Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS One ; 4(12): e8508, 2009 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-20041145

RESUMO

The intracellular gram-negative bacterium Francisella tularensis causes the disease tularemia and is known for its ability to subvert host immune responses. Previous work from our laboratory identified the PI3K/Akt pathway and SHIP as critical modulators of host resistance to Francisella. Here, we show that SHIP expression is strongly down-regulated in monocytes and macrophages following infection with F. tularensis novicida (F.n.). To account for this negative regulation we explored the possibility that microRNAs (miRs) that target SHIP may be induced during infection. There is one miR that is predicted to target SHIP, miR-155. We tested for induction and found that F.n. induced miR-155 both in primary monocytes/macrophages and in vivo. Using luciferase reporter assays we confirmed that miR-155 led to down-regulation of SHIP, showing that it specifically targets the SHIP 3'UTR. Further experiments showed that miR-155 and BIC, the gene that encodes miR-155, were induced as early as four hours post-infection in primary human monocytes. This expression was dependent on TLR2/MyD88 and did not require inflammasome activation. Importantly, miR-155 positively regulated pro-inflammatory cytokine release in human monocytes infected with Francisella. In sharp contrast, we found that the highly virulent type A SCHU S4 strain of Francisella tularensis (F.t.) led to a significantly lower miR-155 response than the less virulent F.n. Hence, F.n. induces miR-155 expression and leads to down-regulation of SHIP, resulting in enhanced pro-inflammatory responses. However, impaired miR-155 induction by SCHU S4 may help explain the lack of both SHIP down-regulation and pro-inflammatory response and may account for the virulence of Type A Francisella.


Assuntos
Citocinas/imunologia , Francisella tularensis/patogenicidade , Francisella/fisiologia , Mediadores da Inflamação/imunologia , MicroRNAs/genética , Monoéster Fosfórico Hidrolases/genética , Animais , Sequência de Bases , Caspase 1/metabolismo , Linhagem Celular , Citocinas/biossíntese , Regulação para Baixo/genética , Endocitose , Ativação Enzimática , Francisella/citologia , Infecções por Bactérias Gram-Negativas/enzimologia , Infecções por Bactérias Gram-Negativas/genética , Humanos , Mediadores da Inflamação/metabolismo , Inositol Polifosfato 5-Fosfatases , Camundongos , MicroRNAs/metabolismo , Viabilidade Microbiana , Modelos Biológicos , Dados de Sequência Molecular , Monoéster Fosfórico Hidrolases/metabolismo , Transdução de Sinais , Receptores Toll-Like , Virulência/genética
2.
J Thorac Oncol ; 4(8): 1028-34, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19474765

RESUMO

Lung cancer is the leading cause of cancer related deaths in the United States. It is estimated that in 2008 there were 215,000 new diagnoses of lung cancer and 163,000 deaths. Despite emerging technologies for potential early diagnosis and discovery of novel targeted therapies, the overall 5-year survival remains a disappointing 15%. Explanations for the poor survival include late presentation of disease, a lack of markers for early detection, and both phenotypic and genotypic heterogeneity within patients of similar histologic classification. To further understand this heterogeneity and thus complexity of lung cancer, investigators have applied various technologies including high throughput analysis of both the genome and proteome. Such approaches have been successful in identifying signatures that may clarify molecular differences in tumors, identify new targets, and improve prognostication. In the last decade, investigators have identified a new mode of gene regulation in the form of noncoding RNAs termed microRNAs (miRNAs or miRs). First determined to be of importance in larval development, microRNAs are approximately 19-22 nucleotide single stranded RNAs that regulate genes by either inducing mRNA degradation or inhibiting translation. MiRNAs have been implicated in several cellular processes including apoptosis, development, proliferation, and differentiation. By regulating hundreds of genes simultaneously, miRNAs have the capacity for regulation of biologic networks. Global alterations in miRNA expression in both solid organ and hematological malignancies suggest their importance in the pathogenesis of disease. To date, both in vivo and in vitro studies in lung cancer demonstrate a dysregulation of miRNA expression. Furthermore, investigators are beginning to identify individual targets and pathways of miRNAs relevant to lung tumorigenesis. Thus, miRNAs may identify critical targets and be important in the pathogenesis of lung cancer.


Assuntos
Neoplasias Pulmonares/etiologia , MicroRNAs/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia
3.
Am J Respir Crit Care Med ; 179(1): 4-10, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18787215

RESUMO

Over the last 15 years, investigators have identified small noncoding RNAs as regulators of gene expression. One type of noncoding RNAs are termed microRNAs (miRNAs). miRNAs are evolutionary conserved, approximately 22-nucleotide single-stranded RNAs that target genes by inducing mRNA degradation or by inhibiting translation. miRNAs are implicated in many critical cellular processes, including apoptosis, proliferation, and differentiation. Furthermore, it is estimated that miRNAs may be responsible for regulating the expression of nearly one-third of the human genome. Despite the identification of greater than 500 mature miRNAs, very little is known about their biological functions and functional targets. In the last 5 years, researchers have increasingly focused on the functional relevance and role that miRNAs play in the pathogenesis of human disease. miRNAs are known to be important in solid organ and hematological malignancies, heart disease, as potential modulators of the immune response, and organ development. It is anticipated that miRNA analysis will emerge as an important complement to proteomic and genomic studies to further our understanding of disease pathogenesis. Despite the application of genomics and proteomics to the study of human lung disease, few studies have examined miRNA expression. This perspective is not meant to be an exhaustive review of miRNA biology but will provide an overview of both miRNA biogenesis and our current understanding of the role of miRNAs in lung disease as well as a perspective on the importance of integrating this analysis as a tool for identifying and understanding the biological pathways in lung-disease pathogenesis.


Assuntos
Pneumopatias/fisiopatologia , MicroRNAs/fisiologia , Linfócitos B/fisiologia , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade Inata/fisiologia , Pneumopatias/metabolismo , MicroRNAs/efeitos dos fármacos , MicroRNAs/imunologia , MicroRNAs/metabolismo , Linfócitos T/fisiologia
4.
Proc Natl Acad Sci U S A ; 105(50): 19678-83, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19066217

RESUMO

The transcription factors E2F and Myc participate in the control of cell proliferation and apoptosis, and can act as oncogenes or tumor suppressors depending on their levels of expression. Positive feedback loops in the regulation of these factors are predicted-and recently shown experimentally-to lead to bistability, which is a phenomenon characterized by the existence of low and high protein levels ("off" and "on" levels, respectively), with sharp transitions between levels being inducible by, for example, changes in growth factor concentrations. E2F and Myc are inhibited at the posttranscriptional step by members of a cluster of microRNAs (miRs) called miR-17-92. In return, E2F and Myc induce the transcription of miR-17-92, thus forming a negative feedback loop in the interaction network. The consequences of the coupling between the E2F/Myc positive feedback loops and the E2F/Myc/miR-17-92 negative feedback loop are analyzed using a mathematical model. The model predicts that miR-17-92 plays a critical role in regulating the position of the off-on switch in E2F/Myc protein levels, and in determining the on levels of these proteins. The model also predicts large-amplitude protein oscillations that coexist with the off steady state levels. Using the concept and model prediction of a "cancer zone," the oncogenic and tumor suppressor properties of miR-17-92 is demonstrated to parallel the same properties of E2F and Myc.


Assuntos
Fatores de Transcrição E2F/metabolismo , Redes Reguladoras de Genes , MicroRNAs/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Fatores de Transcrição E2F/genética , Retroalimentação Fisiológica , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , MicroRNAs/genética , Modelos Biológicos , Neoplasias/genética , Proteínas Oncogênicas , Oncogenes , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Supressoras de Tumor/genética
5.
Cytokine ; 43(2): 114-23, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18554923

RESUMO

The granulocyte colony-stimulating factor receptor (G-CSFR) is a critical regulator of granulopoiesis, but the mechanisms controlling its surface expression are poorly understood. Recent studies using transfected cell lines have suggested the activated G-CSFR is routed to the lysosome and not the proteasome. Here, we examined the role of the ubiquitin/proteasome system in regulating G-CSFR surface expression in both ts20 cells that have a temperature-sensitive E1 ubiquitin-activating enzyme and in primary human neutrophils. We show that the G-CSFR is constitutively ubiquitinated, which increases following ligand binding. In the absence of a functional E1 enzyme, ligand-induced internalization of the receptor is inhibited. Pre-treatment of ts20 transfectants with either chloroquine or MG132 inhibited ligand-induced G-CSFR degradation, suggesting a role for both lysosomes and proteasomes in regulating G-CSFR surface expression in this cell line. In neutrophils, inhibition of the proteasome but not the lysosome was found to inhibit internalization/degradation of the activated G-CSFR. Collectively, these data demonstrate the requirement for a functional ubiquitin/proteasome system in G-CSFR internalization and degradation. Our results suggest a prominent role for the proteasome in physiologic modulation of the G-CSFR, and provide further evidence for the importance of the ubiquitin/proteasome system in the initiation of negative signaling by cytokine receptors.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Animais , Linhagem Celular , Cricetinae , Ligantes , Lisossomos/metabolismo , Neutrófilos/metabolismo , Inibidores de Proteassoma , Ligação Proteica , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Ubiquitinação
6.
PLoS One ; 3(6): e2452, 2008 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-18560579

RESUMO

BACKGROUND: Expression of truncated G-CSFR forms in patients with SCN/AML induces hyperproliferation and prolonged cell survival. Previously, we showed that ligand internalization is delayed and degradation of truncated G-CSFR forms is defective in patients with SCN/AML. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we investigated the potential roles of dileucine and tyrosine-based motifs within the cytoplasmic domain of the G-CSFR in modulating ligand/receptor internalization. Using standard binding assays with radiolabeled ligand and COS-7 cells, substitutions in the dileucine motif or deletion of tyrosine residues in the G-CSFR did not alter internalization. Attachment of the transferrin receptor YTRF internalization motif to a truncated G-CSFR form from a patient with SCN/AML corrected defective internalization, but not receptor degradation suggesting that receptor internalization and degradation occur independently via distinct domains and/or processes. CONCLUSIONS: Our data suggest that distinct domains within the G-CSFR mediate separate processes for receptor internalization and degradation. Our findings using standard binding assays differ from recently published data utilizing flow cytometry.


Assuntos
Endocitose , Leucemia Mieloide Aguda/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Animais , Sequência de Bases , Células COS , Chlorocebus aethiops , Primers do DNA , Leucemia Mieloide Aguda/patologia , Ligantes , Mutagênese Sítio-Dirigida , Receptores de Fator Estimulador de Colônias de Granulócitos/química , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Tirosina/metabolismo
7.
J Leukoc Biol ; 83(5): 1277-85, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18272588

RESUMO

Expression of leucine-rich alpha2 glycoprotein (LRG), a member of the leucine-rich repeat family of proteins, was recently shown to be up-regulated during neutrophil differentiation. Its precise role in granulopoiesis, however, remains unknown. In this paper, we show that the transcription factors PU.1 and C/EBPepsilon that regulate the expression of multiple myeloid-specific genes also bind to the LRG promoter. We also demonstrate that LRG localizes to the same cytoplasmic compartment as myeloperoxidase and that G-CSF treatment of the 32Dcl3 myeloid cell line induces nuclear translocation of LRG. Stable transfection of LRG into 32Dcl3 cells resulted in accelerated, G-CSF-mediated neutrophil differentiation and induction of CD11b expression. In contrast, constitutive expression of LRG in 32Dwt18 cells, expressing a chimeric erythropoietin (Epo)/G-CSFR consisting of the EpoR extracellular domain fused to the G-CSFR transmembrane and cytoplasmic domains, failed to induce accelerated neutrophil differentiation and CD11b expression in response to Epo stimulation. LRG-mediated accelerated differentiation and CD11b expression were found to correlate with an increased level of phospho-Stat3 but not with PU.1 or p27(kip1) levels. Hence, similar to other genes involved in neutrophil differentiation, the expression of LRG also appears to be regulated by PU.1 and C/EBPepsilon. Collectively, these findings suggest a role for LRG in modulating neutrophil differentiation and expression of CD11b via nonredundant G-CSFR signals.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Diferenciação Celular/efeitos dos fármacos , Glicoproteínas/fisiologia , Ativação de Neutrófilo/fisiologia , Neutrófilos/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Animais , Fracionamento Celular , Cromatina/ultraestrutura , Eritropoetina/farmacologia , Citometria de Fluxo , Glicoproteínas/genética , Camundongos , Microscopia Confocal , Neutrófilos/citologia , Regiões Promotoras Genéticas
8.
J Inflamm (Lond) ; 4: 19, 2007 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-17888174

RESUMO

BACKGROUND: The recruitment of mononuclear cells has important implications for tissue inflammation. Previous studies demonstrated enhanced CCR1 and CCR5 expression and decreased CCR2 expression during in vitro monocyte to macrophage differentiation. To date, no study examined the in vivo differences in chemokine receptor expression between human peripheral blood monocytes and alveolar macrophages. METHODS: We examined the expression of these receptors in human peripheral blood monocytes and alveolar macrophages using microarray analysis, reverse-transcriptase PCR, flow cytometry and migration analyses. RESULTS: In contrast to peripheral blood monocytes, alveolar macrophages did not express the CCL2 receptor, CCR2, and did not migrate toward CCL2. In contrast, monocytes and freshly isolated resident alveolar macrophages both migrated towards CCL3. However, up to 6-fold more monocytes migrated toward equivalent concentrations of CCL3 than did alveolar macrophages from the same donor. While peripheral blood monocytes expressed the CCL3 receptor, CCR1, alveolar macrophages expressed the alternate CCL3 receptor, CCR5. The addition of anti-CCR5 blocking antibodies completely abrogated CCL3-induced migration in alveolar macrophages, but did not affect the migration of peripheral blood monocytes. CONCLUSION: These data support the specificity of CCL2 to selectively drive monocyte, but not alveolar macrophage recruitment to the lung and CCR5 as the primary macrophage receptor for CCL3.

9.
Am J Respir Crit Care Med ; 176(1): 78-89, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17431224

RESUMO

RATIONALE: An increase in the number of mononuclear phagocytes in lung biopsies from patients with idiopathic pulmonary fibrosis (IPF) worsens prognosis. Chemokines that recruit mononuclear phagocytes, such as CC chemokine ligand 2 (CCL2), are elevated in bronchoalveolar lavage (BAL) fluid (BALF) from patients with IPF. However, little attention is given to the role of the mononuclear phagocyte survival and recruitment factor, macrophage colony-stimulating factor (M-CSF), in pulmonary fibrosis. OBJECTIVES: To investigate the role of mononuclear phagocytes and M-CSF in pulmonary fibrosis. METHODS: Wild-type, M-CSF-/-, or CCL2-/- mice received intraperitoneal bleomycin. Lung inflammation and fibrosis were measured by immunohistochemistry, ELISA, collagen assay, BAL differentials, real-time polymerase chain reaction, and Western blot analysis. Human and mouse macrophages were stimulated with M-CSF for CCL2 expression. BALF from patients with IPF was examined for M-CSF and CCL2. MEASUREMENTS AND MAIN RESULTS: M-CSF-/- and CCL2-/- mice had less lung fibrosis, mononuclear phagocyte recruitment, collagen deposition, and connective tissue growth factor (CTGF) expression after bleomycin administration than wild-type littermates. Human and mouse macrophages stimulated with M-CSF had increased CCL2 production, and intratracheal administration of M-CSF in mice induced CCL2 production in BALF. Finally, BALF from patients with IPF contained significantly more M-CSF and CCL2 than BALF from normal volunteers. Elevated levels of M-CSF were associated with elevated CCL2 in BALF and the diagnosis of IPF. CONCLUSIONS: These data suggest that M-CSF contributes to the pathogenesis of pulmonary fibrosis in mice and in patients with IPF through the involvement of mononuclear phagocytes and CCL2 production.


Assuntos
Quimiocina CCL2/imunologia , Fator Estimulador de Colônias de Macrófagos/imunologia , Macrófagos Alveolares/imunologia , Fibrose Pulmonar/imunologia , Adulto , Animais , Bleomicina , Líquido da Lavagem Broncoalveolar/citologia , Estudos de Casos e Controles , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Fagócitos/metabolismo , Fibrose Pulmonar/fisiopatologia
10.
Am J Respir Cell Mol Biol ; 36(1): 68-77, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16931806

RESUMO

M-CSF induces PI 3-kinase activation, resulting in reactive oxygen species (ROS) production. Previously, we reported that ROS mediate macrophage colony-stimulating factor (M-CSF)-induced extracellular regulated kinase (Erk) activation and monocyte survival. In this work, we hypothesized that M-CSF-stimulated ROS products modulated Akt1 and p38 activation. Furthermore, we sought to clarify the source of these ROS and the role of ROS and Akt in monocyte/macrophage survival. Macrophages from p47(phox-/-) mice, lacking a key component of the NADPH oxidase complex required for ROS generation, had reduced cell survival and Akt1 and p38 mitogen-activated protein kinase (MAPK) phosphorylation compared with wild-type macrophages in response to M-CSF stimulation, but had no difference in M-CSF-stimulated Erk. To understand how ROS affected monocyte survival and signaling, we observed that NAC and DPI decreased cell survival and Akt1 and p38 MAPK phosphorylation. Using bone marrow-derived macrophages from mice expressing constitutively activated Akt1 (Myr-Akt1) or transfecting Myr-Akt1 constructs into human peripheral monocytes, we concluded that Akt is a positive regulator of monocyte survival. Moreover, the p38 MAPK inhibitor, SB203580, inhibited p38 activity and M-CSF-induced monocyte survival. These findings demonstrate that ROS generated from the NADPH oxidase complex contribute to monocyte/macrophage survival induced by M-CSF via regulation of Akt and p38 MAPK.


Assuntos
Monócitos/fisiologia , NADPH Oxidases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Células da Medula Óssea/fisiologia , Sobrevivência Celular , Células Cultivadas , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Imidazóis/farmacologia , Fator Estimulador de Colônias de Macrófagos/fisiologia , Macrófagos/fisiologia , Camundongos , Fosforilação , Piridinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
11.
Antioxid Redox Signal ; 8(11-12): 2179-86, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17034361

RESUMO

Adiponectin (Ad), a member of the adipocytokine family, has been reported to possess antiinflammatory properties. We investigated the effects of full-length human Ad (hAd) on phorbol 12-myristate 13-acetate (PMA)-induced O2-* generation by human neutrophils. hAd, even at the lowest tested concentration of 0.001 microg/ml, after 30-min pretreatment of cells, significantly inhibited O2-* generation by neutrophils stimulated with PMA (100 nM). However, no relation between the dose of hAd and extent of inhibition of PMA-induced O2-* generation was observed with increasing the concentration of hAd up to 1 microg/ml. hAd also significantly inhibited neutrophil O2-* generation stimulated by N-formyl-methionyl-leucyl-phenylalanine (100 microM) and diacylglycerol (500 nM), as well as the PMA-induced neutrophil nitroblue tetrazolium reduction and H2O2 formation. Pretreatment of neutrophils with pronase-digested hAd failed to inhibit the PMA-induced O2-* generation. For the first time, this study revealed that Ad inhibited O2-* generation by neutrophils, possibly through regulation of NADPH oxidase.


Assuntos
Neutrófilos/efeitos dos fármacos , Superóxidos/antagonistas & inibidores , Adiponectina/farmacologia , Diglicerídeos/antagonistas & inibidores , Diglicerídeos/farmacologia , Relação Dose-Resposta a Droga , Humanos , Peróxido de Hidrogênio/metabolismo , N-Formilmetionina Leucil-Fenilalanina/antagonistas & inibidores , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/metabolismo , Proteínas Recombinantes/farmacologia , Superóxidos/metabolismo , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/antagonistas & inibidores , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo
12.
Circ Res ; 99(5): 477-84, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16873715

RESUMO

Women are at high risk of dying from unrecognized cardiovascular disease. Many differences in cardiovascular disease between men and women appear to be mediated by vascular smooth muscle cells (SMC). Because estrogen reduces the proliferation of SMC, we hypothesized that activation of estrogen receptor-alpha (ERalpha) by agonists or by growth factors altered SMC function. To determine the effect of growth factors, estrogen, and ERalpha expression on SMC differentiation, human aortic SMC were cultured in serum-free conditions for 10 days. SMC from men had lower spontaneous expression of ERalpha and higher levels of the differentiation markers calponin and smooth muscle alpha-actin than SMC from women. When SMC containing low expression of ERalpha were transduced with a lentivirus containing ERalpha, activation of the receptor by ligands or growth factors reduced differentiation markers. Conversely, inhibiting ERalpha expression by small interfering RNA (siRNA) in cells expressing high levels of ERalpha enhanced the expression of differentiation markers. ERalpha expression and activation reduced the phosphorylation of Smad2, a signaling molecule important in differentiation of SMC and initiated cell death through cleavage of caspase-3. We conclude that ERalpha activation switched SMC to a dedifferentiated phenotype and may contribute to plaque instability.


Assuntos
Aorta/citologia , Diferenciação Celular/fisiologia , Receptor alfa de Estrogênio/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Adulto , Aorta/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Ligantes , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fosforilação , RNA Interferente Pequeno/farmacologia , Proteína Smad2/antagonistas & inibidores , Proteína Smad2/metabolismo , Transdução Genética , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1
13.
Cell Signal ; 18(9): 1396-407, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16376521

RESUMO

Our earlier studies have shown that vitamin C at pharmacological doses (mM) induces loss of redox-dependent viability in bovine lung microvascular endothelial cells (BLMVECs) that is mediated by oxidative stress. Therefore, here, we investigated the vitamin C-induced activation of the lipid signaling enzyme, phospholipase D (PLD) in BLMVECs. Monolayer cultures of BLMVECs were treated with vitamin C (0-10 mM) for different time periods (0-2 h) and the activity of PLD was determined. Vitamin C induced activation of PLD in BLMVECs in a time- and dose-dependent fashion that was significantly attenuated by antioxidants, p38 mitogen-activated protein kinase (p38 MAPK)-specific inhibitor (SB203580), extracellular signal-regulated protein kinase (ERK)-specific inhibitor (PD98059), and transient transfection of cells with dominant-negative (DN)-p38 MAPK and DN-ERK1/ERK2. Vitamin C also induced phosphorylation and enhanced the activities of p38 MAPK and ERK in BLMVECs in a time-dependent fashion. It was also evident that vitamin C induced translocation of PLD(1) and PLD(2), association of p38 MAPK and ERK with PLD(1) and PLD(2), threonine phosphorylation of PLD(1) and PLD(2) and SB203580- and PD98059-inhibitable threonine phosphorylation of PLD(1) in BLMVECs. Transient transfection of BLMVECs with DN-p38 MAPK and DN-ERK1/ERK2 resulted in marked attenuation of vitamin C-induced phosphorylation of threonine in PLD(1) and PLD(2). We, for the first time, showed that vitamin C at pharmacological doses, activated PLD in the lung microvascular ECs through oxidative stress and MAPK activation.


Assuntos
Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfolipase D/metabolismo , Animais , Bovinos , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Ativação Enzimática , Proteínas Quinases Ativadas por Mitógeno/genética , Fosfolipase D/genética , Fosforilação , Inibidores de Proteínas Quinases/metabolismo , Treonina/metabolismo
14.
Blood ; 105(9): 3397-404, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15657182

RESUMO

Mutations in the ELA2 gene encoding neutrophil elastase (NE) are present in most patients with severe congenital neutropenia (SCN). However, the mechanisms by which these mutations cause neutropenia remain unknown. To investigate the effects of mutant NE expression on granulopoiesis, we used the HL-60 promyelocytic cell line retrovirally transduced with the G185R NE mutant that is associated with a severe SCN phenotype. We show that the mutant enzyme accelerates apoptosis of differentiating but not of proliferating cells. Using metabolic labeling, confocal immunofluorescence microscopy, and immunoblot analysis of subcellular fractions, we also demonstrate that the G185R mutant is abnormally processed and localizes predominantly to the nuclear and plasma membranes rather than to the cytoplasmic compartment observed with the wild-type (WT) enzyme. Expression of the G185R mutant appeared to alter the subcellular distribution and expression of adaptor protein 3 (AP3), which traffics proteins from the trans-Golgi apparatus to the endosome. These observations provide further insight into potential mechanisms by which NE mutations cause neutropenia and suggest that abnormal protein trafficking and accelerated apoptosis of differentiating myeloid cells contribute to the severe SCN phenotype resulting from the G185R mutation.


Assuntos
Apoptose/genética , Células Precursoras de Granulócitos/citologia , Elastase de Leucócito/genética , Mutação de Sentido Incorreto , Neutropenia/genética , Complexo 3 de Proteínas Adaptadoras , Compartimento Celular , Diferenciação Celular , Proliferação de Células , Células HL-60 , Humanos , Neutropenia/congênito , Neutropenia/etiologia , Transporte Proteico/genética , Transdução Genética/métodos
15.
J Immunol ; 173(11): 6820-30, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15557176

RESUMO

The Src homology 2-containing inositol phosphatase SHIP1 functions in hemopoietic cells to limit activation events mediated by PI3K products, including Akt activation and cell survival. In contrast to the limited cellular expression of SHIP1, the related isoform SHIP2, is widely expressed in both parenchymal and hemopoietic cells. The goal of this study was to determine how SHIP2 functions to regulate M-CSF signaling. We report that 1) SHIP2 was tyrosine-phosphorylated in M-CSF-stimulated human alveolar macrophages, human THP-1 cells, murine macrophages, and the murine macrophage cell line RAW264; 2) SHIP2 associated with the M-CSF receptor after M-CSF stimulation; and 3) SHIP2 associated with the actin-binding protein filamin and localization to the cell membrane, requiring the proline-rich domain, but not on the Src homology 2 domain of SHIP2. Analyzing the function of SHIP2 in M-CSF-stimulated cells by expressing either wild-type SHIP2 or an Src homology 2 domain mutant of SHIP2 reduced Akt activation in response to M-CSF stimulation. In contrast, the expression of a catalytically deficient mutant of SHIP2 or the proline-rich domain of SHIP2 enhanced Akt activation. Similarly, the expression of wild-type SHIP2 inhibited NF-kappaB-mediated gene transcription. Finally, fetal liver-derived macrophages from SHIP2 gene knockout mice enhanced activation of Akt in response to M-CSF treatment. These data suggest a novel regulatory role for SHIP2 in M-CSF-stimulated myeloid cells.


Assuntos
Fator Estimulador de Colônias de Macrófagos/fisiologia , Monoéster Fosfórico Hidrolases/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Proteínas Contráteis/metabolismo , Regulação para Baixo , Filaminas , Humanos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Células NIH 3T3 , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/fisiologia , Fosforilação , Prolina/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/imunologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais/imunologia , Tirosina/metabolismo , Domínios de Homologia de src/fisiologia
16.
J Immunol ; 173(8): 5036-45, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15470047

RESUMO

Mutations in the G-CSF receptor (G-CSFR) in patients with severe congenital neutropenia (SCN) are postulated to contribute to transformation to acute myelogenous leukemia (AML). These mutations result in defective receptor internalization and sustained cellular activation, suggesting a loss of negative signaling by the G-CSFR. In this paper we investigated the roles of SHIP and cytokine-inducible Src homology 2 protein (CIS) in down-modulating G-CSFR signals and demonstrate that loss of their recruitment as a consequence of receptor mutations leads to aberrant signaling. We show that SHIP binds to phosphopeptides corresponding to Tyr744 and Tyr764 in the G-CSFR and that Tyr764 is required for in vivo phosphorylation of SHIP and the formation of SHIP/Shc complexes. Cells expressing a G-CSFR form lacking Tyr764 exhibited hypersensitivity to G-CSF and enhanced proliferation, but to a lesser degree than observed with the most common mutant G-CSFR form in patients with SCN/AML, prompting us to investigate whether suppressor of cytokine signaling proteins also down-modulate G-CSFR signals. G-CSF was found to induce the expression of CIS and of CIS bound to phosphopeptides corresponding to Tyr729 and Tyr744 of the G-CSFR. The expression of CIS was prolonged in cells with the SCN/AML mutant G-CSFR lacking Tyr729 and Tyr744, which also correlated with increased G-CSFR expression. These findings suggest that SHIP and CIS interact with distal phosphotyrosine residues in the G-CSFR to negatively regulate G-CSFR signaling by limiting proliferation and modulating surface expression of the G-CSFR, respectively. Novel therapeutic approaches targeting inhibitory pathways that limit G-CSFR signaling may have promise in the treatment of patients with SCN/AML.


Assuntos
Proteínas Imediatamente Precoces/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Leucemia Mieloide Aguda/patologia , Neutropenia/congênito , Neutropenia/patologia , Monoéster Fosfórico Hidrolases/fisiologia , Receptores de Fator Estimulador de Colônias de Granulócitos/fisiologia , Proteínas de Transporte/análise , Divisão Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Proteínas do Leite/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Repressoras/análise , Fator de Transcrição STAT5 , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Transativadores/metabolismo , Fatores de Transcrição/análise
17.
Am J Hematol ; 74(3): 149-55, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14587040

RESUMO

Neutrophil elastase (NE) is a serine protease stored in the primary granules of neutrophils that proteolytically cleaves multiple cytokines and cell surface proteins on release from activated neutrophils. Recent reports of mutations in the gene encoding this enzyme in some patients with neutropenic syndromes prompted us to investigate whether granulocyte colony-stimulating factor (G-CSF) and its receptor (G-CSFR) are also substrates for NE. To further address this, we examined the effect of NE on G-CSF and the G-CSFR both in solution and on intact cells. Incubation of recombinant G-CSF or a G-CSFR form corresponding to its extracellular domain with purified NE resulted in rapid proteolytic cleavage of both proteins. Addition of NE to tissue culture medium or pretreatment of G-CSF with NE before its addition to media suppressed the growth of G-CSF-responsive cells. NE also cleaved the G-CSFR on the surface of intact cells resulting in a time-dependent reduction in cell surface expression of the G-CSFR. Notably, decreased G-CSFR surface expression resulting from treatment of cells with NE was also associated with a reduction in cell viability and proliferation in response to G-CSF. These results are the first to demonstrate that G-CSF and G-CSFR are proteolytically cleaved by NE and that NE-induced degradation of these proteins correlates with a reduction in the biologic activity of the cytokine and a decrease in the signaling function of the receptor because of decreased G-CSFR surface expression. These findings provide additional insights into mechanisms by which G-CSF/G-CSFR interactions may be modulated.


Assuntos
Fator Estimulador de Colônias de Granulócitos/metabolismo , Elastase de Leucócito/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Cinética , Elastase de Leucócito/farmacologia , Camundongos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA