Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biomolecules ; 11(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34439864

ABSTRACT

Orbital (slow flow) cavernous venous hemangiomas (OCVH) are the most common benign orbital tumors in adults. The c-KIT is a tyrosine kinase receptor, which is expressed on several types of cells, is thought to play a key role in tumor pathogenesis. The purpose of this study was to evaluate the presence of the receptor c-KIT in OCVH. Our retrospective study examined 16 orbital cavernous venous hemangiomas from 16 cases operated on between 2006-2016 at Emek Medical Center. The mean tumor size was 18.4 mm. Symptoms appeared between 6 months and 22 years before operation. All specimens were analyzed for the c-KIT receptor through immunohistochemistry. The c-KIT was expressed by the endothelium in all 16 preparates. Staining was strong in two cases, moderate in six, and weak in eight cases, with no statistically significant correlation between staining and tumor size (p = 0.69) or the symptom duration (p = 0.15). We conclude that c-KIT may play an important role in the pathogenesis of OCVH. This pilot study is significant in that tumor-targeted therapy such as Imatinib Mesylate and Sunitinib may have a role in treating surgically complicated cases located in the orbital apex. A large multicenter collaborative study is necessary to examine the role of c-KIT in OCVH.


Subject(s)
Gene Expression Regulation, Neoplastic , Hemangioma, Cavernous/metabolism , Orbital Neoplasms/metabolism , Proto-Oncogene Proteins c-kit/biosynthesis , Adolescent , Adult , Aged , Child , Female , Hemangioma, Cavernous/genetics , Hemangioma, Cavernous/pathology , Humans , Male , Middle Aged , Orbital Neoplasms/genetics , Orbital Neoplasms/pathology , Proto-Oncogene Proteins c-kit/genetics , Young Adult
2.
PLoS One ; 11(9): e0162984, 2016.
Article in English | MEDLINE | ID: mdl-27690130

ABSTRACT

Human monocyte-derived dendritic cells (mdDCs) are versatile cells that are used widely for research and experimental therapies. Although different culture conditions can affect their characteristics, there are no known subpopulations. Since monocytes differentiate into dendritic cells (DCs) in a variety of tissues and contexts, we asked whether they can give rise to different subpopulations. In this work we set out to characterize two human mdDC subpopulations that we identified and termed small (DC-S) and large (DC-L). Morphologically, DC-L are larger, more granular and have a more complex cell membrane. Phenotypically, DC-L show higher expression of a wide panel of surface molecules and stronger responses to maturation stimuli. Transcriptomic analysis confirmed their separate identities and findings were consistent with the phenotypes observed. Although they show similar apoptotic cell uptake, DC-L have different capabilities for phagocytosis, demonstrate better antigen processing, and have significantly better necrotic cell uptake. These subpopulations also have different patterns of cell death, with DC-L presenting an inflammatory, "dangerous" phenotype while DC-S mostly downregulate their surface markers upon cell death. Apoptotic cells induce an immune-suppressed phenotype, which becomes more pronounced among DC-L, especially after the addition of lipopolysaccharide. We propose that these two subpopulations correspond to inflammatory (DC-L) and steady-state (DC-S) DC classes that have been previously described in mice and humans.

3.
PLoS One ; 7(5): e35602, 2012.
Article in English | MEDLINE | ID: mdl-22590508

ABSTRACT

To reach the lymphatics, migrating dendritic cells (DCs) need to interact with the extracellular matrix (ECM). Heparanase, a mammalian endo-ß-D-glucuronidase, specifically degrades heparan sulfate proteoglycans ubiquitously associated with the cell surface and ECM. The role of heparanase in the physiology of bone marrow-derived DCs was studied in mutant heparanase knock-out (Hpse-KO) mice. Immature DCs from Hpse-KO mice exhibited a more mature phenotype; however their transmigration was significantly delayed, but not completely abolished, most probably due to the observed upregulation of MMP-14 and CCR7. Despite their mature phenotype, uptake of beads was comparable and uptake of apoptotic cells was more efficient in DCs from Hpse-KO mice. Heparanase is an important enzyme for DC transmigration. Together with CCR7 and its ligands, and probably MMP-14, heparanase controls DC trafficking.


Subject(s)
Bone Marrow Cells/immunology , Dendritic Cells/immunology , Glucuronidase/immunology , Transendothelial and Transepithelial Migration/immunology , Animals , Bone Marrow Cells/enzymology , Dendritic Cells/enzymology , Extracellular Matrix/genetics , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Glucuronidase/genetics , Glucuronidase/metabolism , Heparitin Sulfate/genetics , Heparitin Sulfate/immunology , Heparitin Sulfate/metabolism , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/immunology , Mice , Mice, Knockout , Receptors, CCR7/biosynthesis , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Transendothelial and Transepithelial Migration/genetics , Up-Regulation/genetics , Up-Regulation/immunology
4.
PLoS One ; 7(2): e29333, 2012.
Article in English | MEDLINE | ID: mdl-22363402

ABSTRACT

Programmed cell death (PCD) is a fundamental mechanism in tissue and cell homeostasis. It was long suggested that apoptosis regulates the cell number in diverse cell populations; however no clear mechanism was shown. Neutrophils are the short-lived, first-line defense of innate immunity, with an estimated t = 1/2 of 8 hours and a high turnover rate. Here we first show that spontaneous neutrophil constitutive PCD is regulated by cell concentrations. Using a proteomic approach, we identified the S100 A8/9 complex, which constitutes roughly 40% of cytosolic protein in neutrophils, as mediating this effect. We further demonstrate that it regulates cell survival via a signaling mechanism involving MEK-ERK via TLR4 and CD11B/CD18. This mechanism is suggested to have a fine-tuning role in regulating the neutrophil number in bone marrow, peripheral blood, and inflammatory sites.


Subject(s)
Apoptosis , Calgranulin A/metabolism , Calgranulin B/metabolism , MAP Kinase Signaling System , Neutrophils/cytology , Neutrophils/enzymology , Amino Acid Sequence , Apoptosis Regulatory Proteins/metabolism , CD18 Antigens/metabolism , Calgranulin A/chemistry , Calgranulin B/chemistry , Cell Count , Cell Survival , Humans , Mass Spectrometry , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Neutrophil Activation , Proteomics , Proto-Oncogene Proteins c-bcl-2/metabolism , Subcellular Fractions/metabolism , Toll-Like Receptor 4/metabolism
5.
Eur J Immunol ; 40(3): 699-709, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20039295

ABSTRACT

In recent years, it has become apparent that the removal of apoptotic cells by macrophages and DC is not only noninflammatory, but also immune-inhibitory, in most although not all circumstances. Complement may be involved in the uptake of apoptotic cells via direct binding of bridging factors in some physiological circumstances, by opsonization and engagement of the complement receptors. In the current study, we use a complement-dependent system of apoptotic cell clearance by human-derived macrophages and DC. Using a luciferase reporter gene and measuring immune response to non-opsonic zymosan, we show that iC3b-apoptotic cells induce NF-kappaB inhibition in response to zymosan and LPS at the nuclear translocation, transcriptional and post-transcriptional levels, leading to profound inhibition of proinflammatory cytokines. In addition, interaction with iC3b-opsonized apoptotic cells is characterized by macrophage secretion of IL-10 and lack of TGF-beta secretion. In conclusion, in cells with iC3b receptors, opsonized apoptotic cells mediate a distinct anti-inflammatory response and transcriptional NF-kappaB-dependent blockage.


Subject(s)
Anti-Inflammatory Agents/metabolism , Apoptosis/physiology , Complement C3b/metabolism , NF-kappa B/metabolism , Phagocytosis/physiology , Animals , Anti-Inflammatory Agents/immunology , Blotting, Western , Cell Line , Cell Separation , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Macrophages/metabolism , Mice , Microscopy, Fluorescence
6.
Apoptosis ; 12(10): 1769-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17578666

ABSTRACT

One hallmark of programmed cell death (PCD) is redistribution of phosphatidylserine (PS) to the plasma membrane's outer leaflet. Annexin V is widely used in cell death research due to its calcium-dependent ability to bind phosphatidylserine, thus marking apoptotic cells. However, calcium is invariably used at high concentrations in annexin V staining, at doses that can induce cell death. We used flow cytometric annexin V staining, together with propidium iodide and TMRM for determination of dissipation of mitochondrial potential, with a variety of calcium concentrations, cell media, and incubation times, to identify a possible bias in PCD determination of human primary leukocytes. Here we show that measurements of PCD in human monocytes, polymorphonuclear cells, and monocyte-derived dendritic cells using annexin V may be dramatically affected by calcium concentration, time of incubation on ice, and media choice. We propose a method that enables accurate and unbiased annexin V staining, without affecting results.


Subject(s)
Annexin A5/metabolism , Calcium/metabolism , Cell Death/physiology , Enzyme Inhibitors/metabolism , Leukocytes/physiology , Buffers , Cell Membrane/metabolism , Cell Survival , Cells, Cultured , Culture Media/chemistry , Humans , Leukocytes/cytology , Membrane Potentials/physiology , Mitochondria/metabolism , Phosphatidylserines/metabolism , Staining and Labeling , Time Factors
7.
Blood ; 108(10): 3580-9, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16882710

ABSTRACT

Apoptotic cells were shown to induce dendritic cell immune tolerance. We applied a proteomic approach to identify molecules that are secreted from apoptotic monocytes, and thus may mediate engulfment and immune suppression. Supernatants of monocytes undergoing apoptosis were collected and compared using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and differentially expressed proteins were identified using tandem mass spectrometry. Thrombospondin-1 (TSP-1) and its cleaved 26-kDa heparin-binding domain (HBD) were identified. We show that TSP-1 is expressed upon induction of monocyte apoptosis in a caspase-dependent pattern and the HBD is cleaved by chymotrypsin-like serine protease. We further show that CD29, CD36, CD47, CD51, and CD91 simultaneously participate in engulfment induction and generation of an immature dendritic cell (iDC) tolerogenic and phagocytic state. We conclude that apoptotic cell TSP-1, and notably its HBD, creates a signalosome in iDCs to improve engulfment and to tolerate engulfed material prior to the interaction with apoptotic cells.


Subject(s)
Apoptosis/immunology , Dendritic Cells/physiology , Immune Tolerance , Monocytes/metabolism , Phagocytosis , Thrombospondin 1/biosynthesis , Antigens, CD/physiology , Binding Sites , Gene Expression Regulation , Heparin , Humans , Monocytes/cytology , Proteomics/methods , Thrombospondin 1/isolation & purification , Thrombospondin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...