Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Appl Microbiol Biotechnol ; 106(24): 8121-8137, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36401641

RESUMO

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.


Assuntos
Anticorpos Monoclonais , Glicoproteínas , Hibridomas , Fator de Células-Tronco , Humanos , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Biotecnologia , Glicoproteínas/imunologia , Células HEK293 , Fator de Células-Tronco/análise , Fator de Células-Tronco/imunologia , Glicosilação , Ensaio de Imunoadsorção Enzimática , Western Blotting
2.
Front Immunol ; 12: 757967, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34759931

RESUMO

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.


Assuntos
Antígenos de Diferenciação/análise , Subpopulações de Linfócitos/imunologia , Tecido Adiposo Branco/imunologia , Tecido Adiposo Branco/patologia , Animais , Citocinas/metabolismo , Helmintíase/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Homeostase , Humanos , Imunofenotipagem , Inflamação , Intestinos/imunologia , Pulmão/imunologia , Subpopulações de Linfócitos/química , Camundongos , Nutrientes , Especificidade de Órgãos , Proteínas Proto-Oncogênicas c-kit/imunologia , Receptores de Superfície Celular/imunologia , Pele/imunologia , Fator de Células-Tronco/imunologia
3.
Res Vet Sci ; 134: 137-146, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33383491

RESUMO

"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.


Assuntos
Síndrome de Ativação Macrofágica/patologia , Macrófagos/microbiologia , Animais , Antígenos CD34 , Proteína Quinase Ativada por DNA/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Herpesvirus Humano 4 , Histiocitose/imunologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/imunologia , Síndrome de Ativação Macrofágica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Recombinantes/imunologia , Fator de Células-Tronco/imunologia
4.
Immunology ; 159(4): 441-449, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31957000

RESUMO

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.


Assuntos
Interleucina-4/farmacologia , Mastócitos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/genética , Receptores de Interleucina-4/genética , Fator de Transcrição STAT6/genética , Fator de Células-Tronco/farmacologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Hematopoese/imunologia , Interleucina-4/genética , Interleucina-4/imunologia , Mastócitos/citologia , Mastócitos/imunologia , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-kit/imunologia , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/imunologia , Receptores de Interleucina-3/genética , Receptores de Interleucina-3/imunologia , Receptores de Interleucina-4/imunologia , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/imunologia , Fator de Transcrição STAT6/imunologia , Transdução de Sinais , Fator de Células-Tronco/genética , Fator de Células-Tronco/imunologia , beta-Ciclodextrinas/farmacologia
5.
Mucosal Immunol ; 12(2): 445-456, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30617299

RESUMO

Stem cell factor (SCF) binds to the receptor c-Kit that is expressed on a number of myeloid and lymphoid cell populations, including Type 2 innate lymphoid cells (ILC2). However the importance of the SCF/c-Kit interaction in ILC2 has not been studied. Here we investigate the role of a specific SCF isoform, SCF248, in the allergic asthmatic response and SCF/c-Kit in ILC2 activation during chronic allergy. We observed that mice treated with a monoclonal antibody specific for SCF248 attenuated the development of chronic asthmatic disease by decreasing the number of mast cells, ILC2 and eosinophils, as well as reducing the accompanying pathogenic cytokine responses. These data were supported using SCFfl/fl-Col1-Cre-ERT mice and W/Wv mice that demonstrated the importance of the stem cell factor/c-Kit activation during chronic allergy and the accumulation of c-kit+ cells. Finally, these data demonstrate for the first time that SCF could activate ILC2 cells in vitro for the production of key allergic cytokines. Together these findings indicate that SCF is a critical cytokine involved in the activation of ILC2 that lead to more severe outcomes during chronic allergy and that the SCF248 isoform could be an important therapeutic target to control the disease progression.


Assuntos
Asma/imunologia , Pulmão/patologia , Linfócitos/imunologia , Fator de Células-Tronco/metabolismo , Alérgenos/imunologia , Animais , Células Cultivadas , Doença Crônica , Colágeno Tipo I/genética , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/genética , Fator de Células-Tronco/imunologia , Células Th2/imunologia
6.
J Appl Physiol (1985) ; 122(3): 675-682, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28082336

RESUMO

Circulating progenitor cells (CPCs) are a heterogeneous population of stem/progenitor cells in peripheral blood that includes hematopoietic stem and progenitor cells (HSPCs and HSCs), endothelial progenitor cells (EPCs), and mesenchymal stem cells (MSCs) that are involved in tissue repair and adaptation. CPC mobilization during exercise remains uncharacterized in young adults. The purpose of this study was to investigate the kinetics of CPC mobilization during and after submaximal treadmill running and their relationship to mobilization factors. Seven men [age = 25.3 ± 2.4 yr, body mass index = 23.5 ± 1.0 kg/m2, peak O2 uptake (V̇o2peak) = 60.9 ± 2.74 ml·kg-1·min-1] ran on a treadmill for 60 min at 70% V̇o2peak Blood sampling occurred before (Pre), during [20 min (20e), 40 min (40e), 60 min (60e)], and after exercise [15 min (15p), 60 min (60p), 120 min (120p)] for quantification of CPCs (CD34+), HSPCs (CD34+/CD45low), HSCs (CD34+/CD45low/CD38-), CD34+ MSCs (CD45-/CD34+/CD31-/CD105+), CD34- MSCs (CD45-/CD34-/CD31-/CD105+), and EPCs (CD45-/CD34+/CD31+) via flow cytometry. CPC concentration increased compared with Pre at 20e and 40e (2.7- and 2.4-fold, respectively, P < 0.05). HSPCs and HSCs increased at 20e compared with 60p (2.7- and 2.8-fold, respectively, P < 0.05), whereas EPCs and both MSC populations did not change. CXC chemokine ligand (CXCL) 12 (1.5-fold; P < 0.05) and stem cell factor (1.3-fold; P < 0.05) were increased at 40e and remained elevated postexercise. The peak increase in CPCs was positively correlated to concentration of endothelial cells during exercise with no relationship to CXCL12 and SCF. Our data show the kinetics of progenitor cell mobilization during exercise that could provide insight into cellular mediators of exercise-induced adaptations, and have implication for the use of exercise as an adjuvant therapy for CPC collection in hematopoietic stem cell transplant.NEW & NOTEWORTHY Using a comprehensive evaluation of circulating progenitor cells (CPCs), we show that CPC mobilization during exercise is related to tissue damage, and not plasma concentrations of CXC chemokine ligand 12 and stem cell factor. These data have implications for the use of exercise interventions as adjuvant therapy for CPC mobilization in the context of hematopoietic stem cell transplant and also support the role of mobilized progenitor cells as cellular mediators of systemic adaptations to exercise.


Assuntos
Quimiocina CXCL12/sangue , Exercício Físico/fisiologia , Esforço Físico/imunologia , Fator de Células-Tronco/sangue , Células-Tronco/citologia , Células-Tronco/imunologia , Adulto , Movimento Celular/imunologia , Quimiocina CXCL12/imunologia , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Cinética , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Fator de Células-Tronco/imunologia
7.
Arthritis Rheumatol ; 68(10): 2492-502, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27059145

RESUMO

OBJECTIVE: Antinuclear antibodies (ANAs) are detected in ∼18% of females, yet autoimmune disease develops in only 5-8%. Immunologic differences between ANA-positive healthy individuals and patients with systemic lupus erythematosus (SLE) may elucidate the regulatory mechanisms by which ANA-positive individuals avoid transition to clinical autoimmune disease. METHODS: Healthy individuals (n = 790) were screened for autoantibodies specific for 11 antigens associated with lupus, systemic sclerosis, and Sjögren's syndrome. From this screening, 31 European American ANA-positive healthy individuals were selected and demographically matched to ANA-negative controls and SLE patients. Serum cytokine profiles, leukocyte subset frequency, and reactivity were analyzed by multiplex assays, immunophenotyping, and phosphospecific flow cytometry. RESULTS: Of 790 individuals screened, 57 (7%) were ANA-positive. The majority of proinflammatory cytokines, including interferon-γ (IFNγ), tumor necrosis factor, interleukin-17 (IL-17), and granulocyte colony-stimulating factor, exhibited a stepwise increase in serum levels from ANA-negative controls to ANA-positive healthy individuals to SLE patients (P < 0.0001). IFNα, IFNß, IL-12p40, and stem cell factor/c-Kit ligand were increased in SLE patients only (P < 0.05). B lymphocyte stimulator (BlyS) was elevated in SLE patients but decreased in ANA-positive individuals (P < 0.001). Further, IL-1 receptor antagonist (IL-1Ra) was down-regulated in SLE patients only (P < 0.0001). ANA-positive individuals had increased frequencies of monocytes, memory B cells, and plasmablasts and increased levels of pSTAT-1 and pSTAT-3 following IFNα stimulation compared with ANA-negative controls (P < 0.05). CONCLUSION: ANA-positive healthy individuals exhibit dysregulation in multiple immune pathways yet differ from SLE patients by the absence of elevated IFNs, BLyS, IL-12p40, and stem cell factor/c-Kit ligand. Further, severely decreased levels of IL-1Ra in SLE patients compared with ANA-positive individuals may contribute to disease development. These results highlight the importance of IFN-related pathways and regulatory elements in SLE pathogenesis.


Assuntos
Anticorpos Antinucleares/imunologia , Autoimunidade/imunologia , Citocinas/imunologia , Voluntários Saudáveis , Leucócitos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Autoanticorpos/imunologia , Subpopulações de Linfócitos B/imunologia , Estudos de Casos e Controles , Proteína B de Centrômero/imunologia , DNA Topoisomerases Tipo I , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos/imunologia , Humanos , Imunofenotipagem , Interferon-alfa/imunologia , Interferon-alfa/farmacologia , Interferon beta/imunologia , Interferon gama/imunologia , Proteína Antagonista do Receptor de Interleucina 1/imunologia , Subunidade p40 da Interleucina-12/imunologia , Interleucina-17/imunologia , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Monócitos/imunologia , Análise Multivariada , Proteínas Nucleares/imunologia , Fosfoproteínas/efeitos dos fármacos , Plasmócitos/imunologia , Ribonucleoproteínas/imunologia , Proteínas Ribossômicas/imunologia , Fator de Transcrição STAT1/efeitos dos fármacos , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/imunologia , Fatores Sexuais , Fator de Células-Tronco/imunologia , Fator de Necrose Tumoral alfa/imunologia , Adulto Jovem
8.
Am J Reprod Immunol ; 75(4): 493-502, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26791471

RESUMO

PROBLEM: Uterine natural killer (uNK) cells play a significant role in successful human pregnancy. Having previously demonstrated uNK cell progenitors in human endometrium, we hypothesized that abnormal uNK cell maturation contributes to infertility in women with endometriosis. We aimed to characterize uNK cells at different developmental stages in women with and without endometriosis and to investigate possible mechanisms to explain any differences. METHOD OF STUDY: We characterized uNK cell development in women with and without endometriosis using flow cytometry, protein array and in vitro experiments. RESULTS: We found increased proportions of uNK cells at developmental stages 1 and 2 in endometrium from women with endometriosis (n = 36; mean = 21.2%) when compared with healthy fertile women (n = 9; mean = 7.0%). Protein array analysis revealed significantly lower levels of stem cell factor (SCF) in the eutopic endometrium of women with endometriosis when compared to healthy women. Addition of SCF to endometrial progenitor cells in vitro restored uNK cell maturation. CONCLUSION: We have shown that women with endometriosis have low levels of endometrial SCF, which we hypothesize contributes to abnormal maturation of local uNK cell populations. This defect may also compromise embryo implantation and hence contribute to endometriosis-associated infertility. SCF replacement may be a new therapeutic approach.


Assuntos
Endometriose/imunologia , Endométrio/imunologia , Infertilidade Feminina/imunologia , Células Matadoras Naturais/imunologia , Células Progenitoras Linfoides/imunologia , Fator de Células-Tronco/imunologia , Adulto , Endometriose/patologia , Endométrio/patologia , Feminino , Humanos , Infertilidade Feminina/patologia , Células Matadoras Naturais/patologia , Células Progenitoras Linfoides/patologia , Gravidez
9.
Cell Mol Immunol ; 13(1): 47-56, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25418470

RESUMO

Potential crosslinks between inflammation and leukaemia have been discussed for some time, but experimental evidence to support this dogma is scarce. In particular, it is important to understand the mechanisms responsible for potential upregulation of proto-oncogenic growth factor expressions by inflammatory mediators. Here, we investigated the ability of the highly inflammatory cytokine interleukin-1 beta (IL-1ß) to induce the production of stem cell factor (SCF), which is a major hematopoietic growth factor that controls the progression of acute myeloid leukaemia upon malignant transformation of haematopoietic myeloid cells. We found that human IL-1ß induced the expression/secretion of SCF in MCF-7 human epithelial breast cancer cells and that this process depended on the hypoxia-inducible factor 1 (HIF-1) transcription complex. We also demonstrated a crucial role of the phosphatidylinositol-3 kinase (PI-3K)/mammalian target of rapamycin (mTOR) pathway in IL-1ß-induced HIF-1α accumulation in MCF-7 cells. Importantly, mTOR was also found to play a role in IL-1ß-induced SCF production. Furthermore, a tendency for a positive correlation of IL-1ß and SCF levels in the plasma of healthy human donors was observed. Altogether, our results demonstrate that IL-1ß, which normally bridges innate and adaptive immunity, induces the production of the major haematopoietic/proleukaemic growth factor SCF through the PI-3K/mTOR pathway and the HIF-1 transcription complex. These findings strongly support a cross-talk between inflammation and acute myeloid leukaemia.


Assuntos
Regulação Leucêmica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Interleucina-1beta/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Fator de Células-Tronco/imunologia , Serina-Treonina Quinases TOR/imunologia , Imunidade Adaptativa , Hipóxia Celular , Linhagem Celular , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imunidade Inata , Interleucina-1beta/genética , Interleucina-1beta/farmacologia , Células MCF-7 , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Fosfatidilinositol 3-Quinases/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transdução de Sinais , Fator de Células-Tronco/genética , Serina-Treonina Quinases TOR/genética , Transcrição Gênica
10.
PLoS One ; 10(10): e0141485, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26505626

RESUMO

BACKGROUND: Stem cell factor (SCF) can stimulate hematopoietic stem cell (HSC) expansion; however, the specific structural region(s) of SCF protein that are critical for this function are still unknown. A novel monoclonal antibody (named 23C8) against recombinant human SCF (rhSCF) was previously found to inhibit the ability of rhSCF to enhance HSC expansion, making it possible to identify the relevant active region to HSC. METHODS: Eleven polypeptides were synthesized, which were designed to cover the full-length of rhSCF, with overlaps that are at least 3 amino acids long. ELISA was used to identify the polypeptide(s) that specifically react with the anti-SCF. The effects of the synthetic polypeptides on human HSC expansion, or on the ability of the full-length rhSCF to stimulate cell proliferation, were evaluated ex vivo. Total cell number was monitored using hemocytometer whereas CD34+ cell number was calculated based on the proportion determined via flow cytometry on day 6 of culture. RESULTS: Of all polypeptides analyzed, only one, named P0, corresponding to the SCF protein sequence at residues 39-56, was recognized by 23C8 mAb during ELISA. P0 stimulated the expansion of CD34+ cells derived from human umbilical cord blood (UCB). Addition of P0 increased the numbers of total mononucleated cells and CD34+ cells, by ~2 fold on day 6. P0 also showed partial competition against full-length rhSCF in the ex vivo cell expansion assay. CONCLUSION: Residues 39-56 of rhSCF comprise a critical functional region for its ability to enhance expansion of human UCB CD34+ cells. The peptide P0 is a potential candidate for further development as a synthetic substitute for rhSCF in laboratory and clinical applications.


Assuntos
Anticorpos Monoclonais/imunologia , Proliferação de Células , Sangue Fetal/imunologia , Fator de Células-Tronco/imunologia , Anticorpos Monoclonais/metabolismo , Antígenos CD34/imunologia , Ciclo Celular/genética , Sangue Fetal/metabolismo , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Humanos , Fator de Células-Tronco/genética
11.
Cell Metab ; 21(5): 678-91, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25955205

RESUMO

Obesity, insulin resistance, and related pathologies are associated with immune-mediated chronic inflammation. Kit mutant mice are protected from diet-induced obesity and associated co-morbidities, and this phenotype has previously been attributed to their lack of mast cells. We performed a comprehensive metabolic analysis of Kit-dependent Kit(W/Wv) and Kit-independent Cpa3(Cre/+) mast-cell-deficient mouse strains, employing diet-induced or genetic (Lep(Ob/Ob) background) models of obesity. Our results show that mast cell deficiency, in the absence of Kit mutations, plays no role in the regulation of weight gain or insulin resistance. Moreover, we provide evidence that the metabolic phenotype observed in Kit mutant mice, while independent of mast cells, is immune regulated. Our data underscore the value of definitive mast cell deficiency models to conclusively test the involvement of this enigmatic cell in immune-mediated pathologies and identify Kit as a key hematopoietic factor in the pathogenesis of metabolic syndrome.


Assuntos
Resistência à Insulina , Mastócitos/patologia , Obesidade/genética , Fator de Células-Tronco/genética , Animais , Deleção de Genes , Hematopoese , Masculino , Mastócitos/imunologia , Mastócitos/metabolismo , Síndrome Metabólica/genética , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/imunologia , Obesidade/patologia , Fator de Células-Tronco/imunologia , Transcriptoma
12.
Cell Prolif ; 48(3): 293-300, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25899394

RESUMO

OBJECTIVES: Stem cell factor (SCF) is essential in the haematopoietic stem cells (HSCs) niche, and is therefore used extensively in haematopoietic stem and progenitor cells (HSPCs) ex vivo expansion. However, in the literature, dose and schedule of SCF feeding varies widely. We previously proposed a novel SCF feeding regimen with proven effectiveness for HSPCs expansion; however, physiological function of expanded cells with this SCF feeding regimen required further research. MATERIALS AND METHODS: CD34(+) cells were cultured with or without SCF supplementation in serum-free medium for 10 days. Expanded cells were transplanted into sublethally irradiated non-obese diabetic/severe combined immune-deficient (NOD/SCID) mice. Engraftment and multilineage reconstitution of transplanted cells were determined. Also, clonogenic potential of engrafted cells was analysed. RESULTS: Cells, both cultured with and without SCF supplementation, successfully engrafted and reconstituted blood cell lineages in NOD/SCID mice. However, level of engraftment and multilineage reconstitution reduced when cells were expanded without SCF supplementation. Meanwhile, frequencies of colony-forming cells (CFCs) amongst bone marrow cells were higher in mice transplanted with CD34(+) cells expanded with SCF supplementation. CONCLUSIONS: Reconstitution capacity reduced when CD34(+) cells were expanded without SCF supplementation, though this feeding regimen did not have any effect on cell expansion. This finding suggested that SCF was essential for preserving NOD/SCID reconstitution capacity of ex vivo expanded CD34(+) cells.


Assuntos
Antígenos CD34/metabolismo , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Fator de Células-Tronco/metabolismo , Animais , Antígenos CD34/genética , Antígenos CD34/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Proliferação de Células , Feminino , Sangue Fetal/citologia , Sangue Fetal/imunologia , Sangue Fetal/metabolismo , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fator de Células-Tronco/genética , Fator de Células-Tronco/imunologia , Transplante Heterólogo , Irradiação Corporal Total
13.
J Immunol ; 192(12): 5471-5475, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24829419

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Regulação da Expressão Gênica/imunologia , Interleucina-17/imunologia , Subunidade p19 da Interleucina-23/imunologia , Pulmão/imunologia , Metaloproteinase 9 da Matriz/imunologia , Fator de Células-Tronco/imunologia , Animais , Membrana Celular/genética , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/genética , Interleucina-17/genética , Interleucina-17/metabolismo , Subunidade p19 da Interleucina-23/biossíntese , Subunidade p19 da Interleucina-23/genética , Pulmão/citologia , Pulmão/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Fosforilação/genética , Fosforilação/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Células Th1/citologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/citologia , Células Th17/imunologia , Células Th17/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 34(7): 1459-67, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24790137

RESUMO

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.


Assuntos
Permeabilidade Capilar , Células Endoteliais/metabolismo , Fator de Células-Tronco/metabolismo , Junções Aderentes/metabolismo , Animais , Anticorpos Neutralizantes/administração & dosagem , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/etiologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/prevenção & controle , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Injeções Intravítreas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Interferência de RNA , Vasos Retinianos/metabolismo , Transdução de Sinais , Fator de Células-Tronco/genética , Fator de Células-Tronco/imunologia , Fatores de Tempo , Transfecção
15.
J Immunol ; 192(10): 4859-66, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24733849

RESUMO

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.


Assuntos
Quimiotaxia/imunologia , Mastócitos/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-vav/imunologia , Fator de Células-Tronco/imunologia , Quinases da Família src/imunologia , Animais , Quimiotaxia/genética , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Mastócitos/citologia , Camundongos , Camundongos Knockout , Fosforilação/genética , Fosforilação/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-vav/genética , Fator de Células-Tronco/genética , Quinases da Família src/genética
16.
Immunol Lett ; 159(1-2): 11-4, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24534641

RESUMO

Mast cells were first identified by Paul Ehrlich in 1878, when he was still a medical student. Many fundamental aspects of mast cell ontogeny have been elucidated since Ehrlich's first identification. Demonstration of mast cell derivation from bone marrow precursors could be established in 1977 when Kitamura's group first showed reconstitution of mast cells in mast cell-deficient mice by the adaptive transfer of wild type bone marrow and indicated that these cells were of hematopoietic origin. It is now definitively established that development of mast cells in bone marrow occurs along the myeloid pathway. However, several aspects need further clarification. In particular, identification and chemical characterization of growth factors expressing mast cell differentiating properties and the relationship between mast cell and basophils developmental pathways.


Assuntos
Transferência Adotiva/história , Células da Medula Óssea/imunologia , Transplante de Medula Óssea/história , Linhagem da Célula/imunologia , Mastócitos/imunologia , Animais , Basófilos/citologia , Basófilos/efeitos dos fármacos , Basófilos/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular , Expressão Gênica , História do Século XIX , História do Século XX , Humanos , Interleucina-3/imunologia , Interleucina-3/farmacologia , Mastócitos/citologia , Mastócitos/efeitos dos fármacos , Camundongos , Fator de Células-Tronco/imunologia , Fator de Células-Tronco/farmacologia
17.
Clin Exp Allergy ; 43(1): 50-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23278880

RESUMO

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.


Assuntos
Mastócitos/imunologia , Fator de Células-Tronco/imunologia , Feminino , Citometria de Fluxo , Liberação de Histamina/efeitos dos fármacos , Liberação de Histamina/imunologia , Humanos , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Pulmão/citologia , Pulmão/imunologia , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Prostaglandina D2/imunologia , Prostaglandina D2/metabolismo , Fator de Células-Tronco/metabolismo , Fator de Células-Tronco/farmacologia
18.
J Immunol Methods ; 390(1-2): 52-62, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23357051

RESUMO

Activation of KIT, by its ligand, stem cell factor (SCF), results in the initiation of signal transduction pathways that influence mast cell survival and proliferation. Activating mutations in KIT have thus been linked to clonal MC proliferation associated with systemic mastocytosis. SCF also modulates MC function by inducing MC chemotaxis and by potentiating antigen (Ag)/IgE-mediated MC degranulation. Thus, mutations in KIT also have the potential to affect these processes in allergic and other mast cell-related diseases. Studies to determine how native and mutated KIT may modulate MC chemotaxis and activation have, however, been limited due to the lack of availability of a suitable functional MC line lacking native KIT which would allow transduction of KIT constructs. Here we describe a novel mouse MC line which allows the study of normal and mutated KIT constructs. These cells originated from a bone marrow-derived mouse MC culture out of which a rapidly dividing mast cell sub-population spontaneously arose. Over time, these cells lost KIT expression while continuing to express functional high affinity receptors for IgE (FcεRI). As a consequence, these cells degranulated in response to Ag/IgE but did not migrate nor show any evidence of potentiation of Ag/IgE degranulation in response to SCF. Retroviral transduction of the cells with a human (hu)KIT construct resulted in surface expression of huKIT which responded to huSCF by potentiation of Ag/IgE-induced degranulation and chemotaxis. This cell line thus presents a novel system to delineate how MC function is modulated by native and mutated KIT and for the identification of novel inhibitors of these processes.


Assuntos
Antígenos/imunologia , Degranulação Celular/imunologia , Mastócitos/imunologia , Proteínas Proto-Oncogênicas c-kit/imunologia , Animais , Benzamidas/imunologia , Benzamidas/farmacologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Cálcio/imunologia , Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Citometria de Fluxo , Humanos , Mesilato de Imatinib , Immunoblotting , Mastócitos/metabolismo , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Mutação/imunologia , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia , Piperazinas/imunologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/imunologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirimidinas/imunologia , Pirimidinas/farmacologia , Receptores de IgE/genética , Receptores de IgE/imunologia , Receptores de IgE/metabolismo , Fator de Células-Tronco/imunologia , Fator de Células-Tronco/farmacologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia , Serina-Treonina Quinases TOR/metabolismo
19.
J Hematol Oncol ; 5: 73, 2012 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-23228140

RESUMO

Stem cell factor (SCF) activates hematopoietic stem cell (HSC) self-renewal and is being used to stimulate the ex vivo expansion of HSCs. The mechanism by which SCF supports expansion of HSCs remains poorly understood. In cord blood ex vivo expansion assays, a newly produced anti-SCF monoclonal antibody (clone 23C8) was found to significantly inhibit the expansion of CD34+ cells. This antibody appears to bind directly to a part of SCF that is critical for biological activity toward expansion of CD34+ cells, which is located in the first 104 amino acids from the NH2-terminus.


Assuntos
Anticorpos Monoclonais/farmacologia , Sangue Fetal/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas Recombinantes/antagonistas & inibidores , Fator de Células-Tronco/antagonistas & inibidores , Antígenos CD34/metabolismo , Células Cultivadas , Técnicas de Cocultura , Sangue Fetal/citologia , Sangue Fetal/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Fator de Células-Tronco/imunologia , Fator de Células-Tronco/metabolismo
20.
Semin Hematol ; 49(2): 128-37, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22449623

RESUMO

Mast cells and eosinophils often travel in the same biologic circles. In non-clonal states, such as allergic and inflammatory conditions, cell-to-cell contact and the pleiotropic actions of multiple cytokines and chemokines, derived from local tissues or mast cells themselves, foster the co-recruitment of these cells to the same geographic cellular niche. While eosinophils and mast cells serve critical roles as part of the host immune response and in maintenance of normal homeostasis, these cell types can undergo neoplastic transformation due to the development of clonal molecular abnormalities that arise in early hematopoietic progenitors. The dysregulated tyrosine kinases, D816V KIT and FIP1L1-PDGFRA, are the prototypic oncogenic lesions resulting in systemic mastocytosis (SM) and chronic eosinophilic leukemia, respectively. We review the pathobiology of these myeloproliferative neoplasms (MPNs) with a focus on the relationship between mast cells and eosinophils, and discuss murine models, which further elucidate how the phenotype of these diseases can be influenced by stem cell factor (SCF) and expression of the potent eosinophilopoietic cytokine, interleukin-5 (IL-5). Therapy of SM and FIP1L1-PDGFRA-positive disease and the prognostic relevance of increased peripheral blood and tissue mast cells in hematolymphoid malignancies will also be addressed.


Assuntos
Eosinófilos/imunologia , Síndrome Hipereosinofílica/imunologia , Mastócitos/imunologia , Mastocitose/imunologia , Animais , Doença Crônica , Eosinófilos/patologia , Regulação Leucêmica da Expressão Gênica/genética , Regulação Leucêmica da Expressão Gênica/imunologia , Homeostase/genética , Homeostase/imunologia , Humanos , Síndrome Hipereosinofílica/genética , Síndrome Hipereosinofílica/patologia , Síndrome Hipereosinofílica/terapia , Interleucina-5/genética , Interleucina-5/imunologia , Mastócitos/patologia , Mastocitose/genética , Mastocitose/patologia , Mastocitose/terapia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/imunologia , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/imunologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/imunologia , Fator de Células-Tronco/imunologia , Fatores de Poliadenilação e Clivagem de mRNA/genética , Fatores de Poliadenilação e Clivagem de mRNA/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA