32,482 research outputs found

    The usefulness of c-Kit in the immunohistochemical assessment of melanocytic lesions

    Get PDF
    C-Kit (CD117), the receptor for the stem cell factor, a growth factor for melanocyte migra- tion and proliferation, has shown differential immunostaining in various benign and malig- nant melanocytic lesions. The purpose of this study is to compare c-Kit immunostaining in benign nevi and in primary and metastatic malignant melanomas, to determine whether c-Kit can aid in the differential diagnosis of these lesions. c-Kit immunostaining was per- formed in 60 cases of pigmented lesions, including 39 benign nevi (5 blue nevi, 5 intra- dermal nevi, 3 junctional nevi, 15 cases of pri- mary compound nevus, 11 cases of Spitz nevus), 18 cases of primary malignant melanoma and 3 cases of metastatic melanoma. The vast majority of nevi and melanomas examined in this study were posi- tive for c-Kit, with minimal differences between benign and malignant lesions. C-Kit cytoplasmatic immunoreactivity in the intraepidermal proliferating nevus cells, was detected in benign pigmented lesions as well as in malignant melanoma, increasing with the age of patients (P=0.007) in both groups. The patient’s age at presentation appeared to be the variable able to cluster benign and malignant pigmented lesions. The percentage of c-Kit positive intraepidermal nevus cells was better associated with age despite other vari- ables (P=0.014). The intensity and percentage of c-Kit positivity in the proliferating nevus cells in the dermis was significantly increased in malignant melanocytic lesions (P=0.015 and P=0.008) compared to benign lesions (compound melanocytic nevi, Spitz nevi, intradermal nevi, blue nevi). Immunostaning for c-Kit in metastatic melanomas was nega- tive. Interestingly in two cases of melanoma occurring on a pre-existent nevus, the melanoma tumor cells showed strong cytoplas- matic and membranous positivity for c-kit, in contrast with the absence of any immunoreac- tivity in pre-existent intradermal nevus cells. C-Kit does not appear to be a strong immuno- histochemical marker for distinguishing melanoma from melanocytic nevi, if we consid- er c-Kit expression in intraepidermal prolifer- ating cells. The c-Kit expression in proliferat- ing melanocytes in the dermis could help in the differential diagnosis between a superfi- cial spreading melanoma (with dermis inva- sion) and a compound nevus or an intradermal nevus. Finally, c-Kit could be a good diagnostic tool for distinguishing benign compound nevi from malignant melanocytic lesions with der- mis invasion and to differentiate metastatic melanoma from primary melanoma

    GM-CSF Inhibits c-Kit and SCF Expression by Bone Marrow-Derived Dendritic Cells.

    Get PDF
    Stem cell factor (SCF), the ligand of c-kit, is a key cytokine for hematopoiesis. Hematopoietic precursors express c-kit, whereas differentiated cells of hematopoietic lineage are negative for this receptor, with the exception of NK cells, mast cells, and a few others. While it has long been recognized that dendritic cells (DCs) can express c-kit, several questions remain concerning the SCF/c-kit axis in DCs. This is particularly relevant for DCs found in those organs wherein SCF is highly expressed, including the bone marrow (BM). We characterized c-kit expression by conventional DCs (cDCs) from BM and demonstrated a higher proportion of c-kit+ cells among type 1 cDC subsets (cDC1s) than type 2 cDC subsets (cDC2s) in both humans and mice, whereas similar levels of c-kit expression were observed in cDC1s and cDC2s from mouse spleen. To further study c-kit regulation, DCs were generated with granulocyte-macrophage colony-stimulating factor (GM-CSF) from mouse BM, a widely used protocol. CD11c+ cells were purified from pooled non-adherent and slightly adherent cells collected after 7 days of culture, thus obtaining highly purified BM-derived DCs (BMdDCs). BMdDCs contained a small fraction of c-kit+ cells, and by replating them for 2 days with GM-CSF, we obtained a homogeneous population of c-kit+ CD40hi MHCIIhi cells. Not only did BMdDCs express c-kit but they also produced SCF, and both were striking upregulated if GM-CSF was omitted after replating. Furthermore, a small but significant reduction in BMdDC survival was observed upon SCF silencing. Incubation of BMdDCs with SCF did not modulate antigen presentation ability of these cells, nor it did regulate their membrane expression of the chemokine receptor CXCR4. We conclude that the SCF/c-kit-mediated prosurvival circuit may have been overlooked because of the prominent use of GM-CSF in DC cultures in vitro, including those human DC cultures destined for the clinics. We speculate that DCs more prominently rely on SCF in vivo in some microenvironments, with potential implications for graft-versus-host disease and antitumor immunity

    Myeloid conditioning with c-kit-targeted CAR-T cells enables donor stem cell engraftment

    Get PDF
    We report a novel approach to bone marrow (BM) conditioning using c-kit-targeted chimeric antigen receptor T (c-kit CAR-T) cells in mice. Previous reports using anti-c-kit or anti-CD45 antibody linked to a toxin such as saporin have been promising. We developed a distinctly different approach using c-kit CAR-T cells. Initial studies demonstrated in vitro killing of hematopoietic stem cells by c-kit CAR-T cells but poor expansion in vivo and poor migration of CAR-T cells into BM. Pre-treatment of recipient mice with low-dose cyclophosphamide (125 mg/kg) together with CXCR4 transduction in the CAR-T cells enhanced trafficking to and expansion in BM (\u3c1%-13.1%). This resulted in significant depletion of the BM c-ki

    Identification of a lineage of multipotent hematopoietic progenitors

    Get PDF
    All multipotent hematopoietic progenitors in C57BL-Thy-1.1 bone marrow are divided among three subpopulations of Thy-1.1^(lo) Sca-1^+ Lin^(-/lo) c-kit^+ cells: long-term reconstituting Mac-1^-CD4^-c-kit^+ cells and transiently reconstituting Mac-1^(lo)CD4^-or Mac-1^(lo) CD4^(lo) cells. This study shows that the same populations, with similar functional activities, exist in mice whose hematopoietic systems were reconstituted by hematopoietic stem cells after lethal irradiation. We demonstrate that these populations form a lineage of multipotent progenitors from long-term self-renewing stem cells to the most mature multipotent progenitor population. In reconstituted mice, Mac-1- CD4^-c-kit^+ cells gave rise to Mac-1^(lo)CD4^- cells, which gave rise to Mac-1^(lo)CD4^(lo) cells. Mac-1^- CD4^-c-kit^+ cells had long-term self-renewal potential, with each cell being capable of giving rise to more than 10^4 functionally similar Mac-1^-CD4^-c-kit^+ cells. At least half of Mac-1^(lo)CD4^- cells had transient self-renewal potential, detected in the spleen 7 days after reconstitution. Mac-1^(lo)CD4^(lo) cells did not have detectable self-renewal potential. The identification of a lineage of multipotent progenitors provides an important tool for identifying genes that regulate self-renewal and lineage commitment

    c-kit +

    Get PDF

    Src-Like Adaptor Protein (SLAP) differentially regulates normal and oncogenic c-Kit signaling

    Get PDF
    The Src-Like Adaptor Protein (SLAP) is an adaptor protein sharing considerable structural homology with Src. SLAP is expressed in variety of cells regulating receptor tyrosine kinase signaling by direct association. In this report, we show that SLAP associates with both wild-type and oncogenic c-Kit (c-Kit-D816V). The association involves SLAP SH2 domain and receptor phosphotyrosine residues different from those mediating Src interaction. Association of SLAP triggers c-Kit ubiquitination which, in turn, is followed by receptor degradation. Although SLAP depletion potentiates c-Kit downstream signaling by stabilizing the receptor, it remains non-functional in c-Kit-D816V signaling. Ligand-stimulated c-Kit or c-Kit-D816V did not alter membrane localization of SLAP. Interestingly oncogenic c-Kit-D816V, but not wild-type c-Kit, phosphorylates SLAP on Y120, Y258 and Y273 residues. Physical interaction between c-Kit-D816V and SLAP is mandatory for the phosphorylation to take place. Although tyrosine phosphorylated SLAP does not affect c-Kit-D816V signaling, mutation of these tyrosine sites to phenylalanine can restore SLAP activity. Taken together the data demonstrate that SLAP negatively regulates wild-type c-Kit signaling, but not its oncogenic counterpart, indicating a possible mechanism by which the oncogenic c-Kit bypasses the normal cellular negative feedback control

    GAB2 is involved in differential pi3-kinase signaling by two splice forms of C-kit.

    Get PDF
    The stem cell factor receptor/c-Kit plays an important physiological role in hematopoesis, melanogenesis and gametogenesis. It has also been implicated in numerous human malignancies. Signal transduction pathways shown to be of importance for c-Kit mediated transformation include the PI3-kinase/Akt pathway. We have previously shown that two alternative splice forms of c-Kit, denoted GNNK- and GNNK+ respectively, mediate distinctively different signals. In this study we find that in the hematopoietic cell line Ba/F3, the GNNK- c-Kit mediates a substantially stronger activation of PI3-kinase/Akt than the GNNK+ c-Kit. This difference in signaling was shown to be dependent on the association of the scaffolding protein Gab2 to c-Kit and Src-mediated phosphorylation of Gab2, to be independent of the direct association of PI3-kinase with c-Kit. Furthermore, proliferation and survival of Ba/F3 cells expressing a mutant of c-Kit that fails to bind to PI3-kinase directly was slightly decreased compared to wild-type c-Kit expressing cells. Using siRNA technology we further verified a role of Gab2 in inducing activation of PI3-kinase/Akt downstream of c-Kit. To summarize, we show that PI3-kinase activation by c-Kit is both splice form dependent and cell type specific. Furthermore, activation of PI3-kinase by c-Kit is dependent both on the direct PI3-kinase binding site in c-Kit as well as on the phosphorylation of Gab2. The fact that c-Kit has been found mutated in numerous human malignancies including acute myeloid leukemia and that Gab2 often is overexpressed in acute myeloid leukemia suggests a potential role of Gab2 mediated PI3-kinase activation in transformation

    C-KIT EXPRESSION IN VARIOUS DEGREES OF HISTOPATHOLOGY DIFFERENTIATION OF RETINOBLASTOMA IN MOHAMMAD HOESIN HOSPITAL PALEMBANG

    Get PDF
    Retinoblastoma is a malignant intraocular neoplasm in pediatric which composed of embyonic tumour cells from retinoblasts of neuroepithelial origin with a relative incidence of 3% of all pediatric tumours. Histopathology examination can confirmed the diagnosis of retinoblastoma. Immunohistochemical staining with C-kit antibody can be used to identify the expression of C-kit in paraffin block of retinoblastoma sample.This study was conducted to identify C-kit expresion in various degrees of histopathology differentiation’s retinoblastoma in Mohammad Hoesin Hospital Palembang.. A descriptive study used on the paraffin blocks of 19 cases retinoblastoma in RSUP dr. Mohammad Hoesin from January 2012- July 2017. The degree of histopathologic differentiation and invasion of retinoblastoma was investigated by Hematoksilin-Eosin stain, and the C-kit expression stained by immunohistochemistry using antibody C-kit ready to use. Data was being presented in tabular form to identify C-kit expression. Retinoblastoma which expressed C-kit in this study are 63,16% among all sample. C-kit expressions were detected in 25% well differentiated, 50 % moderately differentiated, and 25% poorly differentiated of histopathological differentiation degree, also in 50% sample with optic nerve invasion and 58,33% in scleral invasion. C-kit expression was negative in 36,84% sample. In this study, C-kit expression was found more than half in retinoblastoma sample

    Alterations of oncogenes expression in human NK cells in cancer patients

    Get PDF
    C-kit/SCF signaling play a key role in regulating NK cell homeostasis, maturation, proliferation and cytotoxicity. C-kit-deficiency in NK results in significant reduction of their, suggesting imperative role fore-kit signaling in NKcell immunobiology. We have recently showed that human NK cells express not only c-kit-receptor, but also both membrane-bound and soluble forms of c-kit ligand - Stem cell factor. The goal of this study was to characterize the c-kit/SCF autocrine loop in peripheral blood NK cells obtained from patients with cancer

    Hiding inside? Intracellular expression of non-glycosylated c-kit protein in cardiac progenitor cells

    Get PDF
    Cardiac progenitor cells including c-kit(+) cells and cardiosphere-derived cells (CDCs) play important roles in cardiac repair and regeneration. CDCs were reported to contain only small subpopulations of c-kit(+) cells and recent publications suggested that depletion of the c-kit(+) subpopulation of cells has no effect on regenerative properties of CDCs. However, our current study showed that the vast majority of CDCs from murine heart actually express c-kit, albeit, in an intracellular and non-glycosylated form. Immunostaining and flow cytometry showed that the fluorescent signal indicative of c-kit immunostaining significantly increased when cell membranes were permeabilized. Western blots further demonstrated that glycosylation of c-kit was increased during endothelial differentiation in a time dependent manner. Glycosylation inhibition by 1-deoxymannojirimycin hydrochloride (1-DMM) blocked c-kit glycosylation and reduced expression of endothelial cell markers such as Flk-1 and CD31 during differentiation. Pretreatment of these cells with a c-kit kinase inhibitor (imatinib mesylate) also attenuated Flk-1 and CD31 expression. These results suggest that c-kit glycosylation and its kinase activity are likely needed for these cells to differentiate into an endothelial lineage. In vivo, we found that intracellular c-kit expressing cells are located in the wall of cardiac blood vessels in mice subjected to myocardial infarction. In summary, our work demonstrated for the first time that c-kit is not only expressed in CDCs but may also directly participate in CDC differentiation into an endothelial lineage
    corecore