ABSTRACT
As an important component of solid tumors, mast cells show specific phenotypes in various tumor microenvironments. However, the precise mechanism of mast cell accumulation and the phenotypic features of thyroid cancer (TC) remain largely unknown. Here, we found that mast cells were obviously recruited to tumor tissue by TC-derived stem cell factor (SCF). With tumor progression, mast cell levels increased gradually. In addition, intratumoral mast cells expressed higher levels of the immunosuppressive molecule galectin-9, which effectively suppresses CD8+ T-cell antitumor immunity in vitro. Blocking galectin-9 on tumor-infiltrating mast cells reversed the immunosuppression of CD8+ T cells. In conclusion, our data elucidated novel protumorigenic and immunosuppressive roles of mast cells in TC. In addition, our results indicated that blocking mast cells may impede tumor progression and ameliorate the prognosis of TC patients.
ABSTRACT
Human stem cell factor (hSCF) is an early-acting growth factor that promotes proliferation, differentiation, migration, and survival in several tissues. It plays a crucial role in hematopoiesis, gametogenesis, melanogenesis, intestinal motility, and in development and recovery of nervous and cardiovascular systems. Potential therapeutic applications comprise anemia treatment, mobilization of hematopoietic stem/progenitor cells to peripheral blood, and increasing gene transduction efficiency for gene therapy. Developing new tools to characterize recombinant hSCF in most native-like form as possible is crucial to understand the complexity of its in vivo functions and for improving its biotechnological applications. The soluble domain of hSCF was expressed in HEK293 cells. Highly purified rhSCF showed great molecular mass variability due to the presence of N- and O-linked carbohydrates, and it presented a 2.5-fold increase on proliferative activity compared to bacteria-derived hSCF. Three hybridoma clones producing monoclonal antibodies (mAbs) with high specificity for the glycoprotein were obtained. 1C4 and 2D3 mAbs were able to detect bacteria-derived and glycosylated rhSCF and demonstrated to be excellent candidates to develop a sandwich ELISA assay for rhSCF quantification, with detection limits of 0.18 and 0.07 ng/ml, respectively. Interestingly, 1A10 mAb only recognized glycosylated rhSCF, suggesting that sugar moieties might be involved in epitope recognition. 1A10 mAb showed the highest binding affinity, and it constituted the best candidate for immunodetection of the entire set rhSCF glycoforms in western blot assays, and for intracellular cytokine staining. Our work shows that combining glycosylated rhSCF expression with hybridoma technology is a powerful strategy to obtain specific suitable immunochemical assays and thus improve glycoprotein-producing bioprocesses. KEY POINTS: ⢠Soluble glycosylated human SCF exerted improved proliferative activity on UT-7 cells. ⢠Three mAbs with high specificity targeting glycosylated human SCF were obtained. ⢠mAbs applications comprise sandwich ELISA, western blot, and immunofluorescence assays.
Subject(s)
Antibodies, Monoclonal , Glycoproteins , Hybridomas , Stem Cell Factor , Humans , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Biotechnology , Glycoproteins/immunology , HEK293 Cells , Stem Cell Factor/analysis , Stem Cell Factor/immunology , Glycosylation , Enzyme-Linked Immunosorbent Assay , Blotting, WesternABSTRACT
Triple-negative breast cancer (TNBC) is associated with a poor prognosis and the absence of targeted therapy. c-Kit, a receptor tyrosine kinase (RTK), is considered a molecular target for anticancer drugs. Tyrosine kinase inhibitors (TKIs) recognizing c-Kit are used for the treatment of c-Kit-expressing tumors. However, the expression, function, and therapeutic potential of c-Kit have been little explored in TNBC. Here, we studied the expression and effects of c-Kit in TNBC through in vitro and in silico analysis, and evaluated the response to TKIs targeting c-Kit. Analysis of TNBC cells showed the expression of functional c-Kit at the cell membrane. The stimulation of c-Kit with its ligand induced the activation of STAT3, Akt, and ERK1/2, increasing cell migration, but had no effect on cell proliferation or response to Doxorubicin. Analysis of public datasets showed that the expression of c-Kit in tumors was not associated with patient survival. Finally, TNBC cells were susceptible to TKIs, in particular the effect of Nilotinib was stronger than Doxorubicin in all cell lines. In conclusion, TNBC cells express functional c-Kit, which is a targetable molecule, and show a strong response to Nilotinib that may be considered a candidate drug for the treatment of TNBC.
Subject(s)
Triple Negative Breast Neoplasms , Cell Line, Tumor , Cell Proliferation , Doxorubicin/pharmacology , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/genetics , Receptor Protein-Tyrosine Kinases , Triple Negative Breast Neoplasms/pathologyABSTRACT
[This corrects the article DOI: 10.3389/fbioe.2020.00620.].
ABSTRACT
Cell therapy products exhibit great therapeutic potential but come with a deterring price tag partly caused by their costly manufacturing processes. The development of strategies that lead to cost-effective cell production is key to expand the reach of cell therapies. Growth factors are critical culture media components required for the maintenance and differentiation of cells in culture and are widely employed in cell therapy manufacturing. However, they are expensive, and their common use in soluble form is often associated with decreased stability and bioactivity. Immobilization has emerged as a possible strategy to optimize growth factor use in cell culture. To date, several immobilization techniques have been reported for attaching growth factors onto a variety of biomaterials, but these have been focused on tissue engineering. This review briefly summarizes the current landscape of cell therapy manufacturing, before describing the types of chemistry that can be used to immobilize growth factors for cell culture. Emphasis is placed to identify strategies that could reduce growth factor usage and enhance bioactivity. Finally, we describe a case study for stem cell factor.
ABSTRACT
The objective was to study the effect of mechanical intestinal obstruction in rats on the phenotype of interstitial cells of Cajal (ICC). Healthy Wistar rats were randomly divided into sham-operation group (C), one day obstruction group (M1), two days obstruction group (M2), and three days obstruction group (M3), with 10 rats in each group. The expression of SCF mRNA and c-Kit protein in intestinal tissue was investigated by RT-PCR and immunohistochemistry. Compared with the sham-operation group, the relative expression of SCF mRNA and the expression of c-Kit protein in intestinal tissue were significantly decreased in both obstruction groups. Levels decreased gradually with the prolongation of obstruction time, and significantly decreased on the 3rd day after obstruction (P<0.05). Immunohistochemical staining of the small intestine showed that the number of ICC in the sham-operation group was the highest, and they were gradually decreased with the extension of obstruction time in the M1 to M3 groups. There was a significant difference between groups (P<0.05). Intestinal obstruction caused a decrease in the concentrations of SCF mRNA and c-Kit protein in ICC. With the prolongation of intestinal obstruction, the number of ICCs gradually decreased.
Subject(s)
Animals , Male , Rats , RNA, Messenger/metabolism , Stem Cell Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Interstitial Cells of Cajal/metabolism , Intestinal Obstruction/metabolism , Phenotype , Immunohistochemistry , Rats, Wistar , Disease Models, Animal , Interstitial Cells of Cajal/pathology , Intestinal Obstruction/pathologyABSTRACT
Abstract: The growth factor receptor c-kit (CD117) is expressed in immature T-cells and in some advanced forms of mycosis fungoides. c-kit gene mutation results in unrestricted neoplastic proliferation. We aimed to detect by PCR the most frequent exon mutations in seventeen plaque-stage MF patients, in their perilesional skin and in healthy skin donors. We secondarily evaluated CD117 expression by immunohistochemistry in plaque-stage and tumor-stage MF. We detected no mutation in c-kit gene and low CD117 expression was confirmed on atypical cells in one patient. Complete c-kit exon and intron sequences should be assessed and more sensitive sequencing method could be also applied.
Subject(s)
Humans , Male , Female , Aged , Exons/genetics , Mycosis Fungoides/genetics , Proto-Oncogene Proteins c-kit/genetics , Mutation/genetics , Immunohistochemistry , Case-Control Studies , Gene Expression , Polymerase Chain Reaction , Prospective StudiesABSTRACT
Cell migration of normal cells is tightly regulated. However, tumor cells are exposed to a modified microenvironment that promotes cell migration. Invasive migration of tumor cells is stimulated by receptor tyrosine kinases (RTKs) and is regulated by growth factors. Erythropoietin (Epo) is a glycoprotein hormone that regulates erythropoiesis and is also known to be a potent chemotactic agent that induces cell migration by binding to its receptor (EpoR). Expression of EpoR has been documented in tumor cells, and the potential of Epo to induce cell migration has been explored. Stem cell factor (SCF) is a cytokine that synergizes the effects of Epo during erythropoiesis. SCF is the ligand of c-Kit, a member of the RTKs family. Molecular activity of RTKs is a primary stimulus of cell motility. Thus, expression of the SCF/c-Kit axis is associated with cell migration. In this chapter, we summarize data describing the potential effect of Epo/EpoR and SCF/c-Kit as promoters of cancer cell migration. We also integrate recent findings on molecular mechanisms of Epo/EpoR- and SCF/c-Kit-mediated migration described in various cancer models.
Subject(s)
Cell Movement/physiology , Erythropoietin/metabolism , Neoplasms/metabolism , Stem Cell Factor/metabolism , Gene Expression Regulation, Neoplastic , HumansABSTRACT
Leukemia cells produce acidic metabolites due to their high metabolic condition. An alkaline pHi (intracellular pH) shift, caused by activation of the Na+/H+ exchange, is an important event in the mechanism of growth factor activity. However, the role of the Na(+)/H(+) exchanger in the survival of erythroleukemia TF-1 cells has not yet been studied in detail. The aim of this study was to identify the effects of 5-(N-ethyl-N-isopropyl) amiloride (EIPA), a highly specific blocker of the Na(+)/H(+) exchanger, on the survival of SCF-dependent TF-1 cells. The effects of EIPA on survival and mitochondrial membrane potential were studied when exposing wild type TF-1 cells and TF-1 cells expressing bcl-2 to EIPA for 48h. Ectopic expression of the bcl-2 gene maintained a mildly alkaline pH and prevented the simultaneous appearance of apoptosis and autophagy (typically displayed by TF-1 cells) in the presence of EIPA. Consistent with Stem Cell Factor (SCF) function, we found that this molecule rescued TF-1 cells during autophagy but not apoptosis, allowing these cells to subsequently respond to GM-CSF. Serum deprivation or SCF withdrawal induced cell death at 36h in TF-1 and TF-1 neo cells, whereas TF-1/bcl-2 cells tended to undergo apoptosis and show acidic vacuoles after 96h, pointing to a transient anti-apoptotic effect. The present study shows the suppressive effect of EIPA on the proliferation of leukemia cell line stimulated with SCF, apparently by decreasing the mitochondria membrane potential and averting alkalinization. Through the constitutive expression of bcl-2, TF-1 cells were survival factor independent. Proliferation in these cells was not affected by EIPA at the concentrations used against parental TF-1 cells, indicating that the inhibitory effect in SCF-stimulated cells can be attributed to specific blocking of the Na(+)/H(+) exchanger.
Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Erythroblastic, Acute/metabolism , Leukemia/drug therapy , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/chemistry , Stem Cell Factor/metabolism , Amiloride/analogs & derivatives , Amiloride/chemistry , Apoptosis , Autophagy , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Separation , Cell Survival , Fluorescent Dyes/chemistry , Humans , Hydrogen-Ion Concentration , Leukemia/metabolism , Leukemia, Erythroblastic, Acute/pathology , Membrane Potentials , Mitochondrial Membranes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolismABSTRACT
OBJECTIVE: Endometriosis is linked to altered cell proliferation and stem cell markers c-kit/stem cell factor (SCF) in ectopic endometrium. Our aim was to investigate whether c-kit/SCF also plays a role in eutopic endometrium. DESIGN: Eutopic endometrium obtained from 35 women with endometriosis and 25 fertile eumenorrheic women was analyzed for in situ expression of SCF/c-kit, Ki67, RAC-alpha serine/threonine-protein kinase (Akt), phosphorylated RAC-alpha serine/threonin-protein kinase (pAkt), Glycogen synthase kinase 3 beta (GSK3ß), and phosphorylated glycogen synthase kinase 3 beta (pGSK3ß), throughout the menstrual cycle. RESULTS: Expression of Ki67 and SCF was higher in endometriosis than in control tissue (P < .05) and greater in secretory rather than proliferative (P < .01) endometrium in endometriosis. Expression of c-kit was also higher in endometriosis although similar in both phases. Expression of Akt and GSK3ß was identical in all samples and cycle phases, whereas pAkt and pGSK3ß, opposed to control tissue, remained overexpressed in the secretory phase in endometriosis. CONCLUSION: Unceasing cell proliferation in the secretory phase of eutopic endometriosis is linked to deregulation of c-kit/SCF-associated signaling pathways.
Subject(s)
Cell Proliferation , Endometriosis/enzymology , Endometrium/enzymology , Glycogen Synthase Kinase 3/analysis , Proto-Oncogene Proteins c-akt/analysis , Adult , Biopsy , Case-Control Studies , Endometriosis/pathology , Endometriosis/physiopathology , Endometrium/metabolism , Endometrium/pathology , Female , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Phosphorylation , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Young AdultABSTRACT
Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF). We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes. Male C57/B6 mice (20-25 g) were randomly divided into four groups of 10 animals each. The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL. SCF, ERK1/2 and AKT were measured by Western blotting. In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice. The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 ± 0.077 percent, 2.83 ± 0.156 percent, 1.86 ± 0.094 percent, and 1.78 ± 0.095 percent (mean ± SEM), respectively. SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex. Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex. However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex. The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways. These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.