Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 341
Filtrar
1.
Cells ; 12(23)2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-38067124

RESUMO

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.


Assuntos
Citocinas , Mastócitos , Fator de Células-Tronco , Trifosfato de Adenosina/metabolismo , Alarminas/metabolismo , Citocinas/metabolismo , Interleucina-33/metabolismo , Mastócitos/metabolismo , Fator de Células-Tronco/farmacologia , Fator de Células-Tronco/fisiologia , Masculino , Feminino , Animais , Camundongos , Camundongos Endogâmicos C57BL
2.
Sci Rep ; 10(1): 21732, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33303806

RESUMO

Poor wound closure due to diabetes, aging, stress, obesity, alcoholism, and chronic disease affects millions of people worldwide. Reasons wounds will not close are still unclear, and current therapies are limited. Although stem cell factor (SCF), a cytokine, is known to be important for wound repair, the cellular and molecular mechanisms of SCF in wound closure remain poorly understood. Here, we found that SCF expression in the epidermis is decreased in mouse models of delayed wound closure intended to mimic old age, obesity, and alcoholism. By using SCF conditionally knocked out mice, we demonstrated that keratinocytes' autocrine production of SCF activates a transient c-kit receptor in keratinocytes. Transient activation of the c-kit receptor induces the expression of growth factors and chemokines to promote wound re-epithelialization by increasing migration of skin cells (keratinocytes and fibroblasts) and immune cells (neutrophils) to the wound bed 24-48 h post-wounding. Our results demonstrate that keratinocyte-produced SCF is essential to wound closure due to the increased recruitment of a unique combination of skin cells and immune cells in the early phase after wounding. This discovery is imperative for developing clinical strategies that might improve the body's natural repair mechanisms for treating patients with wound-closure pathologies.


Assuntos
Queratinócitos/metabolismo , Síndrome Metabólica/fisiopatologia , Reepitelização/genética , Reepitelização/fisiologia , Fenômenos Fisiológicos da Pele , Pele/lesões , Fator de Células-Tronco/deficiência , Fator de Células-Tronco/fisiologia , Cicatrização/genética , Cicatrização/fisiologia , Animais , Modelos Animais de Doenças , Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pele/citologia , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo
3.
Elife ; 92020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32242818

RESUMO

Photoreceptor degeneration is a major cause of blindness and a considerable health burden during aging but effective therapeutic or preventive strategies have not so far become readily available. Here, we show in mouse models that signaling through the tyrosine kinase receptor KIT protects photoreceptor cells against both light-induced and inherited retinal degeneration. Upon light damage, photoreceptor cells upregulate Kit ligand (KITL) and activate KIT signaling, which in turn induces nuclear accumulation of the transcription factor NRF2 and stimulates the expression of the antioxidant gene Hmox1. Conversely, a viable Kit mutation promotes light-induced photoreceptor damage, which is reversed by experimental expression of Hmox1. Furthermore, overexpression of KITL from a viral AAV8 vector prevents photoreceptor cell death and partially restores retinal function after light damage or in genetic models of human retinitis pigmentosa. Hence, application of KITL may provide a novel therapeutic avenue for prevention or treatment of retinal degenerative diseases.


Assuntos
Células Fotorreceptoras de Vertebrados/efeitos da radiação , Degeneração Retiniana/prevenção & controle , Fator de Células-Tronco/fisiologia , Animais , Modelos Animais de Doenças , Heme Oxigenase-1/análise , Luz , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/fisiologia , Proteínas Proto-Oncogênicas c-kit/fisiologia , Degeneração Retiniana/etiologia , Degeneração Retiniana/genética , Transdução de Sinais
4.
J Clin Invest ; 128(7): 2848-2861, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29596064

RESUMO

Neurofibromatosis type 1 associates with multiple neoplasms, and the Schwann cell tumor neurofibroma is the most prevalent. A hallmark feature of neurofibroma is mast cell infiltration, which is recruited by chemoattractant stem cell factor (SCF) and has been suggested to sustain neurofibroma tumorigenesis. In the present study, we use new, genetically engineered Scf mice to decipher the contributions of tumor-derived SCF and mast cells to neurofibroma development. We demonstrate that mast cell infiltration is dependent on SCF from tumor Schwann cells. However, removal of mast cells by depleting the main SCF source only slightly affects neurofibroma progression. Other inflammation signatures show that all neurofibromas are associated with high levels of macrophages regardless of Scf status. These findings suggest an active inflammation in neurofibromas and partly explain why mast cell removal alone is not sufficient to relieve tumor burden in this experimental neurofibroma model. Furthermore, we show that plexiform neurofibromas are highly associated with injury-prone spinal nerves that are close to flexible vertebras. In summary, our study details the role of inflammation in neurofibromagenesis. Our data indicate that prevention of inflammation and possibly also nerve injury at the observed tumor locations are therapeutic approaches for neurofibroma prophylaxis and that such treatment should be explored.


Assuntos
Inflamação/complicações , Neurofibroma Plexiforme/etiologia , Microambiente Tumoral , Animais , Carcinogênese , Progressão da Doença , Feminino , Genes da Neurofibromatose 1 , Humanos , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Mastócitos/patologia , Mastócitos/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Neurofibroma Plexiforme/patologia , Neurofibroma Plexiforme/fisiopatologia , Neurofibromatose 1/complicações , Células de Schwann/patologia , Células de Schwann/fisiologia , Fator de Células-Tronco/deficiência , Fator de Células-Tronco/genética , Fator de Células-Tronco/fisiologia , Microambiente Tumoral/fisiologia
5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(7): 920-925, 2017 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-28712399

RESUMO

Objective To study the correlation between the expressions of stem cell factor (SCF) and hypoxia inducible factor 1 alpha (HIF-1α) in pancreatic cancer, and investigate the mechanism by which SCF regulates the expression of HIF-1α. Methods Immunohistochemistry was used to detect the expressions of SCF and HIF-1α in pancreatic cancer specimens and to analyze the correlation between SCF and HIF-1α expressions. Pancreatic cancer PANC-1 cells were treated with different doses of SCF (0, 1, 10, 100 ng/mL) alone or combined with c-KIT inhibitor Gleevec (5 µmol/L). Real-time fluorescent quantitative PCR (qRT-PCR) was performed to detect the level of HIF-1α mRNA, and Western blotting to detect the HIF-1α protein level, the phosphorylation levels of ERK1/2 and AKT. Results SCF and HIF-1α were up-regulated in pancreatic cancer samples and they had an obvious positive correlation. In PANC-1 cells, SCF didn't affect the expression of HIF-1α mRNA, but up-regulated the expression of HIF-1α protein in a dose-dependent manner. Gleevec inhibited the SCF-induced up-regulation of HIF-1α protein, but did not affect the mRNA. And Gleevec blocked the phosphorylation of AKT and ERK1/2. Conclusion SCF/c-KIT can up-regulate the protein expression of HIF-1α by activating AKT and ERK signaling pathways in pancreatic cancer cells.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pancreáticas/metabolismo , Fator de Células-Tronco/fisiologia , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Mesilato de Imatinib/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Células-Tronco/genética
6.
Cytokine ; 95: 35-42, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28235674

RESUMO

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.


Assuntos
Aorta/citologia , Gônadas/citologia , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Mesonefro/citologia , Trombopoetina/fisiologia , Animais , Aorta/embriologia , Aorta/metabolismo , Células Cultivadas , Gônadas/embriologia , Gônadas/metabolismo , Interleucina-3/fisiologia , Mesonefro/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Receptores de Trombopoetina/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Transdução de Sinais , Fator de Células-Tronco/fisiologia , Transdução Genética
7.
J Formos Med Assoc ; 116(7): 542-548, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27707610

RESUMO

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.


Assuntos
Proteínas Proto-Oncogênicas c-kit/fisiologia , Fator de Células-Tronco/fisiologia , Testículo/metabolismo , Varicocele/metabolismo , Animais , Masculino , Proteínas Proto-Oncogênicas c-kit/análise , Ratos , Ratos Sprague-Dawley , Túbulos Seminíferos/patologia , Contagem de Espermatozoides , Espermatogênese , Fator de Células-Tronco/análise
8.
Reprod Fertil Dev ; 29(7): 1356-1368, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27279472

RESUMO

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.


Assuntos
Gatos/genética , Gatos/fisiologia , Folículo Ovariano/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Fator de Células-Tronco/fisiologia , Animais , Fator de Crescimento Epidérmico/administração & dosagem , Fator de Crescimento Epidérmico/fisiologia , Feminino , Técnicas In Vitro , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do FSH/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Células-Tronco/administração & dosagem , Regulação para Cima/efeitos dos fármacos
9.
Sci Rep ; 6: 26812, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27245949

RESUMO

C-kit positive cardiac stem cells (CSCs) have been shown to contribute to myocardial regeneration after infarction. Previously, we have shown that the c-kit ligand stem cell factor (SCF) can induce CSC migration into the infarcted area during myocardial infarction (MI). However, the precise mechanism involved is not fully understood. In this study, we found that CSCs also express C-X-C chemokine receptor type 4 (CXCR4), which is a typical member of the seven transmembrane-spanning G protein-coupled receptor (GPCR). In vitro, activation of c-kit signalling by SCF promotes migration of CSCs with increased phosphorylation of CXCR4-serine 339, p38 mitogen-activated protein kinase (p38 MAPK) and extracellular regulated protein kinases 1/2 (ERK1/2). Knockdown of CXCR4 expression by siRNA reduces SCF/c-kit-induced migration and downstream signalling. As previously reported, CXCR4-serine 339 phosphorylation is mainly regulated by GPCR kinase 6 (GRK6); thus, silencing of GRK6 expression by siRNA impairs CXCR4-serine 339 phosphorylation and migration of CSCs caused by SCF. In vivo, knockdown of GRK6 impairs the ability of CSCs to migrate into peri-infarcted areas. These results demonstrate that SCF-induced CSC migration is regulated by the transactivation of CXCR4-serine 339 phosphorylation, which is mediated by GRK6.


Assuntos
Células-Tronco Adultas/fisiologia , Quimiotaxia/fisiologia , Quinases de Receptores Acoplados a Proteína G/fisiologia , Miocárdio/citologia , Proteínas Proto-Oncogênicas c-kit/fisiologia , Receptores CXCR4/metabolismo , Fator de Células-Tronco/fisiologia , Animais , Células Cultivadas , Ativação Enzimática , Feminino , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/genética , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Fosforilação , Fosfosserina/metabolismo , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/genética , Ativação Transcricional , Transfecção
10.
Radiother Oncol ; 119(3): 537-43, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27178146

RESUMO

BACKGROUND AND PURPOSE: Success of radiotherapy is often limited by therapy resistance and metastasis resulting from cancer cell motility. It was tested in vitro whether this cancer cell motility is affected by growth condition, active SCF/c-Kit pathway or X-irradiation. MATERIALS AND METHODS: Cell motility was measured with BioCoat™ Matrigel™ invasion chamber using four different cancer cell lines (NSCLC: H23, H520, H226 and PrCa: DU145). Cells were grown in 2D or 3D, SCF was knocked down by siRNA and cells were irradiated with 2 or 6Gy. RESULTS: All cell lines except H520 showed a 2-3-fold increase in cell motility when grown in 3D. This effect was considered to result from the EMT-like change seen when cells were grown in 3D as indicated by the enhanced expression of vimentin and N-cadherin and reduction of E-cadherin. Just the opposite trends were found for H520 cells. Knockdown of SCF was found to result in reduced cell motility for both 2D and 3D. In contrast, X-irradiation did not modulate cell motility neither under 2D nor 3D. In line with this, X-irradiation did neither induce the expression of EMT-associated genes nor SCF. CONCLUSION: X-irradiation affects neither the expression of important EMT genes such as vimentin, E-cadherin and N-cadherin nor SCF/c-Kit signaling and, as a consequence, does not alter cell motility.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-kit/fisiologia , Transdução de Sinais/fisiologia , Fator de Células-Tronco/fisiologia , Antígenos CD/fisiologia , Caderinas/fisiologia , Movimento Celular , Transição Epitelial-Mesenquimal , Humanos , Células Tumorais Cultivadas , Raios X
11.
Exp Cell Res ; 342(2): 210-5, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26997528

RESUMO

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.


Assuntos
Diferenciação Celular , Mastócitos/fisiologia , Animais , Biologia Celular/história , História do Século XIX , História do Século XX , Humanos , Fator de Células-Tronco/fisiologia
12.
J Assist Reprod Genet ; 32(12): 1741-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26507072

RESUMO

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.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Ovário/metabolismo , Fator de Células-Tronco/fisiologia , Animais , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/metabolismo , Mesilato de Imatinib/farmacologia , Técnicas In Vitro , Camundongos , Oócitos/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Fator de Células-Tronco/metabolismo
13.
J Invest Dermatol ; 135(2): 516-524, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25178104

RESUMO

The receptor tyrosine kinase KIT and its ligand, stem cell factor (SCF), are essential for the proliferation and survival of normal melanocytes. In melanomas arising on mucosal, acral, and chronically sun-damaged skin, activating KIT mutations have been identified as oncogenic drivers and potent therapeutic targets. Through an initial whole-genome screen for aberrant promoter methylation in melanoma, we identified the KIT promoter as a target for hypermethylation in 43/110 melanoma cell lines, and in 3/12 primary and 11/29 metastatic cutaneous melanomas. Methylation density at the KIT promoter correlated inversely with promoter activity in vitro and in vivo, and the expression of KIT was restored after treatment with the demethylating agent 5-aza-2'-deoxycytidine. Hypermethylation of KIT showed no direct or inverse correlations with well-documented melanoma drivers. Growth of melanoma cells in the presence of SCF led to reduced KIT expression and increased methylation density at the KIT promoter, suggesting that SCF may exert a selection pressure for the loss of KIT. The frequent loss of KIT in cutaneous melanoma by promoter hypermethylation suggests that distinct KIT signaling pathways have opposing roles in the pathogenesis of melanoma subtypes.


Assuntos
Epigênese Genética , Melanoma/genética , Proteínas Proto-Oncogênicas c-kit/genética , Neoplasias Cutâneas/genética , Linhagem Celular Tumoral , Metilação de DNA , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Fator de Células-Tronco/fisiologia
14.
Stem Cells ; 32(7): 1956-67, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24753135

RESUMO

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.


Assuntos
Proliferação de Células , Células-Tronco Hematopoéticas/fisiologia , Proteínas Imediatamente Precoces/genética , Proteínas Tirosina Fosfatases/genética , Animais , Células Cultivadas , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Proteínas Imediatamente Precoces/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-kit/fisiologia , Transdução de Sinais , Fator de Células-Tronco/fisiologia
15.
Stem Cells ; 32(7): 1878-89, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24677703

RESUMO

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.


Assuntos
Células-Tronco Adultas/enzimologia , Células-Tronco Hematopoéticas/enzimologia , Janus Quinase 2/fisiologia , Células-Tronco Adultas/fisiologia , Anemia Aplástica , Animais , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Proliferação de Células , Sobrevivência Celular , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Hemoglobinúria Paroxística/enzimologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Espécies Reativas de Oxigênio/metabolismo , Fator de Células-Tronco/fisiologia , Trombopoetina/fisiologia
16.
Clin Exp Dermatol ; 39(2): 202-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24479586

RESUMO

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.


Assuntos
Benzamidas/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Melanócitos/efeitos dos fármacos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Epidérmicas , Fibroblastos/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Melaninas/metabolismo , Melanócitos/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Modelos Biológicos , Monofenol Mono-Oxigenase/metabolismo , Proteínas Proto-Oncogênicas c-kit/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator de Células-Tronco/fisiologia , Células Tumorais Cultivadas
17.
Arch Pharm Res ; 37(2): 175-85, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23709168

RESUMO

The interaction of stem cell factor (SCF) with its cognate receptor c-Kit is closely associated with the survival and maturation of melanocytes. To investigate novel depigmentation agents, we screened 2,000 plant extracts for c-Kit inhibitors to identify active small molecules by using time-resolved fluorescence enzyme assays. For the active extracts identified as inhibitors of c-Kit enzyme, we evaluated the effects of the active extracts and isolated flavonoids on c-Kit phosphorylation in MO7e/melanocytes. Anti-melanogenic activity was also examined in melanocytes and melanoderm model. The flavonoids such as diosmetin, apigenin, acacetin and luteolin isolated from Chrysanthemum morifolium were found to be active in inhibiting c-Kit both at enzyme and cellular levels. In addition, these flavonoids attenuated SCF-induced proliferation of human primary melanocytes without toxicity and suppressed ultraviolet (UV) B irradiation-mediated melanin synthesis significantly. Among the active flavonoids, diosmetin was found to inhibit SCF-induced melanogenesis in a human melanoderm model. These results strongly suggest that C. morifolium extract and diosmetin have potential to suppress SCF-/UVB-induced melanogenesis, and could be developed as anti-pigmentation agents.


Assuntos
Chrysanthemum/química , Flavonoides/farmacologia , Melaninas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Western Blotting , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Flavonoides/isolamento & purificação , Flores/química , Fluorimunoensaio , Humanos , Melaninas/biossíntese , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanócitos/efeitos da radiação , Camundongos , Microscopia de Contraste de Fase , Modelos Biológicos , Proteínas Proto-Oncogênicas c-kit/genética , Células Sf9 , Spodoptera , Fator de Células-Tronco/farmacologia , Fator de Células-Tronco/fisiologia , Raios Ultravioleta
18.
Am J Chin Med ; 41(6): 1233-49, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24228598

RESUMO

Electroacupuncture (EA) at ST36 is effective for improving gastric motility. However, the underlying mechanism remains poorly understood. The aim of this study was to investigate the effects of EA on gastric contraction and to determine whether interstitial cells of Cajal (ICCs) are involved. Rats were randomized into control, diabetic (DM), diabetic with sham EA (DM + SEA), diabetic with low frequency EA (DM + LEA) and diabetic with high frequency EA (DM + HEA) groups. EA was performed everyday for four and eight weeks. Contractions in antrum strips were explored using the organ bath technique. Western blotting was employed to determine c-kit and transmembrane stem cell factor (M-SCF) expression in the gastric antrum, and levels of soluble stem cell factor (S-SCF) in serum were determined by enzyme-linked immunosorbent assay (ELISA). The distribution of ICCs was further assessed by immunohistochemistry. The results were as follows: (1) Contractions in the DM group were attenuated at four and eight weeks, but LEA and HEA restored the attenuated contraction. (2) ICCs were significantly decreased at eight weeks without alteration at four weeks in DM group, but were rescued in the LEA and HEA groups. (3) Whereas M-SCF and S-SCF in the DM group were slightly decreased at four weeks and were dramatically reduced at eight weeks, LEA and HEA markedly enhanced SCF at eight weeks. Collectively, the data suggest that in diabetic rats, LEA and HEA at ST36 could facilitate contraction of the gastric antrum, possibly by involving the SCF/c-kit pathway.


Assuntos
Pontos de Acupuntura , Diabetes Mellitus Experimental/fisiopatologia , Eletroacupuntura/métodos , Motilidade Gastrointestinal/genética , Motilidade Gastrointestinal/fisiologia , Células Intersticiais de Cajal/fisiologia , Contração Muscular/genética , Contração Muscular/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Antro Pilórico/citologia , Antro Pilórico/fisiologia , Transdução de Sinais/fisiologia , Fator de Células-Tronco/metabolismo , Acetilcolina/farmacologia , Animais , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Imuno-Histoquímica , Técnicas In Vitro , Células Intersticiais de Cajal/metabolismo , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso/citologia , Músculo Liso/fisiologia , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/fisiologia , Ratos , Ratos Sprague-Dawley , Fator de Células-Tronco/genética , Fator de Células-Tronco/fisiologia
19.
J Immunol ; 191(12): 5885-94, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24218457

RESUMO

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.


Assuntos
Calcineurina/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Mastócitos/metabolismo , Proteínas Musculares/fisiologia , Fator de Células-Tronco/fisiologia , Animais , Células da Medula Óssea , Inibidores de Calcineurina , Proteínas de Ligação ao Cálcio , Células Cultivadas , Proteínas de Ligação a DNA , Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Retroalimentação Fisiológica , Sangue Fetal/citologia , Regulação da Expressão Gênica , Humanos , Imunossupressores/farmacologia , Interleucina-6/biossíntese , Interleucina-6/genética , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Fatores de Transcrição NFATC/metabolismo , Proteínas Proto-Oncogênicas c-kit/fisiologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Células-Tronco/farmacologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
20.
Anim Reprod Sci ; 142(3-4): 160-7, 2013 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-24139236

RESUMO

In pigs, primordial to primary follicle transition occur in the late pregnancy. The interactions between Kit ligand (KL) and its receptor (c-Kit), as well as insulin-like growth factor 1 (IGF1) and cognate receptor (IGF1R) are crucial for the primordial follicle activation. It is well established that hormonal disruption induces abnormalities in the developing reproductive system. Hence, this study investigated the influence of antiandrogen, flutamide, on genes involved in the primordial to primary follicle transition. Pregnant gilts were injected with flutamide (50mg/kg bw, seven times, every day) or corn oil (control groups) starting on gestation days 83 (GD90) or 101 (GD108). Fetal ovaries were excised on days 90 and 108 of gestation. The proportion of primordial and primary follicles was determined, and immunohistochemistry for c-Kit and IGF1R was conducted. To assess KL, c-Kit, IGF1 and IGF1R mRNA expression real-time PCR was performed. Ovaries from both GD90 and GD108 animals exhibited a greater proportion of primordial to primary follicles when compared to respective control groups. C-Kit and IGF1R were immunolocalized in the oocytes of primordial and primary follicles. Both c-Kit mRNA and protein levels and KL mRNA expression were diminished in GD90 group. IGF1R expression decreased at mRNA and protein levels, whereas IGF1 mRNA expression was increased in GD90 and GD108 groups. In summary, our findings may indicate that the interactions between KL and c-Kit as well as IGF1 and IGF1R are relevant to the initiation of follicular transition from primordial into primary follicles and can be affected by AR signaling.


Assuntos
Antagonistas de Androgênios/farmacologia , Flutamida/farmacologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Ovário/efeitos dos fármacos , Suínos , Animais , Feminino , Feto/efeitos dos fármacos , Feto/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Fator de Crescimento Insulin-Like I/fisiologia , Folículo Ovariano/embriologia , Ovário/citologia , Ovário/embriologia , Ovário/fisiologia , Gravidez , Proteínas Proto-Oncogênicas c-kit/fisiologia , Receptor IGF Tipo 1/fisiologia , Transdução de Sinais/fisiologia , Fator de Células-Tronco/fisiologia , Suínos/embriologia , Suínos/genética , Suínos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA