Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Oncogene ; 33(39): 4709-21, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-24166497

ABSTRACT

Proteins involved in promoting cell proliferation and viability need to be timely expressed and carefully controlled for the proper development of the brain but also efficiently degraded in order to prevent cells from becoming brain cancer cells. A major pathway for targeted protein degradation in cells is the ubiquitin-proteasome system (UPS). Oncoproteins that drive tumor development and tumor maintenance are often deregulated and stabilized in malignant cells. This can occur when oncoproteins escape degradation by the UPS because of mutations in either the oncoprotein itself or in the UPS components responsible for recognition and ubiquitylation of the oncoprotein. As the pathogenic accumulation of an oncoprotein can lead to effectively sustained cell growth, viability and tumor progression, it is an indisputable target for cancer treatment. The most common types of malignant brain tumors in children and adults are medulloblastoma and glioma, respectively. Here, we review different ways of how deregulated proteolysis of oncoproteins involved in major signaling cancer pathways contributes to medulloblastoma and glioma development. We also describe means of targeting relevant oncoproteins in brain tumors with treatments affecting their stability or therapeutic strategies directed against the UPS itself.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Oncogene Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitination , Animals , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Stability , Proteolysis , Signal Transduction
2.
Leukemia ; 27(5): 1053-62, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23228967

ABSTRACT

Loss of function mutation in FBXW7, an E3 ubiquitin ligase, is associated with good prognosis and early glucocorticoid treatment response in childhood T-cell acute lymphoblastic leukemia (T-ALL) by unknown mechanisms. Here, we show that FBXW7 targets the glucocorticoid receptor α (GRα) for ubiquitylation and proteasomal degradation in a manner dependent on glycogen synthase kinase 3 ß-mediated phsophorylation. FBXW7 inactivation caused elevated GRα levels, and enhanced the transcriptional response to glucocorticoids. There was significant enhancement of GR transcriptional responses in FBXW7-deficient cell lines and primary T-ALL samples, in particular, for those pro-apoptotic regulatory proteins, BIM and PUMA. Reduced FBXW7 expression or function promoted glucocorticoid sensitivity, but not sensitivity to other chemotherapeutic agents used in T-ALL. Moreover, this was a general feature of different cancer cell types. Taken together, our work defines GRα as a novel FBXW7 substrate and demonstrates that favorable patient prognosis in T-ALL is associated with FBXW7 mutations due to enhanced GRα levels and steroid sensitivity. These findings suggest that inactivation of FBXW7, a putative tumor suppressor protein, may create a synthetic lethal state in the presence of specific anticancer therapies.


Subject(s)
Cell Cycle Proteins/physiology , F-Box Proteins/physiology , Glucocorticoids/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Glucocorticoid/drug effects , Ubiquitin-Protein Ligases/physiology , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Line, Tumor , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Humans , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Stability , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/physiology , Ubiquitin-Protein Ligases/genetics
3.
Hum Genet ; 109(5): 542-50, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11735030

ABSTRACT

Rapid progress in the sequencing of the genome of man and other species allows for the comparative analysis of their genetic structure and content. We have used a combined biochemical and computer-based approach to characterize a 146 kb human genomic bacterial artificial chromosome clone from chromosome 5q13 and discovered a novel human elongation-factor gene, hEFG2. The complete human EFG2 cDNA sequence is 3033 bp and contains 21 exons with conserved exon-intron splice junctions encompassing 45 kb of the genomic sequence with its 5'-end residing within a CpG island, characteristic of a housekeeping gene. The complete size of the hEFG2 cDNA was confirmed by Northern blot and reverse transcription/polymerase chain reaction analysis, which showed a single transcript of 3.2 kb ubiquitously expressed in various human tissues. The hEFG2 protein shows significant homology to several bacterial EF-G proteins, including that of Thermus thermophilus, and to the yeast Saccharomyces cerevisiae mitochondrial elongation factor-G ( MEF2). Multiple alignments reveal a novel gene family of mitochondrial EF-G proteins that can by divided into two subgroups, EF-G1 and EF-G2, in several eukaryotic species including S. pombe, Caenorhabditis elegans and Drosophila melanogaster. Using the information contained in the public databases, we also identified and cloned the complete coding sequence of the human EFG1 gene on chromosome 3q25. The cloning and characterization of these human mitochondrial elongation factor genes should permit us to address their role in the regulation of normal mitochondrial function and in various disease states.


Subject(s)
Evolution, Molecular , Mitochondria/genetics , Mitochondrial Proteins , Peptide Elongation Factor G/genetics , Phylogeny , Amino Acid Sequence , Base Sequence , Chromosomes, Artificial, Bacterial , Cloning, Molecular , DNA Primers , DNA, Complementary , Gene Expression , Humans , Molecular Sequence Data , Peptide Elongation Factor G/chemistry , Sequence Homology, Amino Acid
4.
Nature ; 413(6853): 316-22, 2001 Sep 20.
Article in English | MEDLINE | ID: mdl-11565034

ABSTRACT

Cyclin E, one of the activators of the cyclin-dependent kinase Cdk2, is expressed near the G1-S phase transition and is thought to be critical for the initiation of DNA replication and other S-phase functions. Accumulation of cyclin E at the G1-S boundary is achieved by periodic transcription coupled with regulated proteolysis linked to autophosphorylation of cyclin E. The proper timing and amplitude of cyclin E expression seem to be important, because elevated levels of cyclin E have been associated with a variety of malignancies and constitutive expression of cyclin E leads to genomic instability. Here we show that turnover of phosphorylated cyclin E depends on an SCF-type protein-ubiquitin ligase that contains the human homologue of yeast Cdc4, which is an F-box protein containing repeated sequences of WD40 (a unit containing about 40 residues with tryptophan (W) and aspartic acid (D) at defined positions). The gene encoding hCdc4 was found to be mutated in a cell line derived from breast cancer that expressed extremely high levels of cyclin E.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle Proteins/physiology , Cyclin E/metabolism , F-Box Proteins , Mutation , Ubiquitin-Protein Ligases , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Expressed Sequence Tags , F-Box-WD Repeat-Containing Protein 7 , Humans , Molecular Sequence Data , Peptide Synthases/metabolism , Phosphorylation , SKP Cullin F-Box Protein Ligases , Sequence Homology, Amino Acid , Tumor Cells, Cultured , Ubiquitins/metabolism , Yeasts
5.
Blood ; 96(13): 4313-8, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11110707

ABSTRACT

Recently, the derepressed expression of the catalytic subunit of telomerase, human telomerase reverse transcriptase (hTERT), the enzyme that elongates telomeres, has been implicated as an important step in the immortalization process. The exact regulation of hTERT expression, which is the rate-limiting factor for telomerase activity, is at present unclear. As transformed cells seem to be dependent on a constitutive telomerase activity, the availability of inhibitors would potentially be of great value in antineoplastic therapy. Interferons (IFNs) have been successfully used in the treatment of several forms of malignancies, but the underlying molecular mechanisms responsible for the antitumor activity are poorly defined. In this study we have investigated the effects of IFNs on hTERT expression and telomerase activity. We found that IFN-alpha rapidly (commonly within 4 hours) and significantly down-regulates the expression of hTERT and telomerase activity in a number of human malignant hematopoietic cell lines, primary leukemic cells from patients with acute leukemia as well as T-lymphocytes from healthy donors. This effect of IFN-alpha did not seem to depend on IFN-alpha-mediated cell growth arrest or alterations in c-myc expression. The finding that IFN induces a repression of hTERT and a decrease in telomerase activity suggests a novel mechanism that may play a significant role in the antitumor action of IFN. (Blood. 2000;96:4313-4318)


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Interferon-alpha/pharmacology , Neoplasm Proteins/biosynthesis , RNA , T-Lymphocytes/drug effects , Telomerase/biosynthesis , Acute Disease , Burkitt Lymphoma/pathology , Cell Cycle/drug effects , Cycloheximide/pharmacology , DNA Replication/drug effects , DNA, Neoplasm/biosynthesis , DNA-Binding Proteins , Depression, Chemical , Enzyme Induction/drug effects , Humans , Interferon alpha-2 , Leukemia, Myeloid/pathology , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphoma, T-Cell, Cutaneous/pathology , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Recombinant Proteins , T-Lymphocytes/enzymology , Telomerase/genetics , Transcription, Genetic/drug effects , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
6.
Front Biosci ; 5: D479-87, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10762599

ABSTRACT

The interferons (IFNs) are a group of cytokines, which in addition to their antiviral activity are capable of modulating a variety of cellular responses. One such prominent effect of IFNs is their potent antimitogenic action, which can be observed both on malignant and non-malignant cells of many different origins. IFNs are also used in the clinic, mainly in malignant and viral diseases, and their cell growth -inhibitory effect has been suggested to be of major importance in their antitumour and antiviral action. The aim of the present review is to provide insight into the molecular mechanisms by which IFNs modulate cell cycle progression in various cell types. With the recent progress in our understanding of how the cell cycle is regulated at the molecular level, it has become possible to delineate intracellular effectors of IFN in this respect. Understanding the antiproliferative effects of IFN may not only help in understanding its antineoplastic and antiviral activities, but may also provide an insight into cell cycle regulation in general and aid in making IFNs a more useful tool in treating disease.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , Interferons/physiology , Animals , Cell Cycle/physiology , Humans , Signal Transduction/physiology , Tumor Cells, Cultured
7.
Genomics ; 70(3): 327-34, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11161783

ABSTRACT

Previous studies have indicated the presence of a putative tumor suppressor gene on human chromosome 13q14, commonly deleted in patients with B-cell chronic lymphocytic leukemia (B-CLL). We have recently identified a minimally deleted region encompassing parts of two adjacent genes, termed LEU1 and LEU2 (leukemia-associated genes 1 and 2), and several additional transcripts. In addition, 50 kb centromeric to this region we have identified another gene, LEU5/RFP2. To elucidate further the complex genomic organization of this region, we have identified, mapped, and sequenced the homologous region in the mouse. Fluorescence in situ hybridization analysis demonstrated that the region maps to mouse chromosome 14. The overall organization and gene order in this region were found to be highly conserved in the mouse. Sequence comparison between the human deletion hotspot region and its homologous mouse region revealed a high degree of sequence conservation with an overall score of 74%. However, our data also show that in terms of transcribed sequences, only two of those, human LEU2 and LEU5/RFP2, are clearly conserved, strengthening the case for these genes as putative candidate B-CLL tumor suppressor genes.


Subject(s)
Chromosomes, Human, Pair 13 , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Sequence Deletion , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Genes, Tumor Suppressor , Humans , In Situ Hybridization, Fluorescence , Mice , Neoplasm Proteins/genetics
8.
Cell Growth Differ ; 10(8): 575-82, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10470857

ABSTRACT

IFN-alpha exerts prominent regulatory functions on the immune system. One such effect is the inhibition of proliferation of in vitro stimulated T lymphocytes. The exact physiological function of this activity is not known, but it has been implicated in the antiviral effects of IFN, its antitumor action in T-cell malignancies, and the regulation of the in vivo T-cell response. Here, we have investigated the mechanism underlying the IFN-alpha-mediated growth inhibition of normal human PHA- and IL-2-stimulated T lymphocytes by an analysis of how IFN-alpha treatment influences known molecular events that normally accompany the transition from quiescence to proliferation in these cells. IFN-alpha treatment was found to profoundly block S-phase entry of stimulated T lymphocytes. This correlated with a strong inhibition of IL-2-induced changes in G1-regulatory proteins, including the prevented up-regulation of G1 cyclins and cyclin-dependent kinases as well as an abrogation of mitogen-induced reduction of p27Kip1 levels. This latter effect was due to a maintained stability of the p27Kip1 protein in the IFN-alpha-treated cells. In line with these findings, phosphorylation of the pocket proteins was abrogated in IFN-alpha-treated cells. Furthermore, our data indicate that IFN-alpha has selective effects on the pathways that emerge from the IL-2 receptor because IFN-alpha treatment does not block IL-2-induced up-regulation of c-myc or Cdc25A.


Subject(s)
Cell Cycle Proteins/metabolism , Interferon-alpha/pharmacology , Interleukin-2/pharmacology , T-Lymphocytes/drug effects , Tumor Suppressor Proteins , Blotting, Northern , Cell Cycle/drug effects , Cells, Cultured , Cyclin G , Cyclin G1 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Humans , Lymphocyte Activation/drug effects , Microtubule-Associated Proteins/metabolism , Phytohemagglutinins/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-myc/biosynthesis , RNA, Messenger/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Time Factors , Up-Regulation/drug effects , cdc25 Phosphatases/biosynthesis
9.
Oncogene ; 18(18): 2798-810, 1999 May 06.
Article in English | MEDLINE | ID: mdl-10362250

ABSTRACT

One prominent effect of IFNs is their cell growth-inhibitory activity. The mechanism behind this inhibition of proliferation is still not fully understood. In this study, the effect of IFN-alpha treatment on cell cycle progression has been analysed in three lymphoid cell lines, Daudi, U-266 and H9. Examination of the growth-arrested cell populations shows that Daudi cells accumulate in a G0-like state, whereas U-266 cells arrest later in G1. H9 cells are completely resistant to IFN-alpha's cell growth-inhibitory effects. The G0/G1-phase arrest is preceded by a rapid induction of the cyclin-dependent kinase inhibitors (CKIs), p21 and p15. In parallel, the activities of the G1 Cdks are significantly reduced. In addition to p21/p15 induction, IFN-alpha regulates the expression of another CKI, p27, presumably by a post-transcriptional mechanism. In the G1 Cdk-complexes, there is first an increased binding of p21 and p15 to their respective kinases. At longer exposure times, when Cdk-bound p15 and p21 decline, p27 starts to accumulate. Furthermore, we found that IFN-alpha not only suppresses the phosphorylation of pRb, but also alters the phosphorylation and expression of the other pocket proteins p130 and p107. These data suggest that induction of p21/p15 is involved in the primary IFN-alpha response inhibiting G1 Cdk activity, whereas increased p27 expression is part of a second set of events which keep these Cdks in their inactive form. Moreover, elevated levels of p27 correlated with a dissociation of cyclin E/Cdk2-p130 or p107 complexes to yield cyclin E/Cdk2-p27 complexes. In resistant H9 cells, which possess a homozygous deletion of the p15/p16 genes and lack p21 protein expression, IFN-alpha causes no detectable changes in p27 expression and, furthermore, no effects are observed on either pocket proteins in this cell line. Taken together, these data suggest that the early decline in G1 Cdk activity, subsequent changes in phosphorylation of pocket proteins, and G1/G0 arrest following IFN-alpha treatment, is not primarily due to loss of the G1 kinase components, but result from the inhibitory action of CKIs on these complexes.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , G1 Phase/drug effects , Interferon-alpha/pharmacology , Proteins , Proto-Oncogene Proteins , Resting Phase, Cell Cycle/drug effects , Tumor Suppressor Proteins , cdc25 Phosphatases , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/drug effects , Cyclins/drug effects , Cyclins/metabolism , Humans , Interferon-alpha/metabolism , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatases/drug effects , Protein Tyrosine Phosphatases/metabolism , Retinoblastoma Protein/drug effects , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Tumor Cells, Cultured
10.
J Interferon Cytokine Res ; 18(9): 691-5, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9781807

ABSTRACT

Interferon-alpha (IFN-alpha) therapy induces a response in a proportion of patients with metastatic melanoma. However, the mechanism of the antitumor action and reason(s) for resistance to IFN therapy are not known. To investigate whether lack of clinical response may be due to resistance of the melanoma cells to IFN-alpha or to an inability of IFN-alpha to reach the tumor cells during treatment, we investigated the in vivo and in vitro susceptibility of primary tumor cells obtained through fine needle aspiration biopsies and peripheral blood mononuclear cells (PBMC) to the induction of the IFN-induced enzyme 2'5'-oligoadenylate synthetase (2'5'OAS) during initiation of IFN-alpha therapy in 10 patients with metastatic melanoma. None of the patients showed an objective response to IFN-alpha treatment. The melanoma cells from 2 of the 10 patients were resistant to IFN-induced enhancement of 2'5'OAS in vitro. This correlated well with the in vivo induction of 2'5'OAS in the malignant cells, as no in vivo induction was seen in the 2 patients whose malignant cells were resistant in vitro, whereas tumor cells from 7 of 8 of the remaining patients showed enhancement also in vivo. This study shows that it is possible to monitor the cellular susceptibility of tumor cells to IFN-alpha in vivo and that melanoma cells from a small percentage of patients are resistant to the cellular effects of IFN-alpha. Furthermore, the absence of a clinical response to IFN-alpha therapy in the majority of melanoma patients can be explained neither by impaired cellular susceptibility to IFN nor by an inability of IFN-alpha to reach the tumor.


Subject(s)
2',5'-Oligoadenylate Synthetase/biosynthesis , Antineoplastic Agents/therapeutic use , Interferon-alpha/therapeutic use , Leukocytes, Mononuclear/drug effects , Melanoma/drug therapy , Adult , Aged , Enzyme Induction , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Melanoma/metabolism , Melanoma/secondary , Middle Aged , Stimulation, Chemical , Treatment Outcome , Tumor Cells, Cultured
11.
Oncogene ; 17(5): 595-602, 1998 Aug 06.
Article in English | MEDLINE | ID: mdl-9704925

ABSTRACT

Little is known about the molecular background to senescence in T-lymphocytes. In fibroblast systems replicative senescence has been shown to correlate with a number of changes in the expression of the proteins normally regulating progression through the G1 phase of the cell cycle, and recently the Ink4 inhibitor p16 was implicated as a central regulator of replicative senescence in human fibroblasts. It has, however, been claimed that p16 is not expressed in T-lymphocytes. In the present study we have analysed G1 regulating proteins in ageing human T-lymphocytes. We show that PHA and IL-2 stimulated T-lymphocytes cease to proliferate after around 20 population doublings, these cells can not thereafter be restimulated to growth, and were also found to exhibit markers for senescence. We found that T-lymphocytes accumulate p16 and p15 protein during successive population doublings and display high levels of these proteins as they enter into replicative senescence. There was also an increased binding of p16 to the Cdk6 kinase in senescent cells, and a decreased Cdk6 as well as Cdk2 kinase activity. The levels of other G1 regulating proteins were also altered in the senescent cells, such as slightly elevated levels of p21/WAF1, and downregulation of Cdk2 and cyclinD3. The levels of p27/ Kip1 is down regulated in proliferating cells but rise to approximately 15% of the levels in un-stimulated quiescent cells. As a high proportion of T-cell childhood acute lymphoblastic leukaemias have deletions of both p15 and p16, our data suggest that inactivation of these genes makes it possible for leukemic cells to avoid senescence.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Proteins , T-Lymphocytes/metabolism , Tumor Suppressor Proteins , CD28 Antigens/biosynthesis , Cell Division , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinases/metabolism , G1 Phase , Humans , Interleukin-2/pharmacology , Lymphocyte Activation , Phosphoproteins/metabolism , Phosphorylation , Phytohemagglutinins/pharmacology , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p130 , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology
12.
Eur J Haematol ; 59(3): 129-35, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9310119

ABSTRACT

The interferons (IFNs) have become accepted therapy in a range of haematological and non-haematological malignancies. The mechanism behind IFN's antitumour action is, however, unclear. Interferons (IFNs) are capable of modulating a variety of cellular responses. One prominent effect of IFNs is their cell growth inhibitory activity, which has also been suggested to be of major importance in their antitumour action. In the present review we will discuss the cellular events leading to a decreased number of cells following IFN treatment, the molecular mechanisms underlying these phenomena, and the importance of these effects in a clinical perspective.


Subject(s)
Cell Division/drug effects , Interferons/pharmacology , Apoptosis , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Eukaryotic Cells/physiology , Gene Expression Regulation , Interferons/physiology
13.
Cell Growth Differ ; 8(3): 343-52, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9056677

ABSTRACT

IFNs are capable of modulating a variety of cellular responses, including cell growth and apoptosis. The prospective connections between these two biological responses are not fully understood, and the molecular mechanisms underlying the effects of IFNs on these processes are not completely defined. We have investigated the relationship between IFN-alpha-induced apoptosis and cell cycle arrest in three hematopoietic cell lines, Daudi, U-266, and H9. It was found that IFN-alpha was a rapid and potent inducer of apoptosis in H9 and U-266 cells, whereas IFN-alpha-induced cell cycle arrest in Daudi cells is not associated with the onset of apoptosis. In H9 cells, apoptosis occurs without a preceding cell cycle block, whereas in U-266 cells, apoptosis occurs subsequent to G1 arrest. Cell cycle arrest per se, induced by serum starvation or treatment with aphidicolin, had only minor effects on the viability of these cell lines and did not abrogate the apoptosis-inducing capacity of IFN-alpha. Additionally, IFN-alpha-induced apoptosis occurred in cells from all cell cycle phases. Thus, we conclude that IFN-alpha-induced apoptosis seems to occur independent of cell growth inhibition. There were no changes in Bcl-2 or Bax protein levels that could account for the apoptosis-inducing effects of IFN-alpha in these cell lines. Moreover, examination of p53 status suggests that IFN-alpha-induced apoptosis in the U-266 and H9 cell lines occurs through a p53-independent pathway.


Subject(s)
Apoptosis , Hematopoietic Stem Cells/drug effects , Interferon-alpha/pharmacology , Bromodeoxyuridine/analysis , Cell Cycle/drug effects , Cell Division/drug effects , Cell Survival/drug effects , DNA/analysis , Flow Cytometry , Humans , Microscopy, Fluorescence , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
14.
Oncogene ; 14(4): 415-23, 1997 Jan 30.
Article in English | MEDLINE | ID: mdl-9053838

ABSTRACT

One prominent effect of IFNs is their cell growth inhibitory activity. The exact molecular mechanism behind this inhibition of proliferation remains to be elucidated. Possible effectors for IFN-induced growth inhibition are the recently discovered cyclin-dependent kinase inhibitors. The effect of IFN-alpha treatment on the members of the Ink4 and Cip/Kip families of Cdk inhibitors was investigated in three hematopoietic cell lines Daudi, U-266 and H9. Two of these cell lines, Daudi and U-266, respond to IFN-alpha by G1 arrest, whereas the H9 cell line is not growth arrested by IFN-alpha. We show that a p53-independent upregulation of p21 mRNA occurs following IFN-alpha treatment in all three cell lines. In Daudi and U-266 cells, the mRNA induction is accompanied by an increase in p21 protein, followed by an increased binding of p21 to Cdk2 and a subsequent decrease in Cdk2 activity, temporally coinciding with G1 arrest. In both these cell lines, there was also an increased binding of p21 to Cdk4. In contrast, p21 protein was not expressed in H9 cells, despite high levels of p21 mRNA following IFN-alpha treatment. In U-266 cells, IFN-alpha increased not only p21 but also p15 mRNA and protein levels, followed by an increased association of p15 with Cdk4. Furthermore, IFN-alpha treatment caused a four- to sixfold induction of the p16 E1beta transcript in U-266 cells. Expression levels of the other Ink4 and Cip/Kip Cdk inhibitors were not induced by IFN treatment in any of the cell lines. We conclude that IFN-alpha can act as a potent regulator of Cdk-inhibitor expression, correlating with decreased Cdk activity and cell growth inhibition. One mechanism for resistance to IFN may be loss of the ability of cells to upregulate these proteins.


Subject(s)
Blood Proteins/biosynthesis , Carrier Proteins/biosynthesis , Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors , Interferon-alpha/pharmacology , Microtubule-Associated Proteins/biosynthesis , Protein Kinase Inhibitors , Transcription, Genetic/drug effects , Tumor Suppressor Proteins , Cell Line , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p27 , DNA Primers , DNA, Neoplasm/biosynthesis , Hematopoietic Stem Cells , Humans , Interferon alpha-2 , Kinetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Recombinant Proteins , Thymidine/metabolism , Tumor Cells, Cultured
15.
Int J Cancer ; 67(1): 106-12, 1996 Jul 03.
Article in English | MEDLINE | ID: mdl-8690509

ABSTRACT

The tumor suppressor p53 plays a central role in negative growth control, including growth arrest and apoptosis. Interferons (IFNs) are capable of modulating a variety of cellular responses, including apoptosis. In this study, we have evaluated the influence of gamma- and alpha-interferon (IFN) on wild-type (wt) p53-induced apoptosis using a Burkitt lymphoma cell line, BL41, transfected with a temperature-sensitive p53 construct, gamma-IFN, but not alpha-IFN, was found to protect cells from wt p53-induced apoptosis. The gamma-IFN-dependent protection was due neither to down-regulation of p53, nor to the p53-induced genes, p21 (WAF-1) and bax, nor to up-regulation of bcl-2 or bcl-xL. Expression of the proto-oncogene c-myc, implicated in the control of both proliferation and apoptosis, was not affected by gamma-IFN. We conclude that gamma-IFN can suppress p53-induced apoptosis, and that the cytokine microenvironment may be decisive in the cellular response to wt p53 expression.


Subject(s)
Apoptosis/drug effects , Burkitt Lymphoma/pathology , Interferon-gamma/pharmacology , Tumor Suppressor Protein p53/physiology , Humans , Proto-Oncogene Mas , Transfection , Tumor Cells, Cultured
16.
Scand J Gastroenterol ; 31(6): 604-11, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8789901

ABSTRACT

BACKGROUND: The mechanism behind the antiviral action of interferon (IFN) therapy in chronic hepatitis C virus (HCV) infection is not well understood, and, furthermore, few factors have been shown to be good predictors of a favourable response to IFN treatment in chronic HCV infection. METHODS: Freshly explanted liver cells and peripheral blood mononuclear cells (PBMC) from 80 patients with chronic HCV infection were used to study the capacity of IFN to induce the enzyme 2',5'-oligoadenylate synthetase (2'5'-AS) in vitro. The HCV genotype was determined in 53 patients. The induction of 2'5'-AS was correlated to the results of IFN-alpha treatment in 36 patients. RESULTS: Normalization of transaminases during IFN treatment was significantly associated with 2'5'-AS levels in liver cells cultured in the absence of IFN. A similar tendency, although not statistically significant, was found for IFN-induced levels of 2'5'-AS in liver cells. No such associations were found when PBMC were analysed. Six patients showed a sustained biochemical response. These six did not deviate significantly from the remaining patients with regard to base-line or IFN-induced levels of 2'5'-AS in liver cells or PBMC. Eradication of HCV RNA during IFN treatment did not correlate with 2'5'-AS levels in liver cells. Comparison of HCV genotype and clinical response showed that patients with genotype 3a had the most favourable outcome. No association was found between liver histology and treatment outcome. CONCLUSION: These data imply that direct effects of IFN on liver cells are of importance for the response to IFN treatment.


Subject(s)
2',5'-Oligoadenylate Synthetase/biosynthesis , Antiviral Agents/therapeutic use , Hepacivirus/genetics , Hepatitis C/therapy , Hepatitis, Chronic/therapy , Interferon-alpha/therapeutic use , Leukocytes, Mononuclear/enzymology , Liver/enzymology , Cells, Cultured , Enzyme Induction , Genotype , Hepatitis, Chronic/virology , Humans , Interferon alpha-2 , Liver/pathology , Recombinant Proteins
17.
Int J Cancer ; 63(2): 190-2, 1995 Oct 09.
Article in English | MEDLINE | ID: mdl-7591202

ABSTRACT

The expression of the Bcl-2 oncoprotein was studied in pre-treatment bone-marrow samples from 63 patients with multiple myeloma, using an immunohistochemistry technique. A variable expression of the Bcl-2 protein was found in myeloma cells. 43% of the patients had strong expression of the Bcl-2 protein in the malignant cells. Forty patients received alpha-interferon, whereas 23 patients received melphalan/prednisone therapy. A significant association (p = 0.012) was found between high levels of Bcl-2 expression in myeloma cells and resistance to interferon therapy. No such correlation was found in the melphalan/prednisone treated patients. The data indicate that over-expression of Bcl-2 may be a cause for resistance to interferon therapy in myeloma and that staining for Bcl-2 expression in myeloma cells may have a predictive value for this treatment.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Interferon-alpha/therapeutic use , Melphalan/administration & dosage , Multiple Myeloma/drug therapy , Prednisone/administration & dosage , Proto-Oncogene Proteins/metabolism , Humans , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2
18.
Blood ; 84(6): 1942-9, 1994 Sep 15.
Article in English | MEDLINE | ID: mdl-8080997

ABSTRACT

Cells from one-third of chronic lymphocytic leukemia (CLL) patients are resistant to alpha-interferon (alpha-IFN) as measured by induction of blast transformation. We have previously shown that all CLL clones express alpha/beta-IFN receptors, but that the resistant cells are defective in the induction of the enzyme 2',5'-oligoadenylate synthetase (2',5-A synthetase). Thus, the deficiency in IFN sensitivity is localized somewhere between the interaction of the IFN molecule with its receptor and induction of 2',5'-A synthetase. We have now further characterized the resistance of CLL clones to IFN by investigating whether it is associated with a defect in the activation of IFN-stimulated gene factor 3 (ISGF3), which is involved in the activation of alpha-IFN-stimulated genes (ISGs). A defect induction of ISGF3 after alpha-IFN treatment was found in 4 of 12 CLL patients. There was a close correlation between defective induction of ISGF3 and a lack of enhancement of 2',5'-A synthetase as well as induction of blast transformation. Pretreatment with gamma-IFN and mixing experiments with extracts from IFN-sensitive cells indicate that a lack of the gamma-component of ISGF3 was the reason for defect in activation in 2 of the patients. We conclude that a defect in activation of ISGF3 is a possible cause for resistance in CLL cells to IFN-induced blast transformation in vitro.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon-alpha/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Transcription Factors/metabolism , 2',5'-Oligoadenylate Synthetase/biosynthesis , Base Sequence , Cell Nucleus/metabolism , Cell Survival , Cytoplasm/metabolism , Drug Resistance , Enzyme Induction , Humans , Interferon alpha-2 , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Interferon-gamma/pharmacology , Kinetics , Lymphocyte Activation , Molecular Sequence Data , Recombinant Proteins , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...