Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Br J Cancer ; 103(1): 1-5, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20461086

ABSTRACT

Originally identified as the enzymes responsible for catalysing the oxidation of specific, conserved proline residues within hypoxia-inducible factor-1alpha (HIF-1alpha), the additional roles for the prolyl hydroxylase domain (PHD) proteins have remained elusive. Of the four identified PHD enzymes, PHD2 is considered to be the key oxygen sensor, as knockdown of PHD2 results in elevated HIF protein. Several recent studies have highlighted the importance of PHD2 in tumourigenesis. However, there is conflicting evidence as to the exact role of PHD2 in tumour angiogenesis. The divergence seems to be because of the contribution of stromal-derived PHD2, and in particular the involvement of endothelial cells, vs tumour-derived PHD2. This review summarises our current understanding of PHD2 and tumour angiogenesis, focusing on the influences of PHD2 on vascular normalisation and neovascularisation.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic/etiology , Procollagen-Proline Dioxygenase/physiology , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases , Neoplasm Metastasis
2.
Cell Death Differ ; 15(4): 678-85, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18259193

ABSTRACT

Hypoxia-inducible factors (HIFs) are essential mediators of the cellular oxygen-signaling pathway. They are heterodimeric transcription factors consisting of an oxygen-sensitive alpha subunit (HIF-alpha) and a constitutive beta subunit (HIF-beta) that facilitate both oxygen delivery and adaptation to oxygen deprivation by regulating the expression of genes that control glucose uptake, metabolism, angiogenesis, erythropoiesis, cell proliferation, and apoptosis. In most experimental models, the HIF pathway is a positive regulator of tumor growth as its inhibition often results in tumor suppression. In clinical samples, HIF is found elevated and correlates with poor patient prognosis in a variety of cancers. In summary, HIF regulates multiple aspects of tumorigenesis, including angiogenesis, proliferation, metabolism, metastasis, differentiation, and response to radiation therapy, making it a critical regulator of the malignant phenotype.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia/metabolism , Neoplasms/metabolism , Signal Transduction , Adaptation, Physiological , Animals , Cell Differentiation , Cell Hypoxia , Cell Proliferation , Energy Metabolism , Humans , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/radiotherapy , Neovascularization, Pathologic/metabolism , Oxygen/metabolism
3.
Science ; 284(5411): 156-9, 1999 Apr 02.
Article in English | MEDLINE | ID: mdl-10102818

ABSTRACT

The ability of p53 to promote apoptosis in response to mitogenic oncogenes appears to be critical for its tumor suppressor function. Caspase-9 and its cofactor Apaf-1 were found to be essential downstream components of p53 in Myc-induced apoptosis. Like p53 null cells, mouse embryo fibroblast cells deficient in Apaf-1 and caspase-9, and expressing c-Myc, were resistant to apoptotic stimuli that mimic conditions in developing tumors. Inactivation of Apaf-1 or caspase-9 substituted for p53 loss in promoting the oncogenic transformation of Myc-expressing cells. These results imply a role for Apaf-1 and caspase-9 in controlling tumor development.


Subject(s)
Apoptosis , Caspases/physiology , Genes, p53 , Neoplasms, Experimental/pathology , Proteins/physiology , Animals , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspases/genetics , Cell Division , Cell Transformation, Neoplastic , Cells, Cultured , Cytochrome c Group/metabolism , Genes, myc , Genes, ras , Mice , Mice, Nude , Mitochondria/metabolism , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Proteins/genetics , Tumor Suppressor Protein p53/metabolism
4.
Mol Cell Biol ; 21(4): 1196-206, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158306

ABSTRACT

We investigated the role of the cyclin-dependent kinase inhibitors p21(Cip1) and p27(Kip1) in cell cycle regulation during hypoxia and reoxygenation. While moderate hypoxia (1 or 0.1% oxygen) does not significantly impair bromodeoxyuridine incorporation, at very low oxygen tensions (0.01% oxygen) DNA replication is rapidly shut down in immortalized mouse embryo fibroblasts. This S-phase arrest is intact in fibroblasts lacking the cyclin kinase inhibitors p21(Cip1) and p27(Kip1), indicating that these molecules are not essential elements of the arrest pathway. Hypoxia-induced arrest is accompanied by dephosphorylation of pRb and inhibition of cyclin-dependent kinase 2, which results in part from inhibitory phosphorylation. Interestingly, cells lacking the retinoblastoma tumor suppressor protein also display arrest under hypoxia, suggesting that pRb is not an essential mediator of this response. Upon reoxygenation, DNA synthesis resumes by 3.5 h and reaches aerobic levels by 6 h. Cells lacking p21, however, resume DNA synthesis more rapidly upon reoxygenation than wild-type cells, suggesting that this inhibitor may play a role in preventing premature reentry into the cell cycle upon cessation of the hypoxic stress. While p27 null cells did not exhibit rapid reentry into the cell cycle, cells lacking both p21 and p27 entered S phase even more aggressively than those lacking p21 alone, revealing a possible secondary role for p27 in this response. Cdk2 activity is also restored more rapidly in the double-knockout cells when returned to normoxia. These studies reveal that restoration of DNA synthesis after hypoxic stress, but not the S phase arrest itself, is regulated by p21 and p27.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cell Cycle/physiology , Cell Hypoxia/physiology , Cyclins/metabolism , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , Animals , Cells, Cultured , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , DNA/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
5.
Mol Cell Biol ; 14(9): 6264-77, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8065358

ABSTRACT

It has been convincingly demonstrated that genotoxic stresses cause the accumulation of the tumor suppressor gene p53. One important consequence of increased p53 protein levels in response to DNA damage is the activation of a G1-phase cell cycle checkpoint. It has also been shown that G1-phase cell cycle checkpoints are activated in response to other stresses, such as lack of oxygen. Here we show that hypoxia and heat, agents that induce cellular stress primarily by inhibiting oxygen-dependent metabolism and denaturing proteins, respectively, also cause an increase in p53 protein levels. The p53 protein induced by heat is localized in the cytoplasm and forms a complex with the heat shock protein hsc70. The increase in nuclear p53 protein levels and DNA-binding activity and the induction of reporter gene constructs containing p53 binding sites following hypoxia occur in cells that are wild type for p53 but not in cells that possess mutant p53. However, unlike ionizing radiation, the accumulation of cells in G1 phase by hypoxia is not strictly dependent on wild-type p53 function. In addition, cells expressing the human papillomavirus E6 gene, which show increased degradation of p53 by ubiquitination and fail to accumulate p53 in response to DNA-damaging agents, do increase their p53 levels following heat and hypoxia. These results suggest that hypoxia is an example of a "nongenotoxic" stress which induces p53 activity by a different pathway than DNA-damaging agents.


Subject(s)
Cell Cycle , HSP70 Heat-Shock Proteins , Hot Temperature , Hypoxia/metabolism , Repressor Proteins , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Carrier Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , HSC70 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Humans , In Vitro Techniques , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Oncogene Proteins, Viral/metabolism , Transcriptional Activation
6.
Mol Cell Biol ; 18(12): 7371-82, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9819424

ABSTRACT

The putative function of highly conserved regions (HCRs) within 3' untranslated regions (3'UTRs) as regulatory RNA sequences was efficiently and quantitatively assessed by using modular retroviral vectors. This strategy led to the identification of HCRs that alter gene expression in response to oxidative or mitogenic stress. Databases were screened for UTR sequences of >100 nucleotides that had retained 70% identity over more than 300 million years of evolution. The effects of 10 such HCRs on a standard reporter mRNA or protein were studied. To this end, we developed a modular retroviral vector that can allow for a direct comparison of the effects of different HCRs on gene expression independent of their gene-intrinsic 5'UTR, promoter, protein coding region, or poly(A) sequence. Five of the HCRs tested decreased mRNA steady-state levels 2- to 10-fold relative to controls, presumably by altering mRNA stability. One HCR increased translation, and one decreased translation. Elevated mitogen levels caused four HCRs to increase protein levels twofold. One HCR increased protein levels fourfold in response to hypoxia. Although nonconserved UTR sequences may also have a role, these results provide evidence that sequences that are highly conserved during evolution are good candidates for RNA motifs with posttranscriptional regulatory functions in gene expression.


Subject(s)
3' Untranslated Regions/genetics , Conserved Sequence/genetics , RNA/genetics , Stress, Physiological , Animals , Biological Evolution , Cell Line , Flow Cytometry , Gene Expression Regulation/genetics , Genes, Regulator/genetics , Genes, Reporter/genetics , Hypoxia/genetics , Mice , Mitogens/pharmacology , Protein Biosynthesis , RNA, Messenger/genetics , Retroviridae/genetics
7.
Cancer Res ; 55(23): 5512-9, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7585624

ABSTRACT

Exposure of NIH3T3 cells to elevated temperatures induces the phosphorylation and activation of mitogen-activated protein (MAP) kinases [or extracellular signal-regulated kinases (ERKs)]. To investigate the significance of MAP kinase activation by heat shock, we examined the effect of inhibiting the activity of MAP kinase on heat shock protein 70 (hsp 70) expression. Overexpression of a dominant inhibitory mutant of ERK1, but not ERK2, in heat-shocked cells increased hsp70 reporter gene activity, suggesting that ERK1 acts as a repressor of hsp70 gene expression. Increases in ERK1 activity through treatment of cells with sodium vanadate (SV), an inhibitor of the dual-specificity MAP kinase phosphatase 1 (PAC1), resulted in increased phosphorylation of the heat shock transcription factor-1 (HSF-1) in unheated cells, delayed the activation of HSF-1 by heat shock, and inhibited the induction of hsp 70 by heat shock. Furthermore, the induction of thermotolerance was reduced significantly in cells that increased ERK1 activity by SV pretreatment. Immune complex kinase assays of heat shocked or SV-pretreated cells indicated that HSF-1 is a potential in vivo substrate for ERK1 phosphorylation. Taken together, these results suggest that agents that modulate MAP kinase act as negative regulators of the heat shock response in mammalian cells by modulating HSF-1 activity and hsp 70 expression.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , DNA-Binding Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mitogen-Activated Protein Kinases , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Activation , Gene Expression Regulation , Genes, ras , Heat Shock Transcription Factors , Hot Temperature , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Molecular Sequence Data , Phosphorylation/drug effects , Transcription Factors , Vanadates/pharmacology
8.
Cancer Res ; 58(7): 1408-16, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9537241

ABSTRACT

The physiology of solid tumors differs from that of normal tissues in a number of important aspects, the majority of which stem from differences between the two vasculatures. Compared with the regular, ordered vasculature of normal tissues, blood vessels in tumors are often highly abnormal, distended capillaries with leaky walls and sluggish flow. Tumor growth also requires continuous new vessel growth, or angiogenesis. These physiological differences can be problems for cancer treatment; for example, hypoxia in solid tumors leads to resistance to radiotherapy and to some anticancer drugs. However, these differences can also be exploited for selective cancer treatment. Here we review four such areas that are under active investigation: (a) hypoxia-selective cytotoxins take advantage of the unique low oxygen tension in the majority of human solid tumors. Tirapazamine, a drug in the final stages of clinical trials, is one of the more promising of these agents; (b) leaky tumor blood vessels can be exploited using liposomes that have been sterically stabilized to have a long intravascular half-life, allowing them to selectively accumulate in solid tumors; (c) the tumor microenvironment is a stimulus to angiogenenesis, and inhibition of angiogenesis can be a powerful anticancer therapy not susceptible to acquired drug resistance; and (d) we discuss attempts to use gene therapy activated either by the low oxygen environment or by necrotic regions of tumors.


Subject(s)
Neoplasms/physiopathology , Neoplasms/therapy , Cell Hypoxia , Humans , Neoplasms/blood supply
9.
Cancer Res ; 54(6): 1425-30, 1994 Mar 15.
Article in English | MEDLINE | ID: mdl-8137243

ABSTRACT

The response of mammalian cells to stress is controlled by transcriptional regulatory proteins such as nuclear factor kappa B (NF-kappa B) to induce a wide variety of early response genes. In this report, we show that exposure of cells to hypoxia (0.02% O2) results in I kappa B alpha degradation, increased NF-kappa B DNA binding activity, and transactivation of a reporter gene construct containing two NF-kappa B DNA binding sites. Pretreatment of cells with protein tyrosine kinase inhibitors and the dominant negative allele of c-Raf-1 (Raf 301) inhibited I kappa B alpha degradation, NF-kappa B binding, and transactivation of kappa B reporter constructs by hypoxia. To demonstrate a direct link between changes in the phosphorylation pattern of I kappa B alpha with NF-kappa B activation, we immunoprecipitated I kappa B alpha after varying times of hypoxic exposure and found that its tyrosine phosphorylation status increased during hypoxic exposure. Inhibition of the transfer of tyrosine phosphoryl groups onto I kappa B alpha prevented I kappa B alpha degradation and NF-kappa B binding. In comparison to other activators of NF-kappa B such as phorbol myristate acetate or tumor necrosis factor, we did not detect changes in the tyrosine phosphorylation status of I kappa B alpha following treatment with either of these agents. These results suggest that tyrosine phosphorylation of I kappa B alpha during hypoxia is an important proximal step which precedes its dissociation and degradation from NF-kappa B.


Subject(s)
Cell Hypoxia/physiology , DNA-Binding Proteins/metabolism , I-kappa B Proteins , NF-kappa B/metabolism , NF-kappa B/physiology , Tyrosine/metabolism , Base Sequence , Cell Nucleus/physiology , Cells, Cultured , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Kinetics , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Oncogene Protein p65(gag-jun)/genetics , Phosphorylation , Protein-Tyrosine Kinases/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation/genetics , Translocation, Genetic/genetics , Tumor Necrosis Factor-alpha/pharmacology
10.
Cancer Res ; 60(16): 4638-44, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969818

ABSTRACT

Although p53 inactivation is implicated as a mechanism to explain diminished apoptotic response, it is clear that tumor cells that possess transcriptionally functional p53 can also be resistant to diverse apoptotic stimuli. We hypothesize that oncogenic activation and DNA damage are sufficient stimuli to increase the p53-dependent transcription of Fas and thereby establish a situation in which cell to cell contact could be a selective pressure to either lose p53 function or inactivate components of the Fas death pathway. Examination of genetically matched tumor cell lines that possessed either wild-type or null p53 loci indicated that cells possessing functional p53 increased their surface levels of Fas and Fas ligand (FasL) in response to DNA damage. In contrast, stress induced by changes in the tumor microenvironment such as decreased oxygen did not up-regulate Fas or FasL. Cells with wild-type p53 underwent Fas-mediated killing in the presence of either FasL-expressing killer cells or activating Fas antibodies, whereas cells in which p53 was deleted or inactivated were protected from such killing. Furthermore, Fas and FasL expression and induction became transcriptionally repressed in transformed cells with wild-type p53 with increasing passage, whereas other p53 downstream targets and functions, such as p21 inducibility and cell cycle arrest, remained intact. Repression of the Fas locus could be reverted by treatment with the histone deacetylase inhibitor trichostatin A. These results support a model of tumor progression in which oncogenic transformation drives tumor cells to lose either p53 or their Fas sensitivity as a means of promoting their survival and evade immune surveillance.


Subject(s)
Apoptosis/physiology , Tumor Suppressor Protein p53/physiology , fas Receptor/biosynthesis , Animals , Cell Hypoxia/physiology , Cell Line, Transformed , Cell Transformation, Neoplastic/pathology , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Gene Expression Regulation, Neoplastic/physiology , Gene Expression Regulation, Neoplastic/radiation effects , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Kinetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred MRL lpr , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , fas Receptor/genetics , fas Receptor/physiology
11.
Cancer Res ; 61(6): 2429-33, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289110

ABSTRACT

Hypoxia initiates numerous intracellular signaling pathways important in regulating cell proliferation, differentiation, and death. In this study, we investigated the pathway that hypoxia uses to activate Akt and inactivate glycogen synthase kinase-3 (GSK-3), two proteins the functions of which are important in cell survival and energy metabolism. Severe hypoxia (0.01% oxygen) initiated a signaling cascade by inducing the tyrosine phosphorylation of the platelet-derived growth factor (PDGF) receptor within 1 h of treatment and increasing receptor association with the p85 subunit of phosphatidylinositol 3-kinase (PI 3-K). Hypoxia-induced signaling also resulted in PI 3-K-dependent phosphorylation of Akt on Ser-473, a modification of Akt that is important for its activation. This activation of Akt by hypoxia was substantially diminished in cells that possessed mutations in their PDGF receptor-PI 3-K interaction domain. In addition, Akt activation by hypoxia was resistant to treatment with the growth factor receptor poison suramin but was sensitive to treatment with the PI 3-K inhibitor wortmannin. Activation of Akt by hypoxia resulted in the phosphorylation of GSK-3alpha and GSK-3beta at Ser-9 and Ser-21, two well-documented Akt phosphorylation sites, respectively, that are inactivating modifications of each GSK-3 isoform. In support of the phosphorylation data, GSK-3 activity was significantly reduced under hypoxia. In conclusion, we propose that hypoxia activates a growth factor receptor/PI 3-K/Akt cascade that leads to GSK-3 inactivation, a pathway that can impact cell survival, proliferation, and metabolism.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Oxygen/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/physiology , Androstadienes/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Hypoxia/physiology , Cell Line , Dogs , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Fibrosarcoma/enzymology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Kidney/cytology , Kidney/enzymology , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Tumor Cells, Cultured , Tyrosine/metabolism , Wortmannin
12.
Cancer Res ; 57(21): 4703-6, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9354425

ABSTRACT

Cellular checkpoints are important mediators of the response of normal cells following genotoxic damage, and interruption of these checkpoints is a common feature of many solid tumors. Although the effects of loss in checkpoint function in tumor cells are well understood in terms of cell cycle control, there is little information on their role in determining treatment efficacy in vivo. We have examined both the in vitro and in vivo responses of isogenic lines differing only in the p53-transactivated checkpoint gene, p21Waf1/Cip1. When assayed in vitro, loss of p21 in human colon tumor cells results in a selective induction of apoptosis [Waldman, T., et al., Nature (Lond.), 381: 713-716, 1996.] but no difference in the clonogenic survival. However, when grown as xenografts and irradiated in situ, p21-deficient tumors were significantly more sensitive to radiation as assessed both by clonogenic survival and by regrowth of the tumors following treatment. These data indicate that loss of p21 results in increased sensitivity to killing by ionizing radiation that is independent of the induction of apoptosis and cell cycle arrest but that is specific to cells when they are grown as a solid tumor. These results have important implications for assessing both the genetic determinants of sensitivity to anticancer agents and efficacy of anticancer agents.


Subject(s)
Apoptosis , Colonic Neoplasms/radiotherapy , Cyclins/genetics , Gene Deletion , Animals , Apoptosis/genetics , Cell Hypoxia , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , G1 Phase/genetics , G2 Phase/genetics , Humans , Mice , Mice, SCID , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
13.
Cancer Res ; 54(20): 5273-9, 1994 Oct 15.
Article in English | MEDLINE | ID: mdl-7923153

ABSTRACT

We have previously shown that hypoxia causes the activation of nuclear factor-kappa B (NF-kappa B), and the phosphorylation of its inhibitory subunit, I kappa B alpha, on tyrosine residues. With the use of dominant negative mutants of Ha-Ras and Raf-1, we investigated some of the early signaling events leading to the activation of NF-kappa B by hypoxia. Both dominant negative alleles of Ha-Ras and Raf-1 inhibited NF-kappa B induction by hypoxia, suggesting that the hypoxia-induced pathway of NF-kappa B induction is dependent on Ras and Raf-1 kinase activity. Furthermore, although conditions of low oxygen can also activate mitogen-activated protein kinases (ERK1 and ERK2), these kinases do not appear to be involved in regulating NF-kappa B by low oxygen conditions, as dominant negative mutants of mitogen-activated protein kinase do not inhibit NF-kappa B activation by hypoxia. Since Ras and Raf-1 have been previously shown to work downstream from membrane-associated tyrosine kinases such as Src, we determined if the Src membrane-associated kinase was also activated by low oxygen conditions. We detected an increase in Src proto-oncogene activity within 15-30 min of cellular exposure to hypoxia. We postulate that Src activation by hypoxia may be one of the earliest events that precedes Ras activation in the signaling cascade which ultimately leads to the phosphorylation and dissociation of the inhibitory subunit of NF-kappa B, I kappa B alpha.


Subject(s)
Cell Hypoxia , Genes, ras/physiology , Mitogen-Activated Protein Kinases , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , ras Proteins/metabolism , 3T3 Cells , Animals , CSK Tyrosine-Protein Kinase , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Genes, src , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Proto-Oncogene Proteins c-raf , Signal Transduction , src-Family Kinases
14.
Cancer Res ; 55(17): 3726-31, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7543816

ABSTRACT

The multifocal origin of prostate cancer suggests a pan-organ defect in a tumor suppressor pathway. Although structural mutations in the p53 gene have been implicated in late-stage prostate cancer, little is known about the p53 response to genotoxic stress in normal human prostatic epithelial cells from which adenocarcinomas originate. We found that the majority (10 of 12) of epithelial cell cultures derived from histologically normal tissues of radical prostatectomy specimens failed to exhibit p53 accumulation in response to ionizing radiation. Epithelial cell cultures derived from benign prostatic hyperplasia and a primary prostatic adenocarcinoma also failed to accumulate p53 in response to ionizing radiation. In contrast, cultures of prostatic stromal cells derived from normal, benign prostatic hyperplasia, or adenocarcinoma tissues exhibited a 3-9-fold induction of p53 within 1-3 h after irradiation. Since p53 regulates a cell cycle checkpoint through the induction of the cyclin-cdk inhibitor p21, we examined p21 accumulation and cell cycle arrest following exposure to ionizing radiation. With one exception, epithelial cells that did not display increased p53 or p21 induction did not demonstrate a significant G1-S arrest in response to ionizing radiation, whereas stromal cells that accumulated p53 and p21 exhibited a large cell cycle arrest. These results indicate a functional difference between the DNA damage response of epithelial and stromal prostatic cells and suggest a possible mechanism for the increased susceptibility of prostatic epithelial cells to accumulate genetic alterations.


Subject(s)
Adenocarcinoma/metabolism , Cyclins/metabolism , DNA Damage , G1 Phase/radiation effects , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , S Phase/radiation effects , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Epithelium/metabolism , Epithelium/radiation effects , Humans , Male , Phosphorylation , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinases/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
15.
Cancer Res ; 56(15): 3436-40, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8758908

ABSTRACT

Hypoxia can select for cells that have lost their apoptotic potential, thereby making them resistant to adverse conditions. However, long-term survival of transformed cells which have diminished apoptotic sensitivity when exposed to low oxygen conditions would require the activation of their angiogenic program to compensate for an insufficient oxygen supply. In this report, we show that the activity (of oncogenic Ha-ras, either constitutively or transiently, enhances the induction of the angiogenic mitogen, vascular endothelial growth factor (VEGF), by hypoxia. Analysis of the 5' flanking region of the VEGF promoter indicates that a HIF-1-like sequence is to promote a 15-fold increase in reporter gene activity in Ha-ras-transformed cells when exposed to hypoxia, whereas mutations in the same site totally inhibited VEGF induction. Under low oxygen conditions, VEGF induction is inhibited in cells expressing a mutant inhibitory allele of Ha-ras (RasN17), indicating a direct role for Ras in modulating VEGF activity. We propose that the angiogenic switch in Ras-transformed cells may be physiologically promoted by the tumor microenvironment through VEGF induction.


Subject(s)
Cell Transformation, Neoplastic/genetics , Endothelial Growth Factors/biosynthesis , Genes, ras , Lymphokines/biosynthesis , 3T3 Cells/metabolism , 3T3 Cells/physiology , Animals , Cell Hypoxia/physiology , Endothelial Growth Factors/genetics , Gene Expression , Lymphokines/genetics , Mice , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Rats , Stress, Physiological/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , ras Proteins/biosynthesis , ras Proteins/physiology
16.
Cancer Res ; 56(19): 4315-9, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8813114

ABSTRACT

We have developed an animal tumor model system to study the effects of c-Myc activation on apoptosis induction in vivo. Tumors were generated in SCID mice from Rat-1 fibroblasts that constitutively express an inactive c-Myc-estrogen receptor fusion protein (T.D. Littlewood et al, Nucleic Acids Res., 23: 1686 -1690, 1995), which is activated in vivo by the administration of 4-hydroxytamoxifen in time release pellets. We demonstrate that activation of c-Myc results in a substantial increase in the number of apoptotic tumor cells and that this apoptosis is predominant in regions of tumor hypoxia. c-Myc-induced apoptosis of hypoxic cells is inhibited in tumors that overexpress the human Bcl-2 protein. Bcl-2, however, does not prevent p53 protein accumulation or the down-regulation of the cyclin-cdk inhibitor p27 protein following c-Myc activation by 4-hydroxytamoxifen. This result suggests that Bcl-2 does not affect c-Myc function directly but acts downstream of c-Myc to inhibit apoptosis. We propose that the ability of activated c-Myc to enhance cellular proliferation might contribute to the genesis of early neoplasms that are held in check by the alternate ability of c-Myc to induce apoptosis of cells that have outgrown their supply of oxygen or other factors associated with hypoxic regions of solid tumors. Secondary genetic lesions downstream of c-Myc that suppress the apoptotic potential of tumor cells, such as Bcl-2 overexpression, might play an important role in the malignant progression of these tumors because they would disrupt the balance between apoptosis and proliferation initiated by c-Myc deregulation.


Subject(s)
Apoptosis/genetics , Cell Cycle Proteins , Genes, myc , Proto-Oncogene Proteins c-myc/physiology , Tumor Suppressor Proteins , Animals , Cell Hypoxia , Cyclin-Dependent Kinase Inhibitor p27 , Estrogen Antagonists/pharmacology , Fibroblasts/pathology , Fibroblasts/transplantation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, SCID , Microscopy, Fluorescence , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins c-myc/biosynthesis , Rats , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
17.
Cancer Res ; 58(9): 1779-84, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9581811

ABSTRACT

Currently, the contribution of cellular apoptotic sensitivity to tumor response after radiation therapy remains controversial. To address this issue, the survival of Rat-1 fibroblasts containing a 4-hydroxytamoxifen-regulated c-Myc allele, c-MycER (T. D. Littlewood et al., Nucleic Acids Res., 23: 1686-1690, 1995), after single and fractionated doses of radiation was investigated. This model system allows pharmacological regulation of apoptosis sensitivity in the same cells in vitro and as xenograft tumors derived from these cells in vivo (G. I. Evan et al., Cell, 69: 119-128, 1992; R. M. Alarcon et al., Cancer Res., 56: 4315-4319, 1996). Activating c-MycER in vitro resulted in marked sensitization of Rat-1 fibroblasts to the effects of both single-dose and fractionated irradiation as measured by the induction of apoptosis and clonogenic survival. Overexpression of the antiapoptosis protein Bcl-2 suppressed the induction of apoptosis and increased clonogenic survival in cells with activated c-Myc after single-dose and fractionated radiation. Systemic time-release implant delivery of 4-hydroxytamoxifen to severe combined immunodeficient mice bearing Rat-1-MycER tumors over the course of either single-dose (10 Gy) or fractionated (five fractions of 2 Gy) radiotherapy resulted in prolonged tumor growth delay relative to identical tumors from mice that received placebo implants. Furthermore, tumors derived from Rat-1-MycER cells that overexpressed Bcl-2 exhibited shorter tumor growth delays relative to similarly treated Rat-1-MycER tumors. The length of tumor growth delay after single-dose or fractionated radiotherapy strongly correlated with the extent of radiation-induced apoptosis in the xenograft tumors as measured by terminal deoxynucleotidyl transferase-mediated nick end labeling. These in vivo results provide direct evidence that increasing the sensitivity of tumor cells to die by apoptosis increases the efficacy of fractionated radiotherapy by reducing tumor cell clonogenic survival.


Subject(s)
Apoptosis/radiation effects , Neoplasms, Experimental/radiotherapy , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Combined Modality Therapy , DNA Fragmentation , Dose Fractionation, Radiation , Estrogen Antagonists/pharmacology , Fibroblasts/metabolism , Male , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Radiation, Ionizing , Rats , Severe Combined Immunodeficiency/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
18.
Cancer Res ; 59(24): 6046-51, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10626788

ABSTRACT

Hypoxia, a result of DNA-damaging agents such as ionizing radiation, induces the nuclear accumulation of the p53 tumor suppressor protein. However, unlike the effect in ionizing radiation, hypoxia readily induces the nuclear accumulation of p53 in HPV E6-infected cells. In HPV-infected cells, a key regulator of p53 protein levels is the E6 oncoprotein. In association with the endogenous cellular protein E6-associated protein (E6AP), E6 can accelerate the degradation of p53 under aerobic conditions. To better define the mechanism of p53 induction in E6-infected cells by hypoxia, we studied the expression and association of E6 and E6AP with p53 in vivo. We found that hypoxia did not alter the protein levels of E6 or E6AP as compared with those found under aerobic growth conditions, indicating that protein inhibition of E6 or E6AP alone is not sufficient to explain the increased accumulation of p53 under hypoxic conditions. However, p53 did fail to coprecipitate with E6AP under hypoxia, indicating that hypoxia uncouples the interaction of p53 with E6 and E6AP. We also present evidence to indicate that hypoxia decreases the expression of the endogenous cellular regulator of p53 protein, the human MDM2 protein, resulting in an inhibition of p53 export from the nucleus to the cytoplasm for degradation. Taken together, these results suggest that the hypoxic induction of p53 is attributable to the down-regulation of MDM2 protein levels and uncoupling of p53 from its interaction with the E6/E6AP complex.


Subject(s)
Ligases/metabolism , Nuclear Proteins , Oncogene Proteins, Viral/metabolism , Oxygen/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Tumor Suppressor Protein p53/metabolism , Cell Hypoxia , Down-Regulation , Humans , Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF , Ubiquitin-Protein Ligases
19.
Cancer Res ; 51(10): 2636-41, 1991 May 15.
Article in English | MEDLINE | ID: mdl-2021941

ABSTRACT

Since mammalian cells vary widely in their intrinsic thermoresistance, we have investigated the genetic basis underlying this phenomenon in human and rodent cell lines. Typically, human cells are considerably more resistant to killing by heat than rodent cell lines. To determine whether the heat-resistant phenotype is dominant or recessive and to locate the chromosome(s) bearing determinants for heat resistance, we have prepared hybrids of heat-resistant human HT1080 cells and heat-sensitive Chinese hamster ovary (CHO) cells to test their response to heat. For both mass hybrid cultures and individual clones, the heat response of the hybrids was similar to that of the CHO parent. Analysis by in situ hybridization revealed the presence of five to 20 human chromosomes per cell in the mass hybrids and four to eight intact chromosomes plus some fragments in individual clones isolated from the hybrid cell population. A similar result was obtained using a different human cell line, AG1522. These data suggest that heat resistance is a recessive trait. Consistent with this conclusion are the results from a study of a fusion of HT1080 to a CHO mutant, BL-10, which was found to be hypersensitive to heat-induced killing. These hybrids had a normal CHO heat response and not the more heat-resistant phenotype of HT1080 cells. Two hybrid clones, H2 and H4, from the HT1080/BL-10 fusion were studied in more detail. Both clones possess similar amounts of Mr 70,000 heat shock protein (HSP70), despite the fact that H4 contains three human chromosomes (Nos. 6, 14, and 21) which carry HSP70 genes while H2 contains only one (chromosome 6). Both hybrid cell lines have the same response to heat. Although we found a wide range of sensitivities to heat, all cell lines contained a similar amount of constitutive HSP70, suggesting that HSP70 levels per se are not the critical determinant of intrinsic heat resistance.


Subject(s)
Heat-Shock Proteins/biosynthesis , Hybrid Cells/physiology , Animals , Autoradiography , Cell Fusion , Cell Line , Cell Survival , Chromosomes, Human , Cricetinae , Cricetulus , Electrophoresis, Gel, Two-Dimensional , Heat-Shock Proteins/isolation & purification , Hot Temperature , Humans , Hybrid Cells/cytology , Karyotyping , Methionine/metabolism , Molecular Weight , Sulfur Radioisotopes
20.
Cancer Res ; 51(16): 4463-9, 1991 Aug 15.
Article in English | MEDLINE | ID: mdl-1714344

ABSTRACT

As a means to understand the fundamental mechanisms of bleomycin cell killing, we previously isolated 19 bleomycin-sensitive mutants which represent at least six genetically distinct complementation groups (T.D. Stamato, B. Peters, P. Patil, N. Denko, R. Weinstein, and A. Giaccia. Cancer Res., 47: 1588-1592, 1987). One class of mutants represented by the cell line BL-10 displays only hypersensitivity to killing by bleomycin in both acute (16 h) and chronic treatments but no sensitivity to killing by other DNA-damaging agents. Complementation studies between this mutant and human fibroblasts suggested that the human gene which corrects the defect of BL-10 rested on human chromosome 6. It has been reported that the gene for human glutathione S-transferase (GST) alpha also resides on chromosome 6. Measurements of selenium-independent peroxidase (alpha-GST + glutathione peroxidase) activity in wild-type Chinese hamster ovary (CHO) cells, using cumene hydrogen peroxide as a substrate, gave a value of 112 nmol of glutathione oxidized/min/mg protein compared with 88.1 nmol of glutathione oxidized/min/mg protein for BL-10. Measurement of the selenium-dependent peroxidase activity, using H2O2 as a substrate, resulted in 65.9 nmol of reduced glutathione oxidized/min/mg protein in CHO and 81.5 nmol of reduced glutathione oxidized/min/mg protein for BL-10. In other words, BL-10 cells did not exhibit a difference in their ability to metabolize both substrates in contrast to CHO cells. This indicates that BL-10 possesses little alpha-GST activity. Transfection of BL-10 cells with a mammalian expression vector containing the alpha-GST gene increases the survival of BL-10 to bleomycin and does not increase the bleomycin resistance of two other bleomycin mutants which lie in different genetic complementation groups. These data strongly implicate a role for alpha-GST in the resistance of cells to bleomycin.


Subject(s)
Bleomycin/pharmacology , Cell Survival/drug effects , Chromosomes, Human, Pair 6 , Genetic Variation , Glutathione Transferase/genetics , Hybrid Cells/cytology , Animals , Cell Line , Chromosome Banding , Clone Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Resistance/genetics , Female , Genetic Complementation Test , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Humans , Hybrid Cells/drug effects , Ovary
SELECTION OF CITATIONS
SEARCH DETAIL