Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Biol Chem ; 291(19): 10148-61, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26945071

ABSTRACT

The lymphocyte-oriented kinase (LOK), also called serine threonine kinase 10 (STK10), is synthesized mainly in lymphocytes. It is involved in lymphocyte migration and polarization and can phosphorylate ezrin, radixin, and moesin (the ERM proteins). In a T lymphocyte cell line and in purified human lymphocytes, we found LOK to be cleaved by caspases during apoptosis. The first cleavage occurs at aspartic residue 332, located between the kinase domain and the coiled-coil regulation domain. This cleavage generates an N-terminal fragment, p50 N-LOK, containing the kinase domain and a C-terminal fragment, which is further cleaved during apoptosis. Although these cleavages preserve the entire kinase domain, p50 N-LOK displays no kinase activity. In apoptotic lymphocytes, caspase cleavages of LOK are concomitant with a decrease in ERM phosphorylation. When non-apoptotic lymphocytes from mice with homozygous and heterozygous LOK knockout were compared, the latter showed a higher level of ERM phosphorylation, but when apoptosis was induced, LOK(-/-) and LOK(+/-) lymphocytes showed the same low level, confirming in vivo that LOK-induced ERM phosphorylation is prevented during lymphocyte apoptosis. Our results demonstrate that cleavage of LOK during apoptosis abolishes its kinase activity, causing a decrease in ERM phosphorylation, crucial to the role of the ERM proteins in linking the plasma membrane to actin filaments.


Subject(s)
Apoptosis , Caspases/metabolism , Lymphocytes/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Actins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Membrane , Cells, Cultured , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Female , Humans , Immunoenzyme Techniques , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Molecular Sequence Data , Phosphorylation , Sequence Homology, Amino Acid
2.
J Infect Dis ; 210(3): 493-503, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24577508

ABSTRACT

Mucosal sites are continuously exposed to pathogenic microorganisms and are therefore equipped to control respiratory infections. Type 3 innate lymphoid cells (ILC3) are key players in antimicrobial defense in intestinal mucosa, through interleukin 17 and interleukin 22 (IL-22) production. The present study aimed at analyzing the distribution and function of ILC3 in the respiratory tract. We first observed that lung mucosa harbors a discrete population of ILC3 expressing CD127, CD90, CCR6, and the transcriptional factor RORγt. In addition, lung ILC3 were identified as a major source of IL-22 in response to interleukin 23 stimulation. During Streptococcus pneumoniae infection, ILC3 rapidly accumulated in the lung tissue to produce IL-22. In response to S. pneumoniae, dendritic cells and MyD88, an important adaptor of innate immunity, play critical functions in IL-22 production by ILC3. Finally, administration of the Toll-like receptor 5 agonist flagellin during S. pneumoniae challenge exacerbated IL-22 production by ILC3, a process that protects against lethal infection. In conclusion, boosting lung ILC3 might represent an interesting strategy to fight respiratory bacterial infections.


Subject(s)
Interleukins/metabolism , Lung/metabolism , Lymphocytes/classification , Lymphocytes/physiology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Animals , Female , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukins/genetics , Lymphocyte Activation , Mice , Mice, Knockout , Streptococcus pneumoniae , Interleukin-22
3.
Bio Protoc ; 13(7): e4612, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-37056241

ABSTRACT

Cellular senescence is a reprogrammed cell state triggered as an adaptative response to a variety of stresses, most often those affecting the genome integrity. Senescent cells accumulate in most tissues with age and contribute to the development of several pathologies. Studying molecular pathways involved in senescence induction and maintenance, or in senescence escape, can be hindered by the heterogeneity of senescent cell populations. Here, we describe a flow cytometry strategy for sorting senescent cells according to three senescence canonical markers whose thresholds can be independently adapted to be more or less stringent: (i) the senescence-associated-ß-galactosidase (SA-ß-Gal) activity, detected using 5-dodecanoylaminofluorescein Di-ß-D-galactopyranoside (C12FDG), a fluorigenic substrate of ß-galactosidase; (ii) cell size, proportional to the forward scatter value, since increased size is one of the major changes observed in senescent cells; and (iii) cell granularity, proportional to the side scatter value, which reflects the accumulation of aggregates, lysosomes, and altered mitochondria in senescent cells. We applied this protocol to the sorting of normal human fibroblasts at the replicative senescence plateau. We highlighted the challenge of sorting these senescent cells because of their large sizes, and established that it requires using sorters equipped with a nozzle of an unusually large diameter: at least 200 µm. We present evidence of the sorting efficiency and sorted cell viability, as well as of the senescent nature of the sorted cells, confirmed by the detection of other senescence markers, including the expression of the CKI p21 and the presence of 53BP1 DNA damage foci. Our protocol makes it possible, for the first time, to sort senescent cells from contaminating proliferating cells and, at the same time, to sort subpopulations of senescent cells featuring senescent markers to different extents. Graphical abstract.

4.
Am J Pathol ; 174(2): 423-35, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19147823

ABSTRACT

Normal cells reach senescence after a specific time and number of divisions, leading ultimately to cell death. Although escape from this fate may be a requisite step in neoplastic transformation, the mechanisms governing senescent cell death have not been well investigated. We show here, using normal human epidermal keratinocytes, that no apoptotic markers appear with senescence. In contrast, the expression of several proteins involved in the regulation of macroautophagy, notably Beclin-1 and Bcl-2, was found to change with senescence. The corpses occurring at the senescence growth plateau displayed a large central area delimited by the cytokeratin network that contained a huge quantity of autophagic vacuoles, the damaged nucleus, and most mitochondria. 3-methyladenine, an inhibitor of autophagosome formation, but not the caspase inhibitor zVAD, prevented senescent cell death. We conclude that senescent cells do not die by apoptosis, but as a result of high macroautophagic activity that targets the primary vital cell components.


Subject(s)
Autophagy/physiology , Keratinocytes/pathology , Apoptosis Regulatory Proteins/biosynthesis , Beclin-1 , Blotting, Western , Cellular Senescence/physiology , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Humans , In Situ Nick-End Labeling , Keratinocytes/physiology , Membrane Proteins/biosynthesis , Microscopy, Electron, Transmission , Proto-Oncogene Proteins c-bcl-2/biosynthesis
5.
Oncotarget ; 7(20): 29228-44, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27081082

ABSTRACT

Isolation of prostate stem cells (PSCs) is crucial for understanding their biology during normal development and tumorigenesis. In this aim, we used a transgenic mouse model expressing GFP from the stem cell-specific s-SHIP promoter to mark putative stem cells during postnatal prostate development. Here we show that cells identified by GFP expression are present transiently during early prostate development and localize to the basal cell layer of the epithelium. These prostate GFP+ cells are a subpopulation of the Lin- CD24+ Sca-1+ CD49f+ cells and are capable of self-renewal together with enhanced growth potential in sphere-forming assay in vitro, a phenotype consistent with that of a PSC population. Transplantation assays of prostate GFP+ cells demonstrate reconstitution of prostate ducts containing both basal and luminal cells in renal grafts. Altogether, these results demonstrate that s-SHIP promoter expression is a new marker for neonatal basal prostate cells exhibiting stem cell properties that enables PSCs in situ identification and isolation via a single consistent parameter. Transcriptional profiling of these GFP+ neonatal stem cells showed an increased expression of several components of the Wnt signaling pathway. It also identified stem cell regulators with potential applications for further analyses of normal and cancer stem cells.


Subject(s)
Epithelial Cells/cytology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Prostate/cytology , Stem Cells/cytology , Animals , Biomarkers , Epithelial Cells/metabolism , Male , Mice , Mice, SCID , Mice, Transgenic , Prostate/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism
6.
Nat Commun ; 7: 10399, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26822533

ABSTRACT

The main characteristic of senescence is its stability which relies on the persistence of DNA damage. We show that unlike fibroblasts, senescent epithelial cells do not activate an ATM-or ATR-dependent DNA damage response (DDR), but accumulate oxidative-stress-induced DNA single-strand breaks (SSBs). These breaks remain unrepaired because of a decrease in PARP1 expression and activity. This leads to the formation of abnormally large and persistent XRCC1 foci that engage a signalling cascade involving the p38MAPK and leading to p16 upregulation and cell cycle arrest. Importantly, the default in SSB repair also leads to the emergence of post-senescent transformed and mutated precancerous cells. In human-aged skin, XRCC1 foci accumulate in the epidermal cells in correlation with a decline of PARP1, whereas DDR foci accumulate mainly in dermal fibroblasts. These findings point SSBs as a DNA damage encountered by epithelial cells with aging which could fuel the very first steps of carcinogenesis.


Subject(s)
Cellular Senescence , DNA Breaks, Single-Stranded , Epithelial Cells/cytology , Neoplasms/genetics , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Humans , Neoplasms/metabolism , Neoplasms/physiopathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , X-ray Repair Cross Complementing Protein 1
7.
PLoS One ; 8(5): e63607, 2013.
Article in English | MEDLINE | ID: mdl-23675494

ABSTRACT

The incidence of carcinoma increases greatly with aging, but the cellular and molecular mechanisms underlying this correlation are only partly known. It is established that senescent fibroblasts promote the malignant progression of already-transformed cells through secretion of inflammatory mediators. We investigated here whether the senescent fibroblast secretome might have an impact on the very first stages of carcinogenesis. We chose the cultured normal primary human epidermal keratinocyte model, because after these cells reach the senescence plateau, cells with transformed and tumorigenic properties systematically and spontaneously emerge from the plateau. In the presence of medium conditioned by autologous senescent dermal fibroblasts, a higher frequency of post-senescence emergence was observed and the post-senescence emergent cells showed enhanced migratory properties and a more marked epithelial-mesenchymal transition. Using pharmacological inhibitors, siRNAs, and blocking antibodies, we demonstrated that the MMP-1 and MMP-2 matrix metalloproteinases, known to participate in late stages of cancer invasion and metastasis, are responsible for this enhancement of early migratory capacity. We present evidence that MMPs act by activating the protease-activated receptor 1 (PAR-1), whose expression is specifically increased in post-senescence emergent keratinocytes. The physiopathological relevance of these results was tested by analyzing MMP activity and PAR-1 expression in skin sections. Both were higher in skin sections from aged subjects than in ones from young subjects. Altogether, our results suggest that during aging, the dermal and epidermal skin compartments might be activated coordinately for initiation of skin carcinoma, via a paracrine axis in which MMPs secreted by senescent fibroblasts promote very early epithelial-mesenchymal transition of keratinocytes undergoing transformation and oversynthesizing the MMP-activatable receptor PAR-1.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cellular Senescence , Fibroblasts/metabolism , Matrix Metalloproteinases/metabolism , Paracrine Communication , Receptor, PAR-1/metabolism , Skin/metabolism , Adult , Cell Movement/drug effects , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Hepatocyte Growth Factor/pharmacology , Humans , Keratinocytes/metabolism , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinases/genetics , Receptor, PAR-1/genetics , Skin/pathology , Transforming Growth Factor beta1/pharmacology , Young Adult
8.
PLoS One ; 5(9): e12712, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20856861

ABSTRACT

Senescence is a state of growth arrest resulting mainly from telomere attrition and oxidative stress. It ultimately leads to cell death. We have previously shown that, in keratinocytes, senescence is induced by NF-kappaB activation, MnSOD upregulation and H(2)O(2) overproduction. We have also shown that senescent keratinocytes do not die by apoptosis but as a result of high macroautophagic activity that targets the primary vital cell components. Here, we investigated the mechanisms that activate this autophagic cell death program. We show that corpses occurring at the senescence plateau display oxidatively-damaged mitochondria and nucleus that colocalize with autophagic vacuoles. The occurrence of such corpses was decreased by specifically reducing the H(2)O(2) level with catalase, and, conversely, reproduced by overexpressing MnSOD or applying subtoxic doses of H(2)O(2). This H(2)O(2)-induced cell death did occur through autophagy since it was accompanied by an accumulation of autophagic vesicles as evidenced by Lysotracker staining, LC3 vesiculation and transmission electron microscopy. Most importantly, it was partly abolished by 3-methyladenine, the specific inhibitor of autophagosome formation, and by anti-Atg5 siRNAs. Taken together these results suggest that autophagic cell death is activated in senescent keratinocytes because of the upregulation of MnSOD and the resulting accumulation of oxidative damages to nucleus and mitochondria.


Subject(s)
Autophagy , Keratinocytes/cytology , Keratinocytes/enzymology , Superoxide Dismutase/metabolism , Up-Regulation , Cell Death , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Cellular Senescence , Female , Humans , Hydrogen Peroxide/metabolism , Keratinocytes/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress , Superoxide Dismutase/genetics
9.
Cancer Res ; 69(20): 7917-25, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19826058

ABSTRACT

Studies on human fibroblasts have led to viewing senescence as a barrier against tumorigenesis. Using keratinocytes, we show here that partially transformed and tumorigenic cells systematically and spontaneously emerge from senescent cultures. We show that these emerging cells are generated from senescent cells, which are still competent for replication, by an unusual budding-mitosis mechanism. We further present data implicating reactive oxygen species that accumulate during senescence as a potential mutagenic motor of this post-senescence emergence. We conclude that senescence and its associated oxidative stress could be a tumor-promoting state for epithelial cells, potentially explaining why the incidence of carcinogenesis dramatically increases with advanced age.


Subject(s)
Cell Transformation, Neoplastic , Cellular Senescence , DNA Damage , Neoplasms/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Adenoviridae , Adolescent , Adult , Alu Elements , Blotting, Western , Cell Proliferation , Cells, Cultured , Comet Assay , DNA Probes , Epidermis/metabolism , Epidermis/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fluorescent Antibody Technique , Humans , In Situ Hybridization , Karyotyping , Keratinocytes/metabolism , Keratinocytes/pathology , Middle Aged , Neoplasms/metabolism , Superoxide Dismutase/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL