Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
J Invest Dermatol ; 140(9): 1723-1732, 2020 09.
Article in English | MEDLINE | ID: mdl-32061658

ABSTRACT

The ability of cancer cells to invade and disseminate can be affected by components of the surrounding microenvironment. To identify dermal components that regulate the growth of epidermal carcinomas, we studied the genetic disease called xeroderma pigmentosum that bears mutations in genes involved in the nucleotide excision repair of DNA. Patients with xeroderma pigmentosum are more prone to develop cutaneous tumors than the general population and their dermal fibroblasts display the features of dermal cancer-associated fibroblasts, which promote the invasion of keratinocytes. Here, we report that 3-dimensional dermal cultures of fibroblasts from healthy donors but not from patients with xeroderma pigmentosum complementation group C express CLEC2A, which is the ligand of the activating NK cell receptor NKp65. A similar loss of CLEC2A was observed in sporadic dermal cancer-associated fibroblasts and upon the culture of fibroblasts with cutaneous squamous cell carcinoma-conditioned medium. Using an innovative 3-dimensional organotypic skin culture model that contain NK cells in addition to fibroblasts and squamous cell carcinoma cells, we unveiled a key role of CLEC2A that orchestrates a crosstalk between fibroblasts and NK cells, thereby leading to the control of squamous cell carcinoma invasion. These findings indicate that CLEC2A-expressing dermal fibroblasts play a major role in immune surveillance of the skin.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/immunology , Lectins, C-Type/deficiency , Skin Neoplasms/immunology , Xeroderma Pigmentosum/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Cancer-Associated Fibroblasts/immunology , Carcinoma, Squamous Cell/pathology , Cell Communication/immunology , Cells, Cultured , Child , Child, Preschool , Coculture Techniques , DNA-Binding Proteins/genetics , Female , Gene Expression Profiling , Humans , Immunologic Surveillance , Infant , Infant, Newborn , Killer Cells, Natural/immunology , Male , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Primary Cell Culture , Receptors, NK Cell Lectin-Like/metabolism , Skin/immunology , Skin/pathology , Skin Neoplasms/pathology , Tumor Microenvironment/immunology , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/immunology , Young Adult
2.
Curr Protoc Immunol ; 118: 7.42.1-7.42.12, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28762488

ABSTRACT

This unit describes the monitoring and quantification of cellular cytotoxicity using a non-radioactive and real-time cytotoxic assay. The extent of target-cell lysis is monitored over time by imaging and quantifying live fluorescent target cells using a cell-imaging multimode reader. This assay is performed in a 96 well plate in optimized culture conditions at 37°C in the presence of 5% CO2 . The basic protocol describes natural killer cell-mediated cytotoxic assay that can be adapted to include antibodies blocking inhibitory NK receptors or triggering antibody-dependent cell-mediated cytotoxicity (ADCC). The assay is also suitable for antigen specific T-cell cytotoxic assays. Until now, the standard chromium 51 (51 Cr)-release assay has remained the sole sensitive assay but its major drawbacks include cost and hazard of handling radioactivity. The real-time cytotoxic assay is therefore an effective alternative providing a robust and sensitive assay that accurately monitors lysis of target cells over time. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Cytotoxicity Tests, Immunologic , Killer Cells, Natural/immunology , T-Lymphocytes, Cytotoxic/immunology , Biological Assay , Cells, Cultured , Chromium Radioisotopes , Cytotoxicity, Immunologic , Humans
3.
PLoS One ; 10(12): e0145369, 2015.
Article in English | MEDLINE | ID: mdl-26694869

ABSTRACT

Basal cell carcinoma (BCC) is the commonest tumor in human. About 70% sporadic BCCs bear somatic mutations in the PATCHED1 tumor suppressor gene which encodes the receptor for the Sonic Hedgehog morphogen (SHH). PATCHED1 germinal mutations are associated with the dominant Nevoid Basal Cell Carcinoma Syndrome (NBCCS), a major hallmark of which is a high susceptibility to BCCs. Although the vast majority of sporadic BCCs arises exclusively in sun exposed skin areas, 40 to 50% BCCs from NBCCS patients develop in non photo-exposed skin. Since overwhelming evidences indicate that microenvironment may both be modified by- and influence the- epithelial tumor, we hypothesized that NBCCS fibroblasts could contribute to BCCs in NBCCS patients, notably those developing in non photo-exposed skin areas. The functional impact of NBCCS fibroblasts was then assessed in organotypic skin cultures with control keratinocytes. Onset of epidermal differentiation was delayed in the presence of primary NBCCS fibroblasts. Unexpectedly, keratinocyte proliferation was severely reduced and showed high levels of nuclear P53 in both organotypic skin cultures and in fibroblast-led conditioning experiments. However, in spite of increased levels of senescence associated ß-galactosidase activity in keratinocytes cultured in the presence of medium conditioned by NBCCS fibroblasts, we failed to observe activation of P16 and P21 and then of bona fide features of senescence. Constitutive extinction of P53 in WT keratinocytes resulted in an invasive phenotype in the presence of NBCCS fibroblasts. Finally, we found that expression of SHH was limited to fibroblasts but was dependent on the presence of keratinocytes. Inhibition of SHH binding resulted in improved epidermal morphogenesis. Altogether, these data suggest that the repertoire of diffusible factors (including SHH) expressed by primary NBCCS fibroblasts generate a stress affecting keratinocytes behavior and epidermal homeostasis. Our findings suggest that defects in dermo/epidermal interactions could contribute to BCC susceptibility in NBCCS patients.


Subject(s)
Basal Cell Nevus Syndrome/pathology , Carcinoma, Basal Cell/pathology , Fibroblasts/cytology , Receptors, Cell Surface/genetics , Skin Neoplasms/pathology , Tumor Microenvironment , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/metabolism , Carcinoma, Basal Cell/etiology , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mutation , Organ Culture Techniques , Patched Receptors , Patched-1 Receptor , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL