Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Ophthalmol ; 93(2): 126-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25488348

ABSTRACT

PURPOSE: The HELIOS (Health Economics with Lucentis in Observational Settings) study was designed on request of the Dutch Health Authority for an observational study to assess the effectiveness and safety of ranibizumab for neovascular age-related macular degeneration (wet AMD) in daily practice. METHODS: The HELIOS study was a 2-year prospective, observational, open-label, multicentre study involving 14 sites. Patients with wet AMD were enrolled and observed for a period of 24 months. The data were collected at baseline and at the visits closest around the time-points 3, 6, 12, 18 and 24 months after inclusion. RESULTS: Treatment with ranibizumab resulted in prevention of vision loss. The mean ETDRS score increased from 45.1 letters at baseline to 48.5 letters at 24 months. This was achieved with a mean of 7.8 injections over 24 months. Stabilization of visual acuity was also reflected by the scores on the quality of life EQ-5D questionnaire, which did not significantly change over the study period. The more subjective EQ-VAS questionnaire showed an overall improvement. The VFQ-25 questionnaire was also mostly stable over time. After 24 months, 32.2% of the patients gained ≥1 letter and 17.1% gained >15 letters. Patients completing the loading phase were better responders, as demonstrated by increased long-term visual acuity. In addition, ranibizumab was well tolerated and had a safety profile commonly seen in routine clinical practice. CONCLUSION: This study demonstrates that also in daily practice ranibizumab was effective in preventing vision loss over a period of 24 months. No new safety findings were identified.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Wet Macular Degeneration/drug therapy , Aged , Aged, 80 and over , Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Female , Humans , Intravitreal Injections , Male , Prospective Studies , Quality of Life/psychology , Ranibizumab , Sickness Impact Profile , Surveys and Questionnaires , Tomography, Optical Coherence , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vision Disorders/prevention & control , Visual Acuity/physiology , Wet Macular Degeneration/physiopathology , Wet Macular Degeneration/psychology
2.
J Biol Chem ; 282(4): 2211-20, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17132628

ABSTRACT

Leukemic transformation often requires activation of protein kinase B (PKB/c-Akt) and is characterized by increased proliferation, decreased apoptosis, and a differentiation block. PKB phosphorylates and inactivates members of the FOXO subfamily of Forkhead transcription factors. It has been suggested that hyperactivation of PKB maintains the leukemic phenotype through actively repressing FOXO-mediated regulation of specific genes. We have found expression of the transcriptional repressor Id1 (inhibitor of DNA binding 1) to be abrogated by FOXO3a activation. Inhibition of PKB activation or growth factor deprivation also resulted in strong down-regulation of Id1 promoter activity, Id1 mRNA, and protein expression. Id1 is highly expressed in Bcr-Abl-transformed K562 cells, correlating with high PKB activation and FOXO3a phosphorylation. Inhibition of Bcr-Abl by the chemical inhibitor STI571 resulted in activation of FOXO3a and down-regulation of Id1 expression. By performing chromatin immunoprecipitation assays and promoter-mutation analysis, we demonstrate that FOXO3a acts as a transcriptional repressor by directly binding to the Id1 promoter. STI571 treatment, or expression of constitutively active FOXO3a, resulted in erythroid differentiation of K562 cells, which was inhibited by ectopic expression of Id1. Taken together our data strongly suggest that high expression of Id1, through PKB-mediated inhibition of FOXO3a, is critical for maintenance of the leukemic phenotype.


Subject(s)
Cell Differentiation , Cell Transformation, Neoplastic , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Genes, abl , Inhibitor of Differentiation Protein 1/genetics , Animals , Base Sequence , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation/genetics , Cell Line, Transformed , Down-Regulation , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Humans , Inhibitor of Differentiation Protein 1/metabolism , K562 Cells , Leukemia/genetics , Leukemia/pathology , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Transcription, Genetic
3.
Cancer Res ; 66(22): 10760-9, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108112

ABSTRACT

Increased protein kinase B (PKB; c-Akt) activation is a hallmark of diverse neoplasias providing both proliferative and antiapoptotic survival signals. In this study, we investigated the effect of chronic PKB activation on cellular survival and proliferation using cytokine-dependent bone marrow-derived Ba/F3 cells, in which PKBalpha activation can be directly, and specifically, induced by addition of 4-hydroxytamoxifen (4-OHT). Direct activation of PKB rescued Ba/F3 cells from cytokine withdrawal-induced apoptosis; however, surprisingly, these antiapoptotic effects were short lived, cells only being protected for up to 48 hours. We observed that activation of PKB in survival factor-deprived cells led to a dramatic increase of Foxo3a on both the transcriptional and protein level leading to expression of its transcriptional targets Bim and p27(kip1). High levels of PKB activity result in increased aerobic glycolysis and mitochondrial activity resulting in overproduction of reactive oxygen species. To determine whether oxidative stress might itself be responsible for Foxo3a up-regulation, we utilized hydrogen peroxide (H(2)O(2)) as an artificial inducer of oxidative stress and N-acetylcysteine (NAC), a thiol-containing radical oxygen scavenger. Addition of NAC to the culture medium prolonged the life span of cells treated with 4-OHT and prevented the up-regulation of Foxo3a protein levels caused by PKB activation. Conversely, treatment of Ba/F3 cells with H(2)O(2) caused an increase of Foxo3a on both transcriptional and protein levels, suggesting that deregulated PKB activation leads to oxidative stress resulting in Foxo3a up-regulation and subsequently cell death. Taken together, our data provide novel insights into the molecular consequences of uncontrolled PKB activation.


Subject(s)
Apoptosis/physiology , Forkhead Transcription Factors/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/genetics , Enzyme Activation , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Oxidative Stress/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Reactive Oxygen Species/metabolism , Transcription, Genetic , Up-Regulation
4.
FEBS Lett ; 579(1): 191-8, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15620712

ABSTRACT

Phosphatidylinositol 3-kinase (PI3K) and its effector protein kinase B (PKB/c-akt) have been implicated as critical mediators of cytokine-induced survival signals. In this study, we have utilized an IL-5 dependent hematopoietic cell line (TF-1) to investigate the signaling events involved in cytokine-dependent erythroblast survival. We demonstrate that IL-5 rescues TF-1 cells from apoptosis through a PI3K/PKB-dependent signaling pathway. Cytokine-withdrawal leads to activation of the Forkhead transcription factor FOXO3a and subsequent expression of the pro-apoptotic Bcl-2 family member Bim. Bim is itself sufficient to induce apoptosis in these cells. Importantly, activation of an inducible active FOXO3a mutant is alone sufficient for upregulation of Bim expression and induction of apoptosis. These data define a mechanism by which survival factors inhibit the default apoptotic pathway and can regulate TF-1 erythroblast survival.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Erythroblasts/metabolism , Interleukin-5/physiology , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors , Glycogen Synthase Kinase 3/physiology , Humans , Mutation/genetics , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Mol Cell Biol ; 24(22): 10058-71, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509806

ABSTRACT

Cell cycle arrest by FoxO transcription factors involves transcriptional repression of cyclin D, although the exact mechanism remains unclear. In this study, we used the BCR-ABL-expressing cell line BV173 as a model system to investigate the mechanisms whereby FoxO3a regulates cyclin D2 expression. Inhibition of BCR-ABL by STI571 results in down-regulation of cyclin D2 expression, activation of FoxO3a activity, and up-regulation of BCL6 expression. Using reporter gene assays, we demonstrate that STI571, FoxO3a, and BCL6 can repress cyclin D2 transcription through a STAT5/BCL6 site located within the cyclin D2 promoter. We propose that BCR-ABL inhibition leads to FoxO3a activation, which in turn induces the expression of BCL6, culminating in the repression of cyclin D2 transcription through this STAT5/BCL6 site. This process was verified by mobility shift and chromatin immunoprecipitation analyses. We find that conditional activation of FoxO3a leads to accumulation of BCL6 and down-regulation of cyclin D2 at protein and mRNA levels. Furthermore, silencing of FoxO3a and BCL6 in BCR-ABL-expressing cells abolishes STI571-mediated effects on cyclin D2. This report establishes the signaling events whereby BCR-ABL signals are relayed to cyclin D2 to mediate cell cycle progression and defines a potential mechanism by which FoxO proteins regulate cyclin D2 expression.


Subject(s)
Cyclins/genetics , DNA-Binding Proteins/metabolism , Fusion Proteins, bcr-abl/metabolism , Milk Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Base Sequence , Benzamides , Binding Sites/genetics , Cell Line , Cyclin D2 , Cyclins/metabolism , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Milk Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Piperazines/pharmacology , Promoter Regions, Genetic , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-6 , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , STAT5 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic/drug effects
6.
Exp Cell Res ; 299(2): 533-40, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15350550

ABSTRACT

Bcl-2 family proteins regulate apoptosis at the level of mitochondria. To examine the mechanism of Bcl-2 function, we investigated the effects of the protonophore carbonyl cyanide m-chlorophenyl hydrazone (CCCP) on two hematopoietic cell lines and Bcl-2 overexpressing transfectants. CCCP directly interferes with mitochondrial function and induces apoptosis. We show that Bcl-2 inhibits apoptosis and that the antiapoptotic effect of Bcl-2 takes place upstream of caspase activation and nuclear changes associated with apoptosis, since these were markedly inhibited in cells overexpressing Bcl-2. Bcl-2 does not prevent the decrease in mitochondrial membrane potential nor the alterations in cellular ATP content induced by CCCP in FL5.12 and Jurkat cells. A higher number of mitochondria was observed in untreated Bcl-2 transfected cells compared to parental cells, as shown by electron microscopy. Exposure to CCCP induced a dramatic decrease in the number of mitochondria and severely disrupted mitochondrial ultrastructure, with apparent swelling and loss of cristae in parental cells. Bcl-2 clearly diminished the disruption of mitochondrial structure and preserved a higher number of mitochondria. These data suggest that CCCP induces apoptosis by structural disruption of mitochondria and that Bcl-2 prevents apoptosis and mitochondrial degeneration by preserving mitochondrial integrity.


Subject(s)
Apoptosis/drug effects , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Adenosine Triphosphate/metabolism , Animals , Caspases/metabolism , Cell Nucleus , Enzyme Activation/drug effects , Humans , Jurkat Cells , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Membrane Potentials/drug effects , Mice , Mitochondria/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/pharmacology
7.
J Immunol ; 171(4): 1623-9, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12902457

ABSTRACT

Regulation of phosphatidylinositol 3-kinase (PI3K) activity has been demonstrated to be critical for correct lymphocyte function. The molecular targets of this lipid kinase have been the subject of extensive research, and many functional effects of PI3K activation are thought to be mediated by the serine-threonine kinase protein kinase B (PKB/c-akt). Genetic analyses in the nematode worm Caenorhabditis elegans have identified a novel PI3K-regulated signaling pathway that regulates organism lifespan through inhibition of a Forkhead (FOX) transcription factor, DAF-16. Recent studies have subsequently revealed an evolutionarily conserved signaling module in higher eukaryotes in which PKB can directly phosphorylate and inactive a family of Forkhead box class O (FOXO) transcription factors. Phosphorylation results in nuclear exclusion and inhibition of transcription. FOXO transcription factors have been found to play critical roles in regulation of proliferation, apoptosis and control of oxidative stress. This occurs through both activation and repression of target gene expression by multiple mechanisms. Here the regulation and function of these transcription factors is discussed with specific relevance to immune homeostasis. A greater understanding of the regulation and function of this signaling pathway in lymphocytes may provide novel therapeutic opportunities for immune diseases.


Subject(s)
Apoptosis/immunology , DNA-Binding Proteins , Drosophila Proteins/physiology , Homeostasis/immunology , Nerve Tissue Proteins , Nuclear Proteins/physiology , Signal Transduction/immunology , Transcription Factors/physiology , Animals , Cell Survival/immunology , Drosophila Proteins/antagonists & inhibitors , Forkhead Box Protein O1 , Forkhead Transcription Factors , Nuclear Proteins/antagonists & inhibitors , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/physiology , Transcription Factors/antagonists & inhibitors
8.
J Cell Biol ; 156(3): 531-42, 2002 Feb 04.
Article in English | MEDLINE | ID: mdl-11815629

ABSTRACT

Survival signals elicited by cytokines include the activation of phosphatidylinositol 3-kinase (PI3K), which in turn promotes the activation of protein kinase B (PKB). Recently, PKB has been demonstrated to phosphorylate and inactivate forkhead transcription factor FKHR-L1, a potent inducer of apoptosis. To explore the mechanisms underlying the induction of apoptosis after cytokine withdrawal or FKHR-L1 activation, we used a cell line in which FKHR-L1 activity could be specifically induced. Both cytokine withdrawal and FKHR-L1 activation induced apoptosis, which was preceded by an upregulation in p27KIP1 and a concomitant decrease in cells entering the cell cycle. Induction of apoptosis by both cytokine withdrawal and activation of FKHR-L1 correlated with the disruption of mitochondrial membrane integrity and cytochrome c release. This was preceded by upregulation of the pro-apoptotic Bcl-2 family member Bim. Ectopic expression of an inhibitory mutant of FKHR-L1 substantially reduced the levels of apoptosis observed after cytokine withdrawal. Activation of PKB alone was sufficient to promote cell survival, as measured by maintenance of mitochondrial integrity and the resultant inhibition of effector caspases. Furthermore, hematopoietic stem cells isolated from Bim-/- mice exhibited reduced levels of apoptosis upon inhibition of PI3K/PKB signaling. These data demonstrate that activation of FKHR-L1 alone can recapitulate all known elements of the apoptotic program normally induced by cytokine withdrawal. Thus PI3K/PKB--mediated inhibition of this transcription factor likely provides an important mechanism by which survival factors act to prevent programmed cell death.


Subject(s)
Cell Death/physiology , Cell Survival/physiology , Cytokines/deficiency , DNA-Binding Proteins/metabolism , Membrane Proteins , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Annexin A5/metabolism , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Carrier Proteins/metabolism , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p27 , Cytochrome c Group/metabolism , Cytokines/pharmacology , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Male , Membrane Potentials/physiology , Mice , Mice, Knockout , Mitochondria/drug effects , Phosphoinositide-3 Kinase Inhibitors , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Proteins/metabolism , Proto-Oncogene Proteins c-akt , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...