RESUMEN
Stem cell factor (SCF) is a dimeric molecule that exerts its biological functions by binding to and activating the receptor tyrosine kinase c-Kit. Activation of c-Kit leads to its autophosphorylation and initiation of signal transduction. Signaling proteins are recruited to activated c-Kit by certain interaction domains (e.g., SH2 and PTB) that specifically bind to phosphorylated tyrosine residues in the intracellular region of c-Kit. Activation of c-Kit signaling has been found to mediate cell survival, migration, and proliferation depending on the cell type. Signaling from c-Kit is crucial for normal hematopoiesis, pigmentation, fertility, gut movement, and some aspects of the nervous system. Deregulated c-Kit kinase activity has been found in a number of pathological conditions, including cancer and allergy. The observation that gain-of-function mutations in c-Kit can promote tumor formation and progression has stimulated the development of therapeutics agents targeting this receptor, e.g., the clinically used inhibitor imatinib mesylate. Also other clinically used multiselective kinase inhibitors, for instance, sorafenib and sunitinib, have c-Kit included in their range of targets. Furthermore, loss-of-function mutations in c-Kit have been observed and shown to give rise to a condition called piebaldism. This review provides a summary of our current knowledge regarding structural and functional aspects of c-Kit signaling both under normal and pathological conditions, as well as advances in the development of low-molecular-weight molecules inhibiting c-Kit function.
Asunto(s)
Proteínas Proto-Oncogénicas c-kit/fisiología , Transducción de Señal/fisiología , Factor de Células Madre/fisiología , Animales , Supervivencia Celular/fisiología , Humanos , Fosforilación/fisiologíaRESUMEN
Haematopoietic stem cell (HSC) niches provide an environment essential for life-long HSC function. Intense investigation of HSC niches both feed off and drive technology development to increase our capability to assay functionally defined cells with high resolution. A major driving force behind the desire to understand the basic biology of HSC niches is the clear implications for clinical therapies. Here, with particular emphasis on cell type-specific deletion of SCL and CXCL12, we focus on unresolved issues on HSC niches, framed around some very recent advances and novel discoveries on the extrinsic regulation of HSC maintenance. We also provide ideas for possible paths forward, some of which are clearly within reach while others will require both novel tools and vision.
Asunto(s)
Células Madre Hematopoyéticas/fisiología , Nicho de Células Madre , Animales , Movimiento Celular , Quimiocina CXCL12/fisiología , Células Madre Hematopoyéticas/citología , Humanos , Factor de Células Madre/fisiologíaRESUMEN
In the midgestation mouse embryo, hematopoietic cell clusters containing hematopoietic stem/progenitor cells arise in the aorta-gonad-mesonephros (AGM) region. We have previously reported that forced expression of the Sox17 transcription factor in CD45lowc-Kithigh AGM cells, which are the hematopoietic cellular component of the cell clusters, and subsequent coculture with OP9 stromal cells in the presence of three cytokines, stem cell factor (SCF), interleukin-3 (IL-3), and thrombopoietin (TPO), led to the formation and the maintenance of cell clusters with cells at an undifferentiated state in vitro. In this study, we investigated the role of each cytokine in the formation of hematopoietic cell clusters. We cultured Sox17-transduced AGM cells with each of the 7 possible combinations of the three cytokines. The size and the number of Sox17-transduced cell clusters in the presence of TPO, either alone or in combination, were comparable to that observed with the complete set of the three cytokines. Expression of TPO receptor, c-Mpl was almost ubiquitously expressed and maintained in Sox17-transduced hematopoietic cell clusters. In addition, the expression level of c-Mpl was highest in the CD45lowc-Kithigh cells among the Sox17-transduced cell clusters. Moreover, c-Mpl protein was highly expressed in the intra-aortic hematopoietic cell clusters in comparison with endothelial cells of dorsal aorta. Finally, stimulation of the endothelial cells prepared from the AGM region by TPO induced the production of hematopoietic cells. These results suggest that TPO contributes to the formation and the maintenance of hematopoietic cell clusters in the AGM region.
Asunto(s)
Aorta/citología , Gónadas/citología , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Mesonefro/citología , Trombopoyetina/fisiología , Animales , Aorta/embriología , Aorta/metabolismo , Células Cultivadas , Gónadas/embriología , Gónadas/metabolismo , Interleucina-3/fisiología , Mesonefro/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Receptores de Trombopoyetina/metabolismo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transducción de Señal , Factor de Células Madre/fisiología , Transducción GenéticaRESUMEN
The understanding of mast cell (MC) differentiation is derived mainly from in vitro studies of different stages of stem and progenitor cells. The hematopoietic lineage development of human MCs is unique compared to other myeloid-derived cells. Human MCs originate from CD34(+)/CD117(+)/CD13(+)multipotent hematopoietic progenitors, which undergo transendothelial recruitment into peripheral tissues, where they complete differentiation. Stem cell factor (SCF) is a major chemotactic factor for MCs and their progenitors. SCF also elicits cell-cell and cell-substratum adhesion, facilitates the proliferation, and sustains the survival, differentiation, and maturation, of MCs. Because MC maturation is influenced by local microenvironmental factors, different MC phenotypes can develop in different tissues and organs.
Asunto(s)
Diferenciación Celular , Mastocitos/fisiología , Animales , Biología Celular/historia , Historia del Siglo XIX , Historia del Siglo XX , Humanos , Factor de Células Madre/fisiologíaRESUMEN
In the present study we examined the effects of stem cell factor (SCF; 50 vs 100ngmL-1) alone or in combination with epidermal growth factor (EGF; 100ngmL-1) on: (1) the in vitro viability and growth of cat follicles within ovarian cortices; (2) phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK) phosphorylation; and (3) c-kit and FSH receptor (FSHr) mRNA expression. At 100ngmL-1, SCF increased (P≤0.05) the percentage and size of secondary follicles after 14 days of in vitro culture and sustained AKT phosphorylation after 3 days incubation. EGF suppressed this beneficial effect and reduced (P≤0.05) the percentage of structurally normal follicles and FSHr expression when combined with 100ngmL-1 SCF. Expression of c-kit mRNA was higher (P≤0.05) in the presence of 100ngmL-1 SCF compared with fresh follicles and cohorts cultured under other conditions. A c-kit inhibitor suppressed follicle growth and reduced AKT phosphorylation. Collectively, the results demonstrate that SCF promotes cat follicle development by upregulating c-kit mRNA expression and AKT phosphorylation. EGF suppresses the stimulating effect of SCF, leading to downregulation of FSHr expression.
Asunto(s)
Gatos/genética , Gatos/fisiología , Folículo Ovárico/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Factor de Células Madre/fisiología , Animales , Factor de Crecimiento Epidérmico/administración & dosificación , Factor de Crecimiento Epidérmico/fisiología , Femenino , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de HFE/genética , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/administración & dosificación , Regulación hacia Arriba/efectos de los fármacosRESUMEN
BACKGROUND/PURPOSE: Varicocele (VC) is considered by the World Health Organization as the main cause of male infertility. Studies have shown that VC can affect spermatogenesis and then result in male infertility. But the exact mechanism by which VC affects spermatogenesis is still unclear. Stem cell factor (SCF) and c-KIT receptor are crucial molecules during spermatogenesis in testis. This study aims to investigate whether SCF/c-KIT signaling is involved in the pathophysiology of VC on spermatogenesis. METHODS: Rat models of VC were built (n = 13), and sham-operated rats were used as controls (n = 8). The seminiferous tubules of the testis were observed with hematoxylin and eosin staining, expression of SCF was analyzed via enzyme-linked immunosorbent assay and Western blot, and expression of c-KIT was assessed with Western blot and immunofluorescence. RESULTS: Compared with controls, the seminiferous epithelium was disorganized and had significantly fewer cells in the testes of rats with VC. Expression of SCF increased in testes of VC rats, while expression of c-KIT was decreased. CONCLUSION: These results suggest that sperm counts in seminiferous epithelium are affected by VC, and the SCF/c-KIT system is aberrantly expressed in VC testis, which could be involved in male infertility caused by VC.
Asunto(s)
Proteínas Proto-Oncogénicas c-kit/fisiología , Factor de Células Madre/fisiología , Testículo/metabolismo , Varicocele/metabolismo , Animales , Masculino , Proteínas Proto-Oncogénicas c-kit/análisis , Ratas , Ratas Sprague-Dawley , Túbulos Seminíferos/patología , Recuento de Espermatozoides , Espermatogénesis , Factor de Células Madre/análisisRESUMEN
Adult stem cells are crucial for growth, homeostasis and repair of adult animals. The melanocyte stem cell (MSC) and melanocyte regeneration is an attractive model for studying regulation of adult stem cells. The process of melanocyte regeneration can be divided into establishment of the MSC, recruitment of the MSC to produce committed daughter cells, and the proliferation, differentiation and survival of these daughter cells. Reduction of Kit signaling results in dose-dependent reduction of melanocytes during larval regeneration. Here, we use clonal analysis techniques to develop assays to distinguish roles for these processes during zebrafish larval melanocyte regeneration. We use these clonal assays to investigate which processes are affected by the reduction in Kit signaling. We show that the regeneration defect in kita mutants is not due to defects in MSC recruitment or in the proliferation, differentiation or survival of the daughter cells, but is instead due to a defect in stem cell establishment. Our analysis suggests that the kit MSC establishment defect results from inappropriate differentiation of the MSC lineage.
Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/fisiología , Melanocitos/fisiología , Proteínas Proto-Oncogénicas c-kit/fisiología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Proliferación Celular , Supervivencia Celular/genética , Evolución Clonal/genética , Embrión no Mamífero , Células Madre Embrionarias/metabolismo , Dosificación de Gen/fisiología , Melanocitos/metabolismo , Modelos Biológicos , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regeneración/genética , Regeneración/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Factor de Células Madre/fisiología , Pez Cebra/genética , Pez Cebra/fisiologíaRESUMEN
Jak2, a member of the Janus kinase family of nonreceptor protein tyrosine kinases, is activated in response to a variety of cytokines, and functions in survival and proliferation of cells. An activating JAK2V617F mutation has been found in most patients with myeloproliferative neoplasms, and patients treated with Jak2 inhibitors show significant hematopoietic toxicities. However, the role of Jak2 in adult hematopoietic stem cells (HSCs) has not been clearly elucidated. Using a conditional Jak2 knockout allele, we have found that Jak2 deletion results in rapid loss of HSCs/progenitors leading to bone marrow failure and early lethality in adult mice. Jak2 deficiency causes marked impairment in HSC function, and the mutant HSCs are severely defective in reconstituting hematopoiesis in recipient animals. Jak2 deficiency also causes significant apoptosis and loss of quiescence in HSC-enriched LSK (Lin(-)Sca-1(+)c-Kit(+)) cells. Jak2-deficient LSK cells exhibit elevated reactive oxygen species levels and enhanced p38 MAPK activation. Mutant LSK cells also show defective Stat5, Erk, and Akt activation in response to thrombopoietin and stem cell factor. Gene expression analysis reveals significant downregulation of genes related to HSC quiescence and self-renewal in Jak2-deficient LSK cells. These data suggest that Jak2 plays a critical role in the maintenance and function of adult HSCs.
Asunto(s)
Células Madre Adultas/enzimología , Células Madre Hematopoyéticas/enzimología , Janus Quinasa 2/fisiología , Células Madre Adultas/fisiología , Anemia Aplásica , Animales , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Proliferación Celular , Supervivencia Celular , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Hemoglobinuria Paroxística/enzimología , Ratones Endogámicos C57BL , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo , Factor de Células Madre/fisiología , Trombopoyetina/fisiologíaRESUMEN
Hematopoietic stem cell (HSC) self-renewal is tightly controlled by cytokines and other signals in the microenvironment. While stem cell factor (SCF) is an early acting cytokine that activates the receptor tyrosine kinase KIT and promotes HSC maintenance, how SCF/KIT signaling is regulated in HSCs is poorly understood. The protein tyrosine phosphatase 4A (PTP4A) family (aka PRL [phosphatase of regenerating liver] phosphatases), consisting of PTP4A1/PRL1, PTP4A2/PRL2, and PTP4A3/PRL3, represents an intriguing group of phosphatases implicated in cell proliferation and tumorigenesis. However, the role of PTP4A in hematopoiesis remains elusive. To define the role of PTP4A in hematopoiesis, we analyzed HSC behavior in Ptp4a2 (Prl2) deficient mice. We found that Ptp4a2 deficiency impairs HSC self-renewal as revealed by serial bone marrow transplantation assays. Moreover, we observed that Ptp4a2 null hematopoietic stem and progenitor cells (HSPCs) are more quiescent and show reduced activation of the AKT and ERK signaling. Importantly, we discovered that the ability of PTP4A2 to enhance HSPC proliferation and activation of AKT and ERK signaling depends on its phosphatase activity. Furthermore, we found that PTP4A2 is important for SCF-mediated HSPC proliferation and loss of Ptp4a2 decreased the ability of oncogenic KIT/D814V mutant in promoting hematopoietic progenitor cell proliferation. Thus, PTP4A2 plays critical roles in regulating HSC self-renewal and mediating SCF/KIT signaling.
Asunto(s)
Proliferación Celular , Células Madre Hematopoyéticas/fisiología , Proteínas Inmediatas-Precoces/genética , Proteínas Tirosina Fosfatasas/genética , Animales , Células Cultivadas , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Proteínas Inmediatas-Precoces/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/fisiología , Transducción de Señal , Factor de Células Madre/fisiologíaRESUMEN
To investigate the development of follicles and incidence of apoptosis in vitrified cultured mouse ovaries in the presence and absence of Kit ligand, 1-week-old mouse ovaries were cultured in the presence or absence of Kit ligand for 7 days. Development and function of ovarian follicles was evaluated by histology and hormonal analysis. Apoptosis assessment was conducted by analysis of DNA laddering, TdT (terminal deoxynucleotidyl transferase)-mediated dUDP nick-end-labelling and caspase-3/7 activity. The proportion of preantral follicles and the level of 17-ß oestradiol, progesterone and dehydroepiandrosterone were increased in all cultured groups, and it was significantly higher in Kit ligand treated groups than in the control (P < 0.001). The number of apoptotic signals in both vitrified samples is significantly higher than in the non-vitrified control (P < 0.01), and these signals are significantly lower in both Kit ligand treated groups than in non-Kit ligand treated groups (P < 0.001). The level of caspase-3/7 activity was higher in vitrified cultured ovaries than non-vitrified group (P < 0.01). Kit ligand was shown to improve in-vitro development of follicles, and also acted as an anti-apoptotic factor in vitrified ovaries. The developmental potential of follicles in vitrified groups was lower than that in fresh ovaries.
Asunto(s)
Apoptosis/fisiología , Ovario/citología , Factor de Células Madre/fisiología , Vitrificación , Animales , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Células Cultivadas , Femenino , Hormonas Esteroides Gonadales/metabolismo , Ratones , Folículo Ovárico , Ovario/enzimología , Ovario/metabolismoRESUMEN
The receptor for stem cell factor (SCF) is expressed on mast cells and hematopoietic progenitors. SCF-induced signaling pathways remain incompletely defined. In this study, we identified calcineurin and regulator of calcineurin 1 (Rcan1) as novel components in SCF signaling. Calcineurin activity was induced in SCF-stimulated primary mouse and human mast cells. NFAT was activated by SCF in bone marrow-derived mast cells (BMMCs) and mouse bone marrow cells, which contain hematopoietic progenitors. SCF-mediated activation also induced expression of Rcan1 in BMMCs. Rcan1-deficient BMMCs showed increased calcineurin activity and enhanced transcriptional activity of NF-κB and NFAT, resulting in increased IL-6 and TNF production following SCF stimulation. These results suggest that Rcan1 suppresses SCF-induced activation of calcineurin and NF-κB. We further demonstrated that SCF-induced Rcan1 expression is dependent on the transcription factor early growth response 1 (Egr1). Interestingly, SCF-induced Egr1 was also suppressed by Rcan1, suggesting a negative regulatory loop between Egr1 and Rcan1. Together, our findings revealed that calcineurin contributes to SCF-induced signaling, leading to NFAT activation, which, together with NF-κB and Egr1, is suppressed by Rcan1. Considering the wide range of biological functions of SCF, these novel regulatory mechanisms in SCF signaling may have broad implications.
Asunto(s)
Calcineurina/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Mastocitos/metabolismo , Proteínas Musculares/fisiología , Factor de Células Madre/fisiología , Animales , Células de la Médula Ósea , Inhibidores de la Calcineurina , Proteínas de Unión al Calcio , Células Cultivadas , Proteínas de Unión al ADN , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Retroalimentación Fisiológica , Sangre Fetal/citología , Regulación de la Expresión Génica , Humanos , Inmunosupresores/farmacología , Interleucina-6/biosíntesis , Interleucina-6/genética , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Mastocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Factores de Transcripción NFATC/metabolismo , Proteínas Proto-Oncogénicas c-kit/fisiología , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Células Madre/farmacología , Transcripción Genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genéticaRESUMEN
PURPOSE: Foxo3 protein is required in the oocyte nucleus for the maintenance of primordial follicles in a dormant state. PI3K/AKT-dependent phosphorylation of Foxo3 leads to its relocalization to the cytoplasm and subsequent follicular activation. However, the nature of the upstream signals controlling Foxo3 activity and subcellular localization remains unknown. We aimed to study the in vitro effects of Kit ligand (stem cell factor) on the subcellular localization of Foxo3 in primordial follicles within the postnatal mouse ovary. METHODS: This was an in vitro study using explants of intact neonatal mouse ovaries. The study was performed in laboratory animal facility and basic science research laboratory at a University Hospital. The animals used for this study were FVB mice. Neonatal FVB mice ovaries at postnatal day 7 (PD7) were harvested and incubated in culture medium (DMEM) at 37 °C and 5 % CO(2) for 60-90 min with (n = 3) or without (n = 3) Kit ligand at 150 ng/mL (8 nM). Similar experimental conditions were used to establish a dose-response curve for the effects of Kit ligand and assess the effects of imatinib (small molecule inhibitor of the Kit receptor). Immunofluorescence was used to identify the subcellular location of Foxo3 in oocytes. Proportions of cytoplasmic versus nuclear Foxo3 in primordial follicles were determined. RESULTS: Kit ligand treatment increased the cytoplasmic localization of Foxo3 from 40 % in the untreated ovaries to 74 % in the treated group (p = 0.007 in paired samples and p = 0.03 in unpaired samples). Furthermore, this effect was reversible with imatinib (p = 0.005). A dose-response curve for Kit ligand treatment showed that maximum effect was seen at 150 ng/mL. CONCLUSION: Kit ligand treatment in vitro increases the proportion of cytoplasmic Foxo3 in primordial follicles at PD7, lending support to the idea that Kit receptor/ligand controls Foxo3 activity in the context of primordial follicle activation.
Asunto(s)
Factores de Transcripción Forkhead/fisiología , Ovario/metabolismo , Factor de Células Madre/fisiología , Animales , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/metabolismo , Mesilato de Imatinib/farmacología , Técnicas In Vitro , Ratones , Oocitos/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Factor de Células Madre/metabolismoRESUMEN
Mast cell (MC) accumulation has been demonstrated in the lungs of idiopathic pulmonary fibrosis (IPF) patients. Mediators released from MCs may regulate tissue remodeling processes, thereby contributing to IPF pathogenesis. We investigated the role of MC-fibroblast interaction in the progression of lung fibrosis. Increased numbers of activated MCs, in close proximity to fibroblast foci and alveolar type II cells, were observed in IPF lungs. Correspondingly elevated tryptase levels were detected in IPF lung tissue samples. Coculture of human lung MCs with human lung fibroblasts (HLFs) induced MC activation, as evinced by tryptase release, and stimulated HLF proliferation; IPF HLFs exhibited a significantly higher growth rate, compared with control. Tryptase stimulated HLF growth in a PAR-2/PKC-α/Raf-1/p44/42-dependent manner and potentiated extracellular matrix production, but independent of PKC-α, Raf-1, and p44/42 activities. Proproliferative properties of tryptase were attenuated by knockdown or pharmacological inhibition of PAR-2, PKC-α, Raf-1, or p44/42. Expression of transmembrane SCF, but not soluble SCF, was elevated in IPF lung tissue and in fibroblasts isolated from IPF lungs. Coculture of IPF HLFs with MCs enhanced MC survival and proliferation. These effects were cell-contact dependent and could be inhibited by application of anti-SCF antibody or CD117 inhibitor. Thus, fibroblasts and MCs appear to work in concert to perpetuate fibrotic processes and so contribute to lung fibrosis progression.
Asunto(s)
Fibroblastos/fisiología , Mastocitos/fisiología , Fibrosis Pulmonar/patología , Comunicación Celular/fisiología , Recuento de Células , Degranulación de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/patología , Mastocitos/metabolismo , Proteínas Quinasas/fisiología , Fibrosis Pulmonar/metabolismo , Receptor PAR-2/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Células Madre/fisiología , Triptasas/farmacología , Triptasas/fisiologíaRESUMEN
Mast cells, activated by Ag via FcεRI, release an array of proinflammatory mediators that contribute to allergic disorders, such as asthma and anaphylaxis. The KIT ligand, stem cell factor (SCF), is critical for mast cell expansion, differentiation, and survival, and under acute conditions, it enhances mast cell activation. However, extended SCF exposure in vivo conversely protects against fatal Ag-mediated anaphylaxis. In investigating this dichotomy, we identified a novel mode of regulation of the mast cell activation phenotype through SCF-mediated programming. We found that mouse bone marrow-derived mast cells chronically exposed to SCF displayed a marked attenuation of FcεRI-mediated degranulation and cytokine production. The hyporesponsive phenotype was not a consequence of altered signals regulating calcium flux or protein kinase C, but of ineffective cytoskeletal reorganization with evidence implicating a downregulation of expression of the Src kinase Hck. Collectively, these findings demonstrate a major role for SCF in the homeostatic control of mast cell activation with potential relevance to mast cell-driven disease and the development of novel approaches for the treatment of allergic disorders.
Asunto(s)
Mastocitos/inmunología , Mastocitos/metabolismo , Factor de Células Madre/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Degranulación de la Célula/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Homeostasis/inmunología , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Hipersensibilidad/patología , Inmunofenotipificación , Mastocitos/citología , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIHRESUMEN
BACKGROUND: In recent years, increasing attention has been focused on the skin hypopigmentation that develops after the initiation of imatinib mesylate therapy in patients with chronic myeloid leukaemia (CML). AIM: To understand the underlying mechanism of this hypopigmentation effect, and to explore the possibility of using imatinib in the treatment of pigmentation disorders. METHODS: We examined the effects of imatinib on the proliferation, apoptosis, melanin content and melanogenic activity of human primary epidermal melanocytes. The responsible molecular events were also investigated in a mechanism study. RESULTS: We found that imatinib led to a dramatic decrease in total melanin content in cultured melanocytes, by affecting melanocyte number and/or melanogenesis in a dose-dependent manner. This inhibition of melanogenesis was due to suppressed expression of tyrosinase and microphthalmia-associated transcription factor (MiTF). Furthermore, stem cell factor (SCF)-stimulated c-Kit activation and melanocyte proliferation were completely abrogated by imatinib. CONCLUSIONS: Inactivation of c-Kit signalling by imatinib has inhibitory effects on melanocyte survival, proliferation and melanogenesis, which explains the clinical hypopigmentation seen in patients with CML. These results also support using imatinib as a clinical depigmentation agent when dosage being carefully determined.
Asunto(s)
Benzamidas/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Melanocitos/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Epidérmicas , Fibroblastos/efectos de los fármacos , Humanos , Mesilato de Imatinib , Melaninas/metabolismo , Melanocitos/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Modelos Biológicos , Monofenol Monooxigenasa/metabolismo , Proteínas Proto-Oncogénicas c-kit/fisiología , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/fisiología , Células Tumorales CultivadasRESUMEN
Under homeostatic conditions, skin DCs migrate to regional LNs transporting self-antigens (self-Ags). The transport of self-Ags is considered to be critical for maintaining peripheral tolerance. Although the chemokine receptor CCR7 potently induces the migration of skin DCs to regional LNs, Ccr7(-/-) (Ccr7-KO) mice do not show skin auto-immune diseases. To resolve this inconsistency, we examined Ccr7-KO epidermis- or dermis-hyperpigmented transgenic (Tg) mice, in which the transport of skin self-Ags is traceable by melanin granules (MGs). Under CCR7-deficient conditions, the transport of epidermal MGs to regional LNs was impaired at 7 weeks of age. However, epidermal MGs could be transported when they had accumulated in the dermis. Ccr7-KO-dermis-pigmented Tg mice confirmed the presence of CCR7-independent transport from the dermis. Compared with WT-dermis-pigmented Tg mice, the amount of transported melanin and number of MG-laden CD11c(+) cells were both approximately 40% of the WT levels, while the number of MG-laden CD205(+) or CD207(+) cells decreased to about 10% in skin regional LNs of Ccr7-KO-dermis-pigmented Tg mice. Cell sorting highlighted the involvement of CD11c(+) cells in the CCR7-independent transport. Here, we show that CCR7-independent transport of skin self-Ags occurs in the dermis. This system might contribute to the continuous transport of self-Ags, and maintain peripheral tolerance.
Asunto(s)
Autoantígenos/metabolismo , Dermis/inmunología , Receptores CCR7/fisiología , Animales , Transporte Biológico , Antígeno CD11c/análisis , Antígeno CD11c/fisiología , Tolerancia Inmunológica , Ganglios Linfáticos/inmunología , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Células Madre/fisiologíaRESUMEN
Morphogenesis, as illustrated by melanocyte migration and homing to the skin, requires cadherin adhesion, integrin-dependent migration and Kit-ligand growth factor signaling. However, it is not known how Kit ligand regulates integrin or cadherin-dependent intraepidermal melanocyte behavior. To answer this question, we developed specific 2-dimensional (2D) and 3D culture systems analyzing how soluble or immobilized Kit-ligand-regulated melanocyte migration on vitronectin and laminin, or within a monolayer of kidney epithelial cells. In a 2D system, soluble Kit ligand stimulated integrin-dependent melanoblast migration and chemotaxis and accelerated integrin turnover. In contrast, immobilized, but not soluble, Kit ligand, enhanced integrin-dependent melanocyte spreading on suboptimal laminin concentrations. In 3D, membrane-bound Kit ligand induced intraepithelial melanocyte proliferation, survival, and tight adhesion to epithelial cells, while cleavable Kit ligand was less effective. In contrast, melanocyte motility was independent of membrane-bound Kit ligand, but increased in the presence of the cleavable Kit-ligand isoform. Transmembrane-dimerization or basolateral-targeting mutants of Kit ligand altered intraepithelial melanocyte localization, survival, and adhesion to epithelial cells. These data and the identification of c-kit/Kit-ligand clusters at cell contacts suggest that membrane-bound Kit ligand captures cell surface-expressed c-kit, providing mechanical anchoring and survival signaling within intraepithelial niches, and thereby defining a new mechanism for melanocyte homeostasis and requirement for environmental niches.
Asunto(s)
Adhesión Celular/fisiología , Supervivencia Celular/fisiología , Células Epiteliales/citología , Melanocitos/citología , Factor de Células Madre/fisiología , Secuencia de Aminoácidos , Animales , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Perros , Citometría de Flujo , Células de Riñón Canino Madin Darby , Ratones , Datos de Secuencia MolecularRESUMEN
The c-kit-encoded transmembrane tyrosine kinase receptor for stem cell factor (Kit/SCF-R) is required for normal haematopoiesis, melanogenesis and gametogenesis. However, the roles of individual Kit/SCF-R-induced signalling pathways in the control of developmental processes in the intact animal are completely unknown. To examine the function of SCF-induced phosphatidylinositol (PI) 3'-kinase activation in vivo, we employed the Cre-loxP system to mutate the codon for Tyr719, the PI 3'-kinase binding site in Kit/SCF-R, to Phe in the genome of mice by homologous recombination. Homozygous (Y719F/Y719F) mutant mice are viable. The mutation completely disrupted PI 3'-kinase binding to Kit/SCF-R and reduced SCF-induced PI 3'-kinase-dependent activation of Akt by 90%. The mutation induced a gender- and tissue-specific defect. Although there are no haematopoietic or pigmentation defects in homozygous mutant mice, males are sterile due to a block in spermatogenesis, with initially decreased proliferation and subsequent extensive apoptosis occurring at the spermatogonial stem-cell level. In contrast, female homozygotes are fully fertile. This is the first report so far demonstrating the role of an individual signalling pathway downstream of Kit/SCF-R in the intact animal. It provides the first in vivo model for male sterility caused by a discrete signalling pathway defect affecting early germ cells.
Asunto(s)
Fertilidad/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Sustitución de Aminoácidos , Animales , Apoptosis , Codón , Desarrollo Embrionario y Fetal , Activación Enzimática , Exones , Femenino , Biblioteca Genómica , Heterocigoto , Homocigoto , Intrones , Masculino , Ratones , Ratones Mutantes , Mutagénesis Sitio-Dirigida , Proteínas Proto-Oncogénicas c-kit/química , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/farmacología , Factor de Células Madre/fisiologíaRESUMEN
Mast cells (MCs) are sentinel cells which represent an important part of the first line of defense of the immune system. MCs highly express receptors for danger-associated molecular patterns (DAMPs) such as the IL-33R and P2X7, making MCs to potentially effective sensors for IL-33 and adenosine-triphosphate (ATP), two alarmins which are released upon necrosis-induced cell damage in peripheral tissues. Besides receptors for alarmins, MCs also express the stem cell factor (SCF) receptor c-Kit, which typically mediates MC differentiation, proliferation and survival. By using bone marrow-derived MCs (BMMCs), ELISA and flow cytometry experiments, as well as p65/RelA and NFAT reporter MCs, we aimed to investigate the influence of SCF on alarmin-induced signaling pathways and the resulting cytokine production and degranulation. We found that the presence of SCF boosted the cytokine production but not degranulation in MCs which simultaneously sense ATP and IL-33 (ATP/IL-33 co-sensing). Therefore, we conclude that SCF maintains the functionality of MCs in peripheral tissues to ensure appropriate MC reactions upon cell damage, induced by pathogens or allergens.
Asunto(s)
Citocinas , Mastocitos , Factor de Células Madre , Adenosina Trifosfato/metabolismo , Alarminas/metabolismo , Citocinas/metabolismo , Interleucina-33/metabolismo , Mastocitos/metabolismo , Factor de Células Madre/farmacología , Factor de Células Madre/fisiología , Masculino , Femenino , Animales , Ratones , Ratones Endogámicos C57BLRESUMEN
BACKGROUND: Stem cells are located in specific regulatory environments termed niches, which modulate the survival and proliferation of the cells through a variety of both mitogenic and inhibitory cytokines. In the murine prostate, stem cells are located in the proximal region of prostatic ducts. We examined the regulation of murine prostate cells in the stem cell niche by transforming growth factor beta (TGF-ß) and stem cell factor (SCF). METHODS: Prostate cells from the proximal and distal regions of prostatic ducts were cultured in the presence and absence of TGF-ß and SCF, both on collagen-coated wells and in collagen gels. Cell growth on collagen was assessed by determining cell number. Cell growth in collagen gels was quantified by determining the number, size and complexity of prostatic ducts. The basal and luminal phenotype of the cells was determined by immunohistochemistry. RESULTS: Endogenous TGF-ß inhibited proliferation and promoted differentiation of proximal cells towards a luminal phenotype. It also inhibited duct-forming capacity and promoted differentiation of prostatic ducts towards a luminal phenotype. Addition of SCF enhanced proximal cell proliferation on collagen-coated wells and duct formation in collagen gels. Proliferation was further increased by ablation of endogenous TGF-ß. CONCLUSION: Proliferation and the basal/luminal cell composition of cells isolated from the proximal region of prostatic ducts, the stem cell niche, is regulated in part by opposing effects of SCF and endogenous TGF-ß.