Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Appl Microbiol Biotechnol ; 106(24): 8121-8137, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36401641

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales , Glicoproteínas , Hibridomas , Factor de Células Madre , Humanos , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Biotecnología , Glicoproteínas/inmunología , Células HEK293 , Factor de Células Madre/análisis , Factor de Células Madre/inmunología , Glicosilación , Ensayo de Inmunoadsorción Enzimática , Western Blotting
2.
Immunology ; 159(4): 441-449, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31957000

RESUMEN

Signaling by Kit has been extensively studied in hematopoietic cells and is essential for the survival, proliferation and maintenance of hematopoietic stem and progenitor cells. In addition to the activation of intrinsic signaling pathways, Kit has been shown to interact with lineage-restricted type I cytokine receptors and produce cross signals, e.g. erythropoietin receptor, interleukin-7 receptor (IL-7R), IL-3R. Based on the earlier studies, we hypothesize that Kit activate other type I cytokine receptors in a cell-specific manner and execute cell-specific function. To investigate other Kit-activated receptors, we tested Kit and IL-4R cross-receptor activation in murine bone-marrow-derived mast cells, which express both Kit and IL-4R at the surface level. Kit upon activation by Kit ligand (KL), activated IL-4Rα, γC , and signal transducer and activator of transcription 6 independent of its cognate ligand IL-4. Though KL and IL-4 are individually mitogenic, combinations of KL and IL-4 synergistically promoted mast cell proliferation. Furthermore, inhibition of lipid raft formation by methyl-ß-cyclodextrin resulted in loss of synergistic proliferation. Together the data suggest IL-4R as a novel Kit-activated receptor. Such cross-receptor activations are likely to be a universal mechanism of Kit signaling in hematopoiesis.


Asunto(s)
Interleucina-4/farmacología , Mastocitos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-kit/genética , Receptores de Interleucina-4/genética , Factor de Transcripción STAT6/genética , Factor de Células Madre/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Hematopoyesis/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Mastocitos/citología , Mastocitos/inmunología , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-kit/inmunología , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/inmunología , Receptores de Interleucina-3/genética , Receptores de Interleucina-3/inmunología , Receptores de Interleucina-4/inmunología , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/inmunología , Factor de Transcripción STAT6/inmunología , Transducción de Señal , Factor de Células Madre/genética , Factor de Células Madre/inmunología , beta-Ciclodextrinas/farmacología
3.
Eur J Immunol ; 47(1): 206-212, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27739072

RESUMEN

Long-term proliferating, DH JH -rearranged mouse precursor B-cell lines have previously been established in serum- and IL-7-containing media from fetal liver, but not from bone marrow. Serum and stromal cells expose these pre-B cells to undefined factors, hampering accurate analyses of ligand-dependent signaling, which controls pre-B cell proliferation, survival, residence and migration. Here, we describe a novel serum-free, stromal cell-free culture system, which allows us to establish and maintain pre-B cells not only from fetal liver, but also from bone marrow with practically identical efficiencies in proliferation, cloning and differentiation. Surprisingly, recombinant kit-ligand, also called stem cell factor, produced as a kit-ligand-Fc fusion protein, suffices to replace stromal cells and serum, provided that it is presented to cultured pre-B cells in an optimal density in plate-bound, insolubilized, potentially crosslinking form. Additional recombinant CXCL12 and fibronectin have a minor influence on the establishment and maintenance of pre-B cell lines and clones from fetal liver, but are necessary to establish such cell lines from bone marrow.


Asunto(s)
Interleucina-7/metabolismo , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo , Factor de Células Madre/inmunología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Células Clonales , Medios de Cultivo , Interleucina-7/farmacología , Ratones , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/efectos de los fármacos , Factor de Células Madre/farmacología
4.
Brain Behav Immun ; 64: 116-123, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28392427

RESUMEN

Immune dysregulation plays a role in the vulnerability for mood disorders. Immune growth factors, such as Stem Cell Factor (SCF), Insulin-like Growth Factor-Binding Protein-2 (IGF-BP2), Epidermal Growth Factor (EGF), IL-7 and sCD25 have repeatedly been reported altered in patients with mood disorders. The aim of this study was to investigate levels of these factors in serum of adolescent bipolar offspring, who have a heightened risk for mood disorder development and to also analyze the data combined with previously published data. Growth factors were assessed by CBA/ELISA in adolescent bipolar offspring (n=96, mean age=16years) and in age- and gender-matched healthy controls (n=50). EGF belonged to a mutually correlating cluster of mainly neurotrophic compounds including S100B and BDNF, which were in general decreased in serum. IL-7, SCF, IGF-BP2 and sCD25, belonged to a different mutually correlating cluster of immune growth factors, which were in general increased: IGF-BP2 significantly in serum of offspring without a mood disorder, IL-7 and SCF in serum of offspring who had experienced a mood episode. This pattern of de- and increases was not different between bipolar offspring that developed or did not develop a mood disorder over time, apart from the IGF-BP2 level, which was near significantly higher in offspring later developing a mood disorder. Correlations with the previously published immune-cellular abnormalities were not found. In conclusion non-affected adolescents at familial mood disorder development risk were characterized by a distinct pattern of a series of compounds operating in a network of hematopoiesis, neurogenesis and inflammation.


Asunto(s)
Trastorno Bipolar/sangre , Trastorno Bipolar/inmunología , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/inmunología , Adolescente , Trastorno Bipolar/complicaciones , Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/inmunología , Hijo de Padres Discapacitados , Factor de Crecimiento Epidérmico/sangre , Factor de Crecimiento Epidérmico/inmunología , Femenino , Humanos , Inflamación/complicaciones , Inflamación/inmunología , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/inmunología , Interleucina-7/sangre , Interleucina-7/inmunología , Masculino , Subunidad beta de la Proteína de Unión al Calcio S100/sangre , Subunidad beta de la Proteína de Unión al Calcio S100/inmunología , Factor de Células Madre/sangre , Factor de Células Madre/inmunología
5.
Pediatr Res ; 79(4): 637-46, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26705118

RESUMEN

BACKGROUND: Accumulating evidence suggests that c-kit-positive cells are present in the remodeled pulmonary vasculature bed of patients with pulmonary hypertension (PH). Whether stem cell factor (SCF)/c-kit-regulated pathways potentiate pulmonary vascular remodeling is unknown. Here, we tested the hypothesis that attenuated c-kit signaling would decrease chronic hypoxia-induced pulmonary vascular remodeling by decreasing pulmonary vascular cell mitogenesis. METHODS: Neonatal FVB/NJ mice treated with nonimmune IgG (placebo), or c-kit neutralizing antibody (ACK2) as well as c-kit mutant mice (WBB6F1-Kit(W-v/+)) and their congenic controls, were exposed to normoxia (FiO2 = 0.21) or hypoxia (FiO2 = 0.12) for 2 wk. Following this exposure, right ventricular systolic pressure (RVSP), right ventricular hypertrophy (RVH), pulmonary vascular cell proliferation, and remodeling were evaluated. RESULTS: As compared to chronically hypoxic controls, c-kit mutant mice had decreased RVSP, RVH, pulmonary vascular remodeling, and proliferation. Consistent with these findings, administration of ACK2 to neonatal mice with chronic hypoxia-induced PH decreased RVSP, RVH, pulmonary vascular cell proliferation, and remodeling. This attenuation in PH was accompanied by decreased extracellular signal-regulated protein kinase (ERK) 1/2 activation. CONCLUSION: SCF/c-kit signaling may potentiate chronic hypoxia-induced vascular remodeling by modulating ERK activation. Inhibition of c-kit activity may be a potential strategy to alleviate PH.


Asunto(s)
Modelos Animales de Enfermedad , Hipoxia/fisiopatología , Pulmón/irrigación sanguínea , Transducción de Señal , Factor de Células Madre/metabolismo , Remodelación Vascular , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/inmunología , Enfermedad Crónica , Hipoxia/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Mutantes , Factor de Células Madre/inmunología
6.
J Immunol ; 192(10): 4859-66, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24733849

RESUMEN

SHP2 protein-tyrosine phosphatase (encoded by Ptpn11) positively regulates KIT (CD117) signaling in mast cells and is required for mast cell survival and homeostasis in mice. In this study, we uncover a role of SHP2 in promoting chemotaxis of mast cells toward stem cell factor (SCF), the ligand for KIT receptor. Using an inducible SHP2 knockout (KO) bone marrow-derived mast cell (BMMC) model, we observed defects in SCF-induced cell spreading, polarization, and chemotaxis. To address the mechanisms involved, we tested whether SHP2 promotes activation of Lyn kinase that was previously shown to promote mast cell chemotaxis. In SHP2 KO BMMCs, SCF-induced phosphorylation of the inhibitory C-terminal residue (pY507) was elevated compared with control cells, and phosphorylation of activation loop (pY396) was diminished. Because Lyn also was detected by substrate trapping assays, these results are consistent with SHP2 activating Lyn directly by dephosphorylation of pY507. Further analyses revealed a SHP2- and Lyn-dependent pathway leading to phosphorylation of Vav1, Rac activation, and F-actin polymerization in SCF-treated BMMCs. Treatment of BMMCs with a SHP2 inhibitor also led to impaired chemotaxis, consistent with SHP2 promoting SCF-induced chemotaxis of mast cells via a phosphatase-dependent mechanism. Thus, SHP2 inhibitors may be useful to limit SCF/KIT-induced mast cell recruitment to inflamed tissues or the tumor microenvironment.


Asunto(s)
Quimiotaxis/inmunología , Mastocitos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-vav/inmunología , Factor de Células Madre/inmunología , Familia-src Quinasas/inmunología , Animales , Quimiotaxis/genética , Activación Enzimática/genética , Activación Enzimática/inmunología , Mastocitos/citología , Ratones , Ratones Noqueados , Fosforilación/genética , Fosforilación/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-vav/genética , Factor de Células Madre/genética , Familia-src Quinasas/genética
7.
J Immunol ; 192(12): 5471-5475, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24829419

RESUMEN

We reported previously that c-kit ligation by membrane-bound stem cell factor (mSCF) boosts IL-6 production in dendritic cells (DCs) and a Th17-immune response. However, Th17 establishment also requires heterodimeric IL-23, but the mechanisms that regulate IL-23 gene expression in DCs are not fully understood. We show that IL-23p19 gene expression in lung DCs is dependent on mSCF, which is regulated by the metalloproteinase MMP-9. Th1-inducing conditions enhanced MMP-9 activity, causing cleavage of mSCF, whereas the opposite was true for Th17-promoting conditions. In MMP-9(-/-) mice, a Th1-inducing condition could maintain mSCF and enhance IL-23p19 in DCs, promoting IL-17-producing CD4(+) T cells in the lung. Conversely, mSCF cleavage from bone marrow DCs in vitro by rMMP-9 led to reduced IL-23p19 expression under Th17-inducing conditions, with dampening of intracellular AKT phosphorylation. Collectively, these results show that the c-kit/mSCF/MMP-9 axis regulates IL-23 gene expression in DCs to control IL-17 production in the lung.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-17/inmunología , Subunidad p19 de la Interleucina-23/inmunología , Pulmón/inmunología , Metaloproteinasa 9 de la Matriz/inmunología , Factor de Células Madre/inmunología , Animales , Membrana Celular/genética , Membrana Celular/inmunología , Membrana Celular/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/genética , Interleucina-17/genética , Interleucina-17/metabolismo , Subunidad p19 de la Interleucina-23/biosíntesis , Subunidad p19 de la Interleucina-23/genética , Pulmón/citología , Pulmón/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Fosforilación/genética , Fosforilación/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 34(7): 1459-67, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24790137

RESUMEN

OBJECTIVE: Although stem cell factor (SCF) has been shown to play a critical role in hematopoiesis, gametogenesis, and melanogenesis, the function of SCF in the regulation of vascular integrity has not been studied. APPROACH AND RESULTS: We demonstrated that SCF binds to and activates the cKit receptor in endothelial cells, thereby increasing the internalization of vascular endothelial-cadherin and enhancing extravasation of dyes to a similar extent as vascular endothelial growth factor. SCF-mediated cKit activation in endothelial cells enhanced the phosphorylation of endothelial nitric oxide (NO) synthase via the phosphoinositide 3-kinase/Akt signaling pathway and subsequently increased the production of NO. Inhibition of endothelial NO synthase expression and NO synthesis using small interfering RNA knockdown and chemical inhibitors substantially diminished the ability of SCF to increase the internalization of vascular endothelial-cadherin and in vitro endothelial permeability. SCF-induced increase in extravasation of the dyes was abrogated in endothelial NO synthase knockout mice, which indicates that endothelial NO synthase-mediated NO production was responsible for the SCF-induced vascular leakage. Furthermore, we demonstrated that the expression of SCF and cKit was significantly higher in the retina of streptozotocin-injected diabetic mice than in the nondiabetic control animals. Depletion of SCF by intravitreous injection of anti-SCF-neutralizing immunoglobulin G significantly prevented vascular hyperpermeability in the retinas of streptozotocin-injected diabetic mice. CONCLUSIONS: Our data reveal that SCF disrupts the endothelial adherens junction and enhances vascular leakage, as well as suggest that anti-SCF/cKit therapy may hold promise as a potential therapy for the treatment of hyperpermeable vascular diseases.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales/metabolismo , Factor de Células Madre/metabolismo , Uniones Adherentes/metabolismo , Animales , Anticuerpos Neutralizantes/administración & dosificación , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/etiología , Retinopatía Diabética/metabolismo , Retinopatía Diabética/prevención & control , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Interferencia de ARN , Vasos Retinianos/metabolismo , Transducción de Señal , Factor de Células Madre/genética , Factor de Células Madre/inmunología , Factores de Tiempo , Transfección
9.
Eur J Immunol ; 43(7): 1698-701, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23719840

RESUMEN

Expression of the high-affinity receptor for IgE (FcεRI) provides mast cells with the ability to react in a proinflammatory manner to antigens (Ags). In particular, the immediate secretion of preformed mediators from secretory lysosomes (degranulation) is typical for FcεRI-mediated mast cell activation. In addition to the FcεRI, the stem cell factor receptor, KIT, is expressed at high levels on the surface of mast cells. KIT activation controls mast cell differentiation and survival in vivo and potently stimulates the chemotaxis of these cells. Although FcεRI and KIT initiate many of the same early signaling events in mast cells, FcεRI activation results in potent degranulation and a poor chemotactic response while KIT activation triggers very little degranulation and a strong chemotactic response. Novel data published in this issue of the European Journal of Immunology [Smrz et al. Eur. J. Immunol. 2013. 43: 1873-1882] demonstrate that actin de- and repolymerization, involved in both degranulation and chemotaxis, make all the difference: Pharmacological suppression of F-actin formation converts activated KIT into a strong degranulator. The possible implications for mast cell physiology and pathophysiology are discussed in this Commentary.


Asunto(s)
Actinas/metabolismo , Degranulación de la Célula/inmunología , Quimiotaxis de Leucocito/inmunología , Mastocitos/metabolismo , Factor de Células Madre/inmunología , Humanos
10.
Eur J Immunol ; 43(7): 1873-82, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23616175

RESUMEN

Following antigen/IgE-mediated aggregation of high affinity IgE-receptors (FcεRI), mast cells (MCs) degranulate and release inflammatory mediators leading to the induction of allergic reactions including anaphylaxis. Migration of MCs to resident tissues and sites of inflammation is regulated by tissue chemotactic factors such as stem cell factor (SCF (KIT ligand)). Despite inducing similar early signaling events to antigen, chemotactic factors, including SCF, produce minimal degranulation in the absence of other stimuli. We therefore investigated whether processes regulating MC chemotaxis are rate limiting for MC mediator release. To investigate this issue, we disrupted actin polymerization, a requirement for MC chemotaxis, with latrunculin B and cytochalasin B, then examined chemotaxis and mediator release in human (hu)MCs induced by antigen or SCF. As expected, such disruption minimally affected early signaling pathways, but attenuated SCF-induced human mast cell chemotaxis. In contrast, SCF, in the absence of other stimuli, induced substantial degranulation in a concentration-dependent manner following actin disassembly. It also moderately enhanced antigen-mediated human mast cell degranulation which was further enhanced in the presence of SCF. These observations suggest that processes regulating cell migration limit MC degranulation as a consequence of cytoskeletal reorganization.


Asunto(s)
Actinas/metabolismo , Degranulación de la Célula/inmunología , Quimiotaxis de Leucocito/inmunología , Mastocitos/metabolismo , Factor de Células Madre/inmunología , Actinas/inmunología , Degranulación de la Célula/efectos de los fármacos , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Citometría de Flujo , Humanos , Immunoblotting , Mastocitos/inmunología , Microscopía Confocal , Factor de Células Madre/farmacología
11.
Clin Exp Allergy ; 43(1): 50-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23278880

RESUMEN

BACKGROUND: Stem cell factor (SCF) is a growth factor that is involved in mast cell differentiation and proliferation. SCF primes human lung mast cells for enhanced responses to IgE-directed activation but is not generally recognized as a direct activator. SCF mediates its effects through c-kit. OBJECTIVE: The aim of this study was to reappraise the effects of SCF on human lung mast cells. METHODS: Mast cells were isolated from human lung. Mast cells were challenged with anti-IgE or SCF and the generation of histamine, cysteinyl-leukotrienes (cys-LTs) and prostaglandin D(2) (PGD(2) ) was assessed as was expression of the activation marker, CD63. The effects of c-kit inhibitors on mediator release were evaluated. RESULTS: Stem cell factor (10 ng/mL) alone was unable to induce mediator release but primed mast cells for enhanced IgE-dependent secretion. At higher concentrations (≥ 30 ng/mL), SCF had more varied effects and even when used alone was able to drive substantial levels of histamine release in about a third of all preparations studied. Similarly, SCF (100 ng/mL) alone was effective in stimulating the generation of cys-LTs in half of the preparations studied. SCF (100 ng/mL) was even more effective at stimulating PGD(2) generation as almost all preparations generated substantial quantities of the prostanoid. Mediator release induced by SCF was accompanied by the up-regulation of the activation marker, CD63. There was a positive correlation between the extent of mediator release induced by SCF and c-kit receptor expression. The effects of SCF on mediator release from mast cells were reversed by the c-kit inhibitor imatinib. CONCLUSIONS AND CLINICAL RELEVANCE: These data demonstrate that the responses of mast cells to SCF are heterogeneous. SCF can drive much greater levels of mediator release from mast cells, especially of PGD(2), than hitherto appreciated and this could be important in the context of respiratory diseases.


Asunto(s)
Mastocitos/inmunología , Factor de Células Madre/inmunología , Femenino , Citometría de Flujo , Liberación de Histamina/efectos de los fármacos , Liberación de Histamina/inmunología , Humanos , Inmunoglobulina E/inmunología , Inmunoglobulina E/metabolismo , Pulmón/citología , Pulmón/inmunología , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Prostaglandina D2/inmunología , Prostaglandina D2/metabolismo , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología
12.
J Immunol ; 187(6): 2974-81, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21859956

RESUMEN

Deficient thymopoiesis is a pivotal determinant of impaired immune competence following hematopoietic stem cell transplantation (HSCT). Stem cell factor (SCF) is essentially involved in early thymopoiesis. We evaluated whether SCF administration would improve recovery of thymopoiesis following HSCT in immunodeficient mice receiving: 1) bone marrow (BM) transplantation of congenic mice; or 2) human fetal liver HSCT in the human immune system mouse model. Following murine BM transplantation, SCF significantly enhanced thymopoiesis and peripheral T cell recovery in lymph nodes and spleen. SCF did not affect BM lymphoid progenitor recovery and/or expansion. Median thymic cellularity increased from 0.9 in PBS- to 266 × 10(4)/thymus in SCF-treated mice (p = 0.05). Following human HSCT in human immune system mice, higher thymic cellularity was observed in SCF-treated mice. Double-negative and early double-positive thymocyte subsets increased, but especially late double-positive, CD4 single-positive, and CD8 single-positive thymocyte subsets were significantly enhanced (p < 0.05). These results show that exogenous supply of SCF may significantly improve murine and human posttransplant thymopoiesis, for which the effect is probably exerted by directly promoting T cell development intrathymically rather than by enhanced entry of prethymically expanded lymphoid progenitors.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Trasplante de Células Madre Hematopoyéticas , Linfopoyesis/inmunología , Factor de Células Madre/inmunología , Timo/citología , Animales , Diferenciación Celular/inmunología , Separación Celular , Citometría de Flujo , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/cirugía , Ratones , Ratones Endogámicos C57BL , Linfocitos T/citología
13.
Cancer Immunol Immunother ; 61(3): 303-12, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21877248

RESUMEN

Mast cells may have either antitumor or tumor-promoting potential. Nevertheless, mast cells in tumor microenvironment have been found to promote tumor growth. So far the mechanisms underlying the modulation of mast cell function in tumor microenvironment remains to be fully elucidated. Here, we report that tumor-promoting potential of mast cells could be augmented by molecules released from damaged tumor cells through cooperative stimulation of stem cell factor (SCF) and ligand for Toll-like receptor 4 (TLR4). Co-simulation with SCF and TLR4 ligand inhibited mast cell degranulation, but efficiently induced the production and secretion of VEGF, PDGF, and IL-10. Although TLR4 ligand alone may induce IL-12 expression in mast cells, co-stimulation with SCF and TLR4 ligand induced the expression of IL-10, but not IL-12, in mast cells. The phosphorylation of GSK3ß was crucial for the effect of SCF and TLR4 ligand. In addition to inducing phosphorylation of GSK3ß at Ser9 through PI3K pathway, SCF and TLR4 ligand cooperated to induce phosphorylation of GSK3ß at Tyr216 by simultaneous activation of ERK and p38MAPK pathways. Both phospho-Ser9 and phospho-Tyr216 of GSK3ß were required for IL-10 expression induced by SCF/TLR4 ligand, whereas suppressive effect of SCF/TLR4 ligand on mast cell degranulation was related to phospho-Tyr216. Importantly, the effect of SCF and TLR4 ligand on mast cells could be abrogated by inhibiting phosphorylation of GSK3ß at Tyr216. These findings disclose the mechanisms underlying the modulation of mast cell function in tumor microenvironment, and suggest that inhibiting GSK3ß in mast cells will be beneficial to the treatment of cancer.


Asunto(s)
Mastocitos/inmunología , Neoplasias Experimentales/inmunología , Factor de Células Madre/inmunología , Receptor Toll-Like 4/inmunología , Animales , Western Blotting , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/fisiología , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas , Cromolin Sódico/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Ligandos , Lipopolisacáridos/farmacología , Mastocitos/efectos de los fármacos , Mastocitos/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología , Receptor Toll-Like 4/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
14.
J Immunol ; 184(11): 6114-23, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20427772

RESUMEN

Tissue-resident mast cells (MCs) are important in allergic diseases. In a mouse model of allergic airways inflammation, an increase in peribronchiolar MCs was associated with increased concentrations of the chemokine CCL2 in lung lavage. MC progenitors (MCps) arising in bone marrow (BM) are recruited to tissues by transendothelial migration, and we found that CCL2 is chemotactic for MCps in freshly isolated BM in vitro. Immature, but not mature, BM-derived MCs migrated in response to CCL2 when cultured in IL-3+stem cell factor (SCF) but not when cultured in IL-3 alone. However, the cells under both culture conditions expressed mRNA for CCR2, the receptor for CCL2, and bound the radiolabeled chemokine with similar affinities, highlighting SCF as a key mediator in coupling CCR2 to downstream events, culminating in chemotaxis. Immature BM-derived MCs from IL-3 +SCF cultures, when administered i.v., accumulated at skin sites injected with CCL2 in vivo. MCp recruitment to the allergen-sensitized/challenged lung was significantly reduced in CCR2(-/-) and CCL2(-/-) mouse strains. However, reconstitution studies of sublethally irradiated and BM-reconstituted mice indicated that BM cells and stromal elements could provide CCL2, whereas the CCR2 function resided with stromal elements rather than BM cells. These experiments revealed a new function of SCF in chemokine receptor coupling, but they suggest a complex role of the CCL2/CCR2 axis in recruiting MCps during pulmonary inflammation.


Asunto(s)
Quimiocina CCL2/inmunología , Quimiotaxis de Leucocito/inmunología , Mastocitos/inmunología , Receptores CCR2/inmunología , Alérgenos/inmunología , Alérgenos/farmacología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocina CCL2/metabolismo , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Mastocitos/citología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Receptores CCR2/metabolismo , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Células Madre/inmunología , Factor de Células Madre/metabolismo
15.
Gene Ther ; 18(11): 1078-86, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21544097

RESUMEN

Cytokines are required for γ-retroviral transduction of human CD34+ cells. However, cytokines may reduce engraftment of CD34+ cells and may not be necessary for their lentiviral transduction. We sought to optimize transduction and engraftment of human CD34+ cells using lentiviral vectors. Single 24 h transduction of human CD34+ cells with human immunodeficiency virus type 1 (HIV1)-based lentiviral vectors in media containing stem cell factor (SCF), FMS-like tyrosine kinase 3 (FLT3) ligand, thrombopoietin (each 100 ng ml⁻¹) and 10% fetal bovine serum was compared with various cytokine conditions during ex vivo culture and assayed using humanized xenograft mice for 6 months after transplantation. Serum-free media improved transduction efficiency of human CD34+ cells. Interleukin-3 (20 ng ml⁻¹) had little effect on transduction efficiency or engraftment. Threefold higher cytokine mixture (each 300 ng ml⁻¹) reduced engraftment of CD34+ cells. SCF alone (100 ng ml⁻¹) proved insufficient for maintaining engraftment ability and reduced transduction efficiency. Short-term prestimulation had little effect on transduction efficiency or engraftment, yet 24 h prestimulation showed higher transduction efficiency, higher gene expression levels and lower engraftment. In summary, 24 h prestimulation followed by single 24-h lentiviral transduction in serum-free media with SCF, FLT3 ligand and thrombopoietin yields high transduction efficiency to engrafting human CD34+ cells, and is applicable in human clinical gene therapy trials.


Asunto(s)
Antígenos CD34/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Interleucina-3/farmacología , Lentivirus/genética , Transducción Genética , Animales , Medio de Cultivo Libre de Suero , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Animales , Factor de Células Madre/inmunología , Trombopoyetina/inmunología , Trasplante Heterólogo , Tirosina Quinasa 3 Similar a fms/inmunología
16.
Clin Immunol ; 140(3): 276-83, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21550856

RESUMEN

We examined the capacities of bone marrow (BM) plasmacytoid dendritic cells (pDC) to differentiate into type B synoviocyte-like cells. BM aspiration samples were obtained from 24 rheumatoid arthritis (RA) patients and 19 osteoarthritis (OA) patients during joint operations from the iliac crest. CD34+ cells and pDC purified from BM mononuclear cells were cultured with or without SCF, GM-CSF, and TNF-α for 2-4 weeks. RA BM pDC as well as OA BM pDC comparably differentiated into fibroblast-like cells (FLC), expressing cadherin-11 and producing MMP-1, especially in the presence of TNF-α. Of note, depletion of BDCA4+ pDC from RA BM CD34+ cells significantly diminished their capacities to differentiate into FLC, which were restored by addition of BDCA4+cells in a dose-response manner. These results indicate that pDC is one of the progenitors of type B synoviocytes, suggesting that BM pDC might be involved in the pathogenesis of RA and OA.


Asunto(s)
Artritis Reumatoide/inmunología , Médula Ósea/inmunología , Osteoartritis/inmunología , Membrana Sinovial/inmunología , Antígenos CD34/inmunología , Antígenos de Superficie , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Cadherinas/biosíntesis , Cadherinas/inmunología , Diferenciación Celular/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Masculino , Metaloproteinasa 1 de la Matriz/biosíntesis , Persona de Mediana Edad , Factor de Células Madre/inmunología , Factor de Necrosis Tumoral alfa/inmunología
17.
Immunol Cell Biol ; 89(1): 149-59, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20498673

RESUMEN

In mast cells, crosslinking the high-affinity IgE receptor (FcɛRI) results in a dynamic reorganization of the actin cytoskeleton that is associated with membrane ruffling. Although the signaling involved in degranulation has been well described, it is less understood in morphological changes. In this study, we investigated the specific role of conventional protein kinase C (cPKC), a crucial signal for degranulation, in antigen-induced membrane ruffling of mast cells. In RBL-2H3 mast cells, antigen induced a long-lasting membrane ruffling, which was blocked with late-added Gö6976, a specific cPKC inhibitor, indicating that sustained activation of cPKC is required for maintaining the reaction. Immunofluorescence staining of endogenous PKCα/ß and real-time imaging of transfected green fluorescent protein-tagged PKCα/ß demonstrated that in response to antigen both PKCα and PKCßI quickly translocated to the plasma membrane and were colocalized with actin filaments at the ruffling sites. These reactions were blocked by expression of kinase-negative PKCßI, but not kinase-negative PKCα, and by treatment with a specific PKCß inhibitor, LY333531. The adhesion, spreading and membrane ruffling of mouse bone marrow-derived mast cells (BMMCs), which are mostly nonadhesive, were promoted by both antigen and thymeleatoxin. Treatment with Gö6976 abolished all these reactions. Antigen-mediated migration of BMMC was also sensitive to Gö6076 and LY333531. In addition, BMMC adhesion by and migration toward stem cell factor were shown to be dependent on cPKC. Thus, cPKC, at least PKCß subtype, may be critical for the dynamic morphological changes that lead to the migration of mast cells.


Asunto(s)
Mastocitos/citología , Mastocitos/enzimología , Proteína Quinasa C/metabolismo , Actinas/metabolismo , Animales , Antígenos/inmunología , Antígenos/metabolismo , Adhesión Celular/fisiología , Degranulación de la Célula/inmunología , Línea Celular , Membrana Celular/inmunología , Membrana Celular/metabolismo , Movimiento Celular/inmunología , Extensiones de la Superficie Celular/inmunología , Extensiones de la Superficie Celular/metabolismo , Fibronectinas/fisiología , Mastocitos/inmunología , Ratones , Proteína Quinasa C/inmunología , Transporte de Proteínas/inmunología , Conejos , Receptores de IgE/inmunología , Receptores de IgE/metabolismo , Transducción de Señal/inmunología , Factor de Células Madre/inmunología , Factor de Células Madre/metabolismo
18.
J Immunol ; 183(2): 803-13, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19570824

RESUMEN

NK cells induce apoptosis in target cells via the perforin-mediated delivery of granzyme molecules. Cytotoxic human NK cells can be generated by IL-15-mediated differentiation of CD34(+) cells in vitro and these cultures have been used extensively to analyze the development of the NK cell surface phenotype. We have used NK cell differentiation in vitro together with protease-deficient human NK cells to analyze the acquisition of the cytotoxic phenotype. Granzymes are synthesized as inactive zymogens and are proteolytically activated by the cysteine protease cathepsin C. Cathepsin C is also synthesized as a zymogen and activated by proteolysis. We show that human NK cells generated in vitro undergo granule exocytosis and induce the caspase cascade in target cells. IL-15 and stem cell factor (IL-15 plus SCF) induced the expression of the granzyme B and perforin genes and the activation of cathepsin C and granzyme B zymogens. Perforin activation is also mediated by a cysteine protease and IL-15 plus SCF-mediated differentiation was accompanied by perforin processing. However, cathepsin C-deficient human NK cells revealed that perforin processing could occur in the absence of cathepsin C activity. The combination of IL-15 plus SCF is therefore sufficient to coordinate the development of the NK cell surface phenotype with the expression and proteolytic activation of the cytotoxic machinery, reflecting the central role of IL-15 in NK cell development.


Asunto(s)
Diferenciación Celular , Citotoxicidad Inmunológica , Interleucina-15/fisiología , Células Asesinas Naturales/citología , Péptido Hidrolasas/fisiología , Factor de Células Madre/fisiología , Antígenos de Superficie , Caspasas/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Exocitosis , Granzimas , Humanos , Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Péptido Hidrolasas/inmunología , Perforina , Vesículas Secretoras , Factor de Células Madre/inmunología
19.
Res Vet Sci ; 134: 137-146, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33383491

RESUMEN

"Humanized" immunodeficient mice generated via the transplantation of CD34+ human hematopoietic stem cells (hHSC) are an important preclinical model system. The triple transgenic NOD.Cg-PrkdcscidIl2rgtm1Wjl Tg(CMV-IL3,CSF2,KITLG)1Eav/MloySzJ (NSGS) mouse line is increasingly used as recipient for CD34+ hHSC engraftment. NSGS mice combine the features of the highly immunodeficient NSG mice with transgenic expression of the human myeloid stimulatory cytokines GM-CSF, IL-3, and Kit ligand. While generating humanized NSGS (huNSGS) mice from two independent cohorts, we encountered a fatal macrophage activation syndrome (MAS)-like phenotype resulting from the transplantation of CD34+ hHSC. huNSGS mice exhibiting this phenotype declined clinically starting at approximately 10 weeks following CD34+ hHSC engraftment, with all mice requiring euthanasia by 16 weeks. Gross changes comprised small, irregular liver, splenomegaly, cardiomegaly, and generalized pallor. Hematological abnormalities included severe thrombocytopenia and anemia. Pathologically, huNSGS spontaneously developed a disseminated histiocytosis with infiltrates of activated macrophages and hemophagocytosis predominantly affecting the liver, spleen, bone marrow, and pancreas. The infiltrates were chimeric with a mixture of human and mouse macrophages. Immunohistochemistry suggested activation of the inflammasome in both human and murine macrophages. Active Epstein-Barr virus infection was not a feature. Although the affected mice exhibited robust chimerism of the spleen and bone marrow, the phenotype often developed in the face of low chimerism of the peripheral blood. Given the high penetrance and early lethality associated with the MAS-like phenotype here described, we urge caution when considering the use of huNSGS mice for the development of long-term studies.


Asunto(s)
Síndrome de Activación Macrofágica/patología , Macrófagos/microbiología , Animales , Antígenos CD34 , Proteína Quinasa Activada por ADN/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Herpesvirus Humano 4 , Histiocitosis/inmunología , Humanos , Subunidad gamma Común de Receptores de Interleucina/inmunología , Síndrome de Activación Macrofágica/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes/inmunología , Factor de Células Madre/inmunología
20.
Front Immunol ; 12: 757967, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34759931

RESUMEN

Innate lymphoid cells (ILCs) are the most recently described group of lymphoid subpopulations. These tissue-resident cells display a heterogeneity resembling that observed on different groups of T cells, hence their categorization as cytotoxic NK cells and helper ILCs type 1, 2 and 3. Each one of these groups is highly diverse and expresses different markers in a context-dependent manner. Type 2 innate lymphoid cells (ILC2s) are activated in response to helminth parasites and regulate the immune response. They are involved in the etiology of diseases associated with allergic responses as well as in the maintenance of tissue homeostasis. Markers associated with their identification differ depending on the tissue and model used, making the study and understanding of these cells a cumbersome task. This review compiles evidence for the heterogeneity of ILC2s as well as discussion and analyses of molecular markers associated with their identity, function, tissue-dependent expression, and how these markers contribute to the interaction of ILC2s with specific microenvironments to maintain homeostasis or respond to pathogenic challenges.


Asunto(s)
Antígenos de Diferenciación/análisis , Subgrupos Linfocitarios/inmunología , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/patología , Animales , Citocinas/metabolismo , Helmintiasis/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Homeostasis , Humanos , Inmunofenotipificación , Inflamación , Intestinos/inmunología , Pulmón/inmunología , Subgrupos Linfocitarios/química , Ratones , Nutrientes , Especificidad de Órganos , Proteínas Proto-Oncogénicas c-kit/inmunología , Receptores de Superficie Celular/inmunología , Piel/inmunología , Factor de Células Madre/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA