Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35012988

ABSTRACT

Early diagnosis of oral squamous cell carcinoma (OSCC) remains an unmet clinical need. Therefore, elucidating the initial events of OSCC preceding tumor development could benefit OSCC prognosis. Here, we define the Langerhans cells (LCs) of the tongue and demonstrate that LCs protect the epithelium from carcinogen-induced OSCC by rapidly priming αßT cells capable of eliminating γH2AX+ epithelial cells, whereas γδT and natural killer cells are dispensable. The carcinogen, however, dysregulates the epithelial resident mononuclear phagocytes, reducing LC frequencies, while dendritic cells (DCs), macrophages, and plasmacytoid DCs (pDCs) populate the epithelium. Single-cell RNA-sequencing analysis indicates that these newly differentiated cells display an immunosuppressive phenotype accompanied by an expansion of T regulatory (Treg) cells. Accumulation of the Treg cells was regulated, in part, by pDCs and precedes the formation of visible tumors. This suggests LCs play an early protective role during OSCC, yet the capacity of the carcinogen to dysregulate the differentiation of mononuclear phagocytes facilitates oral carcinogenesis.


Subject(s)
Antineoplastic Agents/metabolism , Carcinogens/toxicity , Langerhans Cells/metabolism , 4-Nitroquinoline-1-oxide/toxicity , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/pathology , Epithelial Cells/metabolism , Epithelium/drug effects , Epithelium/pathology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Histones/metabolism , Humans , Immunity/drug effects , Langerhans Cells/drug effects , Phagocytes/drug effects , Phagocytes/metabolism , Phagocytes/pathology , Quinolones/toxicity , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tongue/pathology , Transcriptome/genetics
2.
Cell Host Microbe ; 29(2): 197-209.e5, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33412104

ABSTRACT

Postnatal host-microbiota interplay governs mucosal homeostasis and is considered to have life-long health consequences. The intestine monolayer epithelium is critically involved in such early-life processes; nevertheless, the role of the oral multilayer epithelium remains ill defined. We demonstrate that unlike the intestine, the neonate oral cavity is immensely colonized by the microbiota that decline to adult levels during weaning. Neutrophils are present in the oral epithelium prenatally, and exposure to the microbiota postnatally further recruits them to the preamble neonatal epithelium by γδT17 cells. These neutrophils virtually disappear during weaning as the epithelium seals. The neonate and adult epithelium display distinct turnover kinetics and transcriptomic signatures, with neonate epithelium reminiscent of the signature found in germ-free mice. Microbial reduction during weaning is mediated by the upregulation of saliva production and induction of salivary antimicrobial components by the microbiota. Collectively, unique postnatal interactions between the multilayer epithelium and microbiota shape oral homeostasis.


Subject(s)
Bacterial Load , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Neutrophils/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Saliva/microbiology , Animals , Animals, Newborn/growth & development , Animals, Newborn/microbiology , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouth Mucosa/cytology , Mouth Mucosa/growth & development , Th17 Cells/immunology
3.
Front Immunol ; 9: 1712, 2018.
Article in English | MEDLINE | ID: mdl-30158922

ABSTRACT

Peri-implantitis is a destructive inflammatory process affecting tissues surrounding dental implants and it is considered a new global health concern. Human studies have suggested that the frequencies of Langerhans cells (LCs), the main antigen-presenting cells (APCs) of the oral epithelium, are dysregulated around the implants. Since LCs play a role in regulating oral mucosal homeostasis, we studied the impact of dental titanium implants on LC differentiation using a novel murine model. We demonstrate that whereas the percentage of LC precursors (CD11c+MHCII+) increased in the peri-implant epithelium, the frequencies of LCs (CD11c+MHCII+EpCAM+langerin+) were significantly reduced. Instead, a population of partially developed LCs expressing CD11c+MHCII+EpCAM+ but not langerin evolved in the peri-implant mucosa, which was also accompanied by a considerable leukocyte infiltrate. In line with the increased levels of LC precursors, expression of CCL2 and CCL20, chemokines mediating their translocation to the epithelium, was elevated in the peri-implant epithelium. However, expression of TGF-ß1, the major cytokine driving final differentiation of LCs, was reduced in the epithelium. Further analysis revealed that while the expression of the TGF-ß1 canonical receptor activing-like kinase (ALK)5 was upregulated, expression of its non-canonical receptor ALK3 was decreased. Since titanium ions releasing from implants were proposed to alter APC function, we next analyzed the impact of such ions on TGF-ß1-induced LC differentiation cultures. Concurring with the in vivo studies, the presence of titanium ions resulted in the generation of partially developed LCs that express CD11c+MHCII+EpCAM+ but failed to upregulate langerin expression. Collectively, these findings suggest that titanium dental implants have the capacity to impair the development of oral LCs and might subsequently dysregulate immunity in the peri-implant mucosa.


Subject(s)
Cell Differentiation , Dental Implants , Langerhans Cells/cytology , Langerhans Cells/metabolism , Mouth Mucosa/cytology , Mouth Mucosa/metabolism , Titanium , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biomarkers , Cells, Cultured , Cytokines/metabolism , Dental Implants/adverse effects , Gingiva/cytology , Ions/adverse effects , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Leukocyte Count , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Mice , Mouth Mucosa/pathology , Peri-Implantitis/etiology , Peri-Implantitis/metabolism , Peri-Implantitis/pathology , Stem Cells/cytology , Stem Cells/metabolism , Titanium/adverse effects
4.
Proc Natl Acad Sci U S A ; 115(25): E5736-E5745, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29871951

ABSTRACT

AXL, a member of the TYRO3, AXL, and MERTK (TAM) receptor tyrosine kinase family, has been shown to play a role in the differentiation and activation of epidermal Langerhans cells (LCs). Here, we demonstrate that growth arrest-specific 6 (GAS6) protein, the predominant ligand of AXL, has no impact on LC differentiation and homeostasis. We thus examined the role of protein S (PROS1), the other TAM ligand acting primarily via TYRO3 and MERTK, in LC function. Genetic ablation of PROS1 in keratinocytes resulted in a typical postnatal differentiation of LCs; however, a significant reduction in LC frequencies was observed in adult mice due to increased apoptosis. This was attributed to altered expression of cytokines involved in LC development and tissue homeostasis within keratinocytes. PROS1 was then excised in LysM+ cells to target LCs at early embryonic developmental stages, as well as in adult monocytes that also give rise to LCs. Differentiation and homeostasis of LCs derived from embryonic precursors was not affected following Pros1 ablation. However, differentiation of LCs from bone marrow (BM) precursors in vitro was accelerated, as was their capability to reconstitute epidermal LCs in vivo. These reveal an inhibitory role for PROS1 on BM-derived LCs. Collectively, this study highlights a cell-specific regulation of LC differentiation and homeostasis by TAM signaling.


Subject(s)
Carrier Proteins/metabolism , Epidermis/metabolism , Langerhans Cells/metabolism , Protein S/metabolism , Animals , Bone Marrow/metabolism , Calcium-Binding Proteins , Cell Differentiation/physiology , Homeostasis/physiology , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , c-Mer Tyrosine Kinase/metabolism
5.
J Exp Med ; 215(2): 481-500, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29343501

ABSTRACT

Mucosal Langerhans cells (LCs) originate from pre-dendritic cells and monocytes. However, the mechanisms involved in their in situ development remain unclear. Here, we demonstrate that the differentiation of murine mucosal LCs is a two-step process. In the lamina propria, signaling via BMP7-ALK3 promotes translocation of LC precursors to the epithelium. Within the epithelium, TGF-ß1 finalizes LC differentiation, and ALK5 is crucial to this process. Moreover, the local microbiota has a major impact on the development of mucosal LCs, whereas LCs in turn maintain mucosal homeostasis and prevent tissue destruction. These results reveal the differential and sequential role of TGF-ß1 and BMP7 in LC differentiation and highlight the intimate interplay of LCs with the microbiota.


Subject(s)
Bone Morphogenetic Protein 7/immunology , Langerhans Cells/immunology , Microbiota/immunology , Transforming Growth Factor beta1/immunology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Humans , Immunity, Mucosal , Langerhans Cells/cytology , Langerhans Cells/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Male , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouth Mucosa/cytology , Mouth Mucosa/immunology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction/immunology , Stem Cells/cytology , Stem Cells/immunology , Transcriptome , Transforming Growth Factor beta1/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...