical (IHC) analysis of EGFR and ErbB2 showed decreased expression of EGFR and ErbB2 proteins in AD lesional skin as compared to skin from healthy individuals. Interestingly, we found that EGFR and ErbB2 were reciprocally expressed in an in vitro model of keratinocyte proliferation and differentiation, paralleling the expression patterns found in epidermis of healthy skin. The highest levels of EGFR transcripts were found in proliferating cells, while ErbB2 was found in differentiated cells. We show that blocking EGFR activity combined with co-stimulation of the Th2-cytokine IL4 in keratinocytes leads to induction of the inflammatory chemokine CCL26/eotaxin-3 in vitro. Accordingly, increased CCL26 transcriptional levels were observed in AD lesional skin. Taken together, suppression of EGFR may contribute to the pathogenesis of AD via the regulation of inflammatory chemokines.


Subject(s)
Dermatitis, Atopic/metabolism , ErbB Receptors/biosynthesis , Keratinocytes/metabolism , Receptor, ErbB-2/biosynthesis , Skin/metabolism , Adolescent , Adult , Cell Differentiation/drug effects , Cell Line , Chemokine CCL26 , Chemokines, CC/metabolism , Dermatitis, Atopic/genetics , ErbB Receptors/genetics , Female , Humans , Inflammation Mediators/metabolism , Interleukin-4/immunology , Keratinocytes/drug effects , Male , Middle Aged , Quinazolines/pharmacology , Receptor, ErbB-2/genetics , Th2 Cells/immunology , Transcription, Genetic , Young Adult
15.
PLoS One ; 6(12): e28254, 2011.
Article in English | MEDLINE | ID: mdl-22164253

ABSTRACT

BACKGROUND: Several common genetic and environmental disease mechanisms are important for the pathophysiology behind atopic dermatitis (AD). Filaggrin (FLG) loss-of-function is of great significance for barrier impairment in AD and ichthyosis vulgaris (IV), which is commonly associated with AD. The molecular background is, however, complex and various clusters of genes are altered, including inflammatory and epidermal-differentiation genes. OBJECTIVE: The objective was to study whether the functional and molecular alterations in AD and IV skin depend directly on FLG loss-of-function, and whether FLG genotype determines the type of downstream molecular pathway affected. METHODS AND FINDINGS: Patients with AD/IV (n = 43) and controls (n = 15) were recruited from two Swedish outpatient clinics and a Swedish AD family material with known FLG genotype. They were clinically examined and their medical history recorded using a standardized questionnaire. Blood samples and punch biopsies were taken and trans-epidermal water loss (TEWL) and skin pH was assessed with standard techniques. In addition to FLG genotyping, the STS gene was analyzed to exclude X-linked recessive ichthyosis (XLI). Microarrays and quantitative real-time PCR were used to compare differences in gene expression depending on FLG genotype. Several different signalling pathways were altered depending on FLG genotype in patients suffering from AD or AD/IV. Disease severity, TEWL and pH follow FLG deficiency in the skin; and the number of altered genes and pathways are correlated to FLG mRNA expression. CONCLUSIONS: We emphasize further the role of FLG in skin-barrier integrity and the complex compensatory activation of signalling pathways. This involves inflammation, epidermal differentiation, lipid metabolism, cell signalling and adhesion in response to FLG-dependent skin-barrier dysfunction.


Subject(s)
Dermatitis, Atopic/genetics , Genotype , Intermediate Filament Proteins/genetics , Skin/pathology , Adult , Aged , Cell Adhesion , Cell Differentiation , Dermatitis, Atopic/pathology , Female , Filaggrin Proteins , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Ichthyosis Vulgaris/genetics , Ichthyosis Vulgaris/pathology , Inflammation , Lipid Metabolism , Male , Middle Aged , Multigene Family , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Signal Transduction , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL
...