Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(21): 2687-97, 2016 05.
Article in English | MEDLINE | ID: mdl-26364599

ABSTRACT

Antiangiogenic therapy resistance occurs frequently in patients with metastatic renal cell carcinoma (RCC). The purpose of this study was to understand the mechanism of resistance to sunitinib, an antiangiogenic small molecule, and to exploit this mechanism therapeutically. We hypothesized that sunitinib-induced upregulation of the prometastatic MET and AXL receptors is associated with resistance to sunitinib and with more aggressive tumor behavior. In the present study, tissue microarrays containing sunitinib-treated and untreated RCC tissues were stained with MET and AXL antibodies. The low malignant RCC cell line 786-O was chronically treated with sunitinib and assayed for AXL, MET, epithelial-mesenchymal transition (EMT) protein expression and activation. Co-culture experiments were used to examine the effect of sunitinib pretreatment on endothelial cell growth. The effects of AXL and MET were evaluated in various cell-based models by short hairpin RNA or inhibition by cabozantinib, the multi-tyrosine kinases inhibitor that targets vascular endothelial growth factor receptor, MET and AXL. Xenograft mouse models tested the ability of cabozantinib to rescue sunitinib resistance. We demonstrated that increased AXL and MET expression was associated with inferior clinical outcome in patients. Chronic sunitinib treatment of RCC cell lines activated both AXL and MET, induced EMT-associated gene expression changes, including upregulation of Snail and ß-catenin, and increased cell migration and invasion. Pretreatment with sunitinib enhanced angiogenesis in 786-0/human umbilical vein endothelial cell co-culture models. The suppression of AXL or MET expression and the inhibition of AXL and MET activation using cabozantinib both impaired chronic sunitinib treatment-induced prometastatic behavior in cell culture and rescued acquired resistance to sunitinib in xenograft models. In summary, chronic sunitinib treatment induces the activation of AXL and MET signaling and promotes prometastatic behavior and angiogenesis. The inhibition of AXL and MET activity may overcome resistance induced by prolonged sunitinib therapy in metastatic RCC.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Renal Cell/drug therapy , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Pyrroles/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Mice , Mice, Nude , Molecular Targeted Therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Sunitinib , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
2.
Oncogene ; 33(9): 1093-100, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-23435419

ABSTRACT

Metastatic melanoma remains a devastating disease with a 5-year survival rate of less than five percent. Despite recent advances in targeted therapies for melanoma, only a small percentage of melanoma patients experience durable remissions. Therefore, it is critical to identify new therapies for the treatment of advanced melanoma. Here, we define connective tissue growth factor (CTGF) as a therapeutic target for metastatic melanoma. Clinically, CTGF expression correlates with tumor progression and is strongly induced by hypoxia through HIF-1 and HIF-2-dependent mechanisms. Genetic inhibition of CTGF in human melanoma cells is sufficient to significantly reduce orthotopic tumor growth, as well as metastatic tumor growth in the lung of severe combined immunodeficient (SCID) mice. Mechanistically, inhibition of CTGF decreased invasion and migration associated with reduced matrix metalloproteinase-9 expression. Most importantly, the anti-CTGF antibody, FG-3019, had a profound inhibitory effect on the progression of established metastatic melanoma. These results offer the first preclinical validation of anti-CTGF therapy for the treatment of advanced melanoma and underscore the importance of tumor hypoxia in melanoma progression.


Subject(s)
Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/genetics , Melanoma/drug therapy , Melanoma/genetics , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Hypoxia/genetics , Hypoxia-Inducible Factor 1/genetics , Matrix Metalloproteinase 9/genetics , Melanoma/pathology , Mice , Mice, SCID
3.
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
4.
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
5.
Clin Cancer Res ; 10(14): 4806-12, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15269156

ABSTRACT

PURPOSE: Our study was undertaken to determine the utility of plasma proteomic profiling using surface-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) mass spectrometry for the detection of head and neck squamous cell carcinomas (HNSCCs). EXPERIMENTAL DESIGN: Pretreatment plasma samples from HNSCC patients or controls without known neoplastic disease were analyzed on the Protein Biology System IIc SELDI-TOF mass spectrometer (Ciphergen Biosystems, Fremont, CA). Proteomic spectra of mass:charge ratio (m/z) were generated by the application of plasma to immobilized metal-affinity-capture (IMAC) ProteinChip arrays activated with copper. A total of 37356 data points were generated for each sample. A training set of spectra from 56 cancer patients and 52 controls were applied to the "Lasso" technique to identify protein profiles that can distinguish cancer from noncancer, and cross-validation was used to determine test errors in this training set. The discovery pattern was then used to classify a separate masked test set of 57 cancer and 52 controls. In total, we analyzed the proteomic spectra of 113 cancer patients and 104 controls. RESULTS: The Lasso approach identified 65 significant data points for the discrimination of normal from cancer profiles. The discriminatory pattern correctly identified 39 of 57 HNSCC patients and 40 of 52 noncancer controls in the masked test set. These results yielded a sensitivity of 68% and specificity of 73%. Subgroup analyses in the test set of four different demographic factors (age, gender, and cigarette and alcohol use) that can potentially confound the interpretation of the results suggest that this model tended to overpredict cancer in control smokers. CONCLUSIONS: Plasma proteomic profiling with SELDI-TOF mass spectrometry provides moderate sensitivity and specificity in discriminating HNSCC. Further improvement and validation of this approach is needed to determine its usefulness in screening for this disease.


Subject(s)
Head and Neck Neoplasms/blood , Neoplasms, Squamous Cell/blood , Proteome/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Female , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/metabolism , Humans , Male , Middle Aged , Neoplasms, Squamous Cell/diagnosis , Neoplasms, Squamous Cell/metabolism , Reproducibility of Results , Sensitivity and Specificity
6.
Gene Ther ; 9(4): 291-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11896468

ABSTRACT

A major obstacle in cancer gene therapy is selective tumor delivery. Previous studies have suggested that genetically engineered anaerobes of the genus Clostridium might be gene therapy vectors because of their ability to proliferate selectively in the hypoxic/necrotic regions common to solid tumors. However, the tumor colonization efficiency of the strain previously used was insufficient to produce any antitumor effect. Here we describe for the first time the successful transformation of C. sporogenes, a clostridial strain with the highest reported tumor colonization efficiency, with the E. coli cytosine deaminase (CD) gene and show that systemically injected spores of these bacteria express CD only in the tumor. This enzyme can convert the nontoxic prodrug 5-fluorocytosine (5-FC) to the anticancer drug 5-fluorouracil (5-FU). Furthermore, systemic delivery of 5-FC into mice previously injected with CD-transformed spores of C. sporogenes produced greater antitumor effect than maximally tolerated doses of 5-FU. Since most human solid tumors have hypoxic and necrotic areas this vector system has considerable promise for tumor-selective gene therapy.


Subject(s)
Carcinoma, Squamous Cell/therapy , Clostridium/genetics , Gene Targeting/methods , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Animals , Carcinoma, Squamous Cell/pathology , Cell Hypoxia , Clostridium/enzymology , Cytosine Deaminase , Flucytosine/therapeutic use , Fluorouracil/therapeutic use , Mice , Mice, Inbred C3H , Necrosis , Neoplasm Transplantation , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , Prodrugs/therapeutic use
7.
Novartis Found Symp ; 240: 115-28; discussion 128-32, 2001.
Article in English | MEDLINE | ID: mdl-11727925

ABSTRACT

The p53 tumour suppressor gene is capable of activating both death receptor and mitochondrial-signalled forms of apoptotic cell death in response to diverse stimuli. Studies have suggested that impairment of the mitochondrial-signalled Apaf/caspase 9 pathway and not the death receptor Fas pathway results in almost complete resistance to apoptotic cell death induced by a low oxygen environment. However, it is unclear how p53 signals the activation of this pathway and whether it is through already identified p53 effector genes such as the pro-apoptotic gene bax, or through novel effectors such as BNIP-3/BNIP-3L. Comparison of cell lines genetically matched at the bax, cytochrome c, apaf, caspase 9 and caspase 3 loci indicated that except for bax, all of these genes were essential for hypoxia induced apoptosis both in cell culture and in transplanted tumours. These data imply that cytochrome c plays a pivotal role in signalling cell death by apoptosis under hypoxic conditions, and that the release of cytochrome c is independent of both Bax and p53. In contrast to cytochrome c, p53 modulates the magnitude of apoptosis under hypoxic conditions, but in itself is not required for the activation of the caspase cascade.


Subject(s)
Apoptosis/physiology , Cell Hypoxia/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Proto-Oncogene Proteins c-bcl-2 , Aerobiosis , Animals , Caspase 9 , Caspases/metabolism , Cell Line , Cytochrome c Group/metabolism , Embryo, Mammalian , Mice , Proto-Oncogene Proteins/genetics , Tumor Necrosis Factor-alpha/pharmacology , bcl-2-Associated X Protein
8.
J Clin Endocrinol Metab ; 86(6): 2653-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11397868

ABSTRACT

Intrauterine growth restriction (IUGR) can be a consequence of decreased uterine blood flow (uteroplacental insufficiency) and maternal and fetal hypoxia. Insulin-like growth factors (IGFs) and their binding proteins (IGFBPs) are key elements in fetal growth. IGF-I is a major growth promoter in utero. IGFBP-1 is primarily made in the liver, and it mostly inhibits IGF actions at the cellular level. IGFBP-1 is elevated in the fetal circulation of human and animal pregnancies complicated by IUGR caused by placental insufficiency and in utero hypoxia and is believed to restrict fetal growth by sequestering IGFs. In this study, we developed a protocol to establish highly pure primary cultures of human fetal hepatocytes in vitro and investigated their expression of IGFBP-1 messenger RNA (mRNA) and protein and the effects of hypoxia on their expression of IGFBP-1 mRNA and protein. Hepatocytes were isolated from second-trimester human fetal livers (n = 7) and purified by Percoll gradient centrifugation. Hepatocyte cultures were characterized by immunocytochemistry and were compared with hepatocytes in situ in human fetal liver tissue, by immunohistochemistry, using specific antibodies and indirect immunofluorescence. Cultures consisted primarily (>90%) of cells positive for cytokeratin 18, fibrinogen, and IGFBP-1, with less than 2% vascular cells and less than 8% macrophages. Identification of isolated hepatocytes was further confirmed by morphology. Hepatocytes were cultured in defined medium, and Northern analysis revealed expression of a 1.5-kb IGFBP-1 mRNA transcript in hepatocytes cultured under normoxic conditions, for 24 h, that did not increase in steady-state levels after 48 h in culture. Under hypoxic conditions (2% O(2)), IGFBP-1 mRNA expression increased 3- to 4-fold, compared with normoxic controls. Cells cultured under 10% O(2) did not demonstrate an increase in IGFBP-1 mRNA levels. IGFBP-1 protein in conditioned medium (CM) was measured by immunoradiometric assay and increased 3- to 4-fold under hypoxic (2% O(2)), compared with normoxic, conditions. Western ligand blot analysis of CM revealed the presence of IGFBP-1, IGFBP-2, IGFBP-3, and IGFBP-4. IGFBP-1 was the most abundant IGFBP in CM, and densitometric analysis revealed a 2.5-fold increase in IGFBP-1 under hypoxic, compared with normoxic, conditions, supporting the immunoradiometric assay results. A 3-fold increase in IGFBP-3 mRNA, but not other IGFBPs, was noted under hypoxic, compared with normoxic, conditions. This study demonstrates that human fetal hepatocytes can be cultured in defined medium, as primary cultures with high purity, and that they express IGFBP-1 mRNA and secrete IGFBP-1 protein in vitro. In addition, the data demonstrate that hypoxia up-regulates fetal hepatocyte IGFBP-1 mRNA steady-state levels and protein, with this being the major IGFBP derived from the fetal hepatocyte. The data support a role for the fetal liver as a source of elevated circulating levels of IGFBP-1 in fetuses with in utero hypoxia and IUGR.


Subject(s)
Fetus/metabolism , Hepatocytes/metabolism , Hypoxia/metabolism , Insulin-Like Growth Factor Binding Protein 1/metabolism , Uterine Diseases/complications , Cells, Cultured , Female , Fetal Growth Retardation/etiology , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Liver/cytology , Liver/embryology , Placental Insufficiency/complications , Pregnancy , RNA, Messenger/metabolism , Reference Values
9.
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
11.
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
12.
J Clin Endocrinol Metab ; 85(10): 3821-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11061545

ABSTRACT

Insulin-like growth factor-binding protein 1 (IGFBP-1) is important in regulating minute-to-minute IGF bioavailability in the circulation and is primarily an inhibitor of IGF action systemically and in most cellular systems. Understanding regulation of IGFBP-1 is, thus, important in understanding regulation of IGF actions. The IGFBP-1 promoter contains a cAMP response element, and cAMP stimulates IGFBP-1 gene expression at the transcriptional level. Recently, we have found three consensus sequences for the hypoxia response element in intron 1 of the IGFBP-1 gene. Herein, we have investigated the effects of hypoxia and a cAMP analog, 8-bromoadenosine-3',5'-cyclic monophosphate (8-Br-cAMP), on IGFBP-1 expression in HepG2 cells, a model system for IGFBP-1 gene regulation. HepG2 cells were exposed to normoxia (20% pO2) or hypoxia (2% pO2) for 24 h in the absence or presence of 8-Br-cAMP (0.1, 0.5, and 1 mM). Western ligand blotting revealed IGFBP-1 as the predominant IGFBP in HepG2-conditioned media, which increased in a dose-dependent manner after incubation with 8-Br-cAMP in normoxia and hypoxia (3-fold and 7-fold at 1 mM, respectively). Under hypoxic, compared with normoxic, conditions, IGFBP-1 protein and messenger RNA (mRNA) levels increased approximately 10-fold and 20-fold, respectively. In normoxia, 8-Br-cAMP stimulated IGFBP-1 protein and mRNA levels in a dose-dependent manner (7-fold and 10-fold at 1 mM). Hypoxia and 8-Br-cAMP showed additive stimulatory effects on IGFBP-1 protein and mRNA levels (35-fold and 50-fold at 1 mM) that were time and dose dependent. Primary transcripts of IGFBP-1 mRNA were increased concordantly with IGFBP-1 mRNA. The half-life of the IGFBP-1 mRNA was markedly increased (approximately 6-fold) by hypoxia, and cAMP minimally enhanced this effect. These results demonstrate that hypoxia and compounds that increase intracellular cAMP additively regulate IGFBP-1 gene expression by transcriptional and posttranscriptional mechanisms. Regulation of IGFBP-1 mRNA and protein by cAMP and hypoxia may be important for understanding the physiologic and pathophysiologic roles of IGFBP-1.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation/physiology , Hypoxia/metabolism , Insulin-Like Growth Factor Binding Protein 1/biosynthesis , Liver Neoplasms, Experimental/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Blotting, Northern , Blotting, Western , Half-Life , Humans , Immunoradiometric Assay , Insulin-Like Growth Factor Binding Protein 1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Tumor Cells, Cultured
13.
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
14.
J Clin Endocrinol Metab ; 85(8): 2714-21, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10946870

ABSTRACT

Nitric oxide (NO) is believed to play an important, but as yet undefined, role in regulating hypoxia inducible gene expression. Recently, we have reported evidence suggesting that the human insulin-like growth factor-binding protein-1 (IGFBP-1) gene is directly regulated by hypoxia through the hypoxia-inducible factor-1 pathway. The goal of the current study was to investigate NO regulation of hypoxic induction of IGFBP-1 gene expression using HepG2 cells, a model system of hepatic gene expression. We report that a NO generator, sodium nitroprusside, significantly diminishes hypoxic activation of IGFBP-1 protein and messenger ribonucleic acid expression. Furthermore, these effects are independent of guanylate cyclase/ cGMP signaling, as two different inhibitors, LY 83583, a specific inhibitor of guanylate cyclase, and KT 5823, a protein kinase G inhibitor, had no effect on IGFBP-1 induction by hypoxia. Hypoxic induction of a reporter gene containing four tandemly ligated hypoxia response elements was completely blocked by sodium nitroprusside, but not by 8-bromo-cGMP, an analog ofcGMP. These results suggest that NO blocks hypoxic induction of IGFBP-1 by a guanylate cyclase/ cGMP-independent pathway, possibly at the level of oxygen sensing. The impaired hypoxia regulation of IGFBP-1 by nitric oxide may play a key role in the hyperinduction of IGFBP-1 observed in pathophysiological conditions such as fetal hypoxia and preeclampsia where dysregulation of NO has been observed.


Subject(s)
Carbazoles , Cell Hypoxia , Gene Expression Regulation , Indoles , Insulin-Like Growth Factor Binding Protein 1/genetics , Nitric Oxide/physiology , Nitroprusside/pharmacology , Alkaloids/pharmacology , Aminoquinolines/pharmacology , Base Sequence , Binding Sites , Carcinoma, Hepatocellular , Conserved Sequence , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP/physiology , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Guanylate Cyclase/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Kinetics , Liver/metabolism , Liver Neoplasms , Luciferases/genetics , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Biosynthesis , Recombinant Fusion Proteins/biosynthesis , Signal Transduction/drug effects , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured
15.
Clin Cancer Res ; 6(7): 2941-50, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914744

ABSTRACT

Tumor angiogenesis, the development of new blood vessels during malignant progression, is a regulated process that has both genetic and physiological controls. Physiologically, angiogenesis is stimulated by decreases in tissue oxygenation (i.e., hypoxia). We investigated the effect of hypoxia on the expression of two angiogenic factors reported to be genetically regulated by the p53 tumor suppressor gene: (a) the angiogenic inhibitor thrombospondin 1 (TSP-1); and (b) the angiogenic inducer vascular endothelial growth factor (VEGF). Analysis of rodent cells that differ in their p53 genotype (p53+/+ or p53-/-) indicated that in vitro exposure to hypoxia simultaneously suppressed TSP-1 and induced VEGF expression, regardless of the p53 genotype. On transformation of these cells with E1A and oncogenic H-ras, the basal level of TSP-1 expression was strongly diminished, whereas that of VEGF could still be induced by hypoxia. Consistent with these in vitro findings, sections of tumors derived from the transformed p53+/+ and p53-/- cells showed that VEGF protein overlapped with regions of hypoxia, whereas TSP-1 protein was below the limits of detection in tumor tissue. Using a panel of normal/immortalized and transformed human cells, it was found that the ability of hypoxia to inhibit TSP-1 expression depends on the cell type and/or the degree of transformation. In contrast, VEGF expression was induced by hypoxia in all of the human cell types examined. Together, these findings suggest that hypoxic and oncogenic signals could interact in the tumor microenvironment to inhibit TSP-1 and induce VEGF expression, promoting the switch to the angiogenic phenotype.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Hypoxia , Endothelial Growth Factors/genetics , Gene Expression Regulation , Genes, p53 , Lymphokines/genetics , Thrombospondin 1/genetics , Uterine Cervical Neoplasms/genetics , Animals , Carcinoma, Squamous Cell/pathology , Cell Division , Cells, Cultured , Female , Humans , Mice , Mice, SCID , Transfection , Tumor Suppressor Protein p53/genetics , Uterine Cervical Neoplasms/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
16.
Exp Cell Res ; 258(1): 82-91, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10912790

ABSTRACT

X-ray-induced damage leads to cell-cycle "checkpoint" arrest by p53-dependent induction of the cyclin-dependent kinase inhibitor p21 (Waf1/Cip1/Sdi1). Human tumor cells that lack this response fail to arrest after exposure to DNA-damaging agents, undergo multiple rounds of endoreduplicative DNA synthesis, and eventually commit to an apoptotic cell death. Since low oxygen tension can also induce p53 protein accumulation, and can lead to cell-cycle arrest or apoptosis, we examined the expression of p21 in tumor cells under normoxic and hypoxic conditions. In a survey of cells, mRNA for the p21 gene was induced two- to threefold in response to hypoxia in a seemingly p53-independent manner. We therefore examined genetically matched cells that differ in their p21 and p53 status for response to ionizing radiation and hypoxia. We found that both p21-deficient and p53-deficient cells exhibit an increase in chromosome instability, an increased level of apoptosis, and a failure to arrest after exposure to ionizing radiation. However, cells that lack either p21 or p53 exhibit no increase in chromosome instability or elevated apoptosis and still arrest in response to hypoxia. Thus, the mechanism responsible for the differential response to either hypoxia or X rays presumably lies in the control of cell-cycle progression in response to stress and its dependence on p21. Since the loss of a DNA-damage-dependent checkpoint does not sensitize cells to killing by stresses that elicit a DNA-damage-independent checkpoint, targeting the function of p21 pharmacologically will not kill tumor cells in situ in the absence of a DNA damage signal.


Subject(s)
Cell Cycle/physiology , Cell Hypoxia/physiology , Cyclins/physiology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA Damage , Enzyme Inhibitors/metabolism , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , Humans , Kinetics , Time Factors , Tumor Cells, Cultured , X-Rays
17.
Gene Ther ; 7(6): 493-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10757022

ABSTRACT

We are developing new gene therapy vectors whose expression is selectively activated by hypoxia, a unique feature of human solid tumors. As vascular endothelial growth factor (VEGF) is upregulated by hypoxia, such regulatory mechanisms would enable us to achieve hypoxia-inducible expression of therapeutic genes. Constructs with five copies of hypoxia-responsive elements (HREs) derived from the 5'-untranslated region (UTR) of the human VEGF showed excellent transcriptional activation at low oxygen tension relevant to tumor hypoxia. In an attempt to achieve higher responsiveness, various combinations of HREs and promoters were examined. In addition, we also investigated whether the 3' UTR of the VEGF gene would confer increased post-transcriptional mRNA stability under hypoxic conditions. However, despite increases in the hypoxic/aerobic ratio of luciferase activity, gene expression with 3' UTR was lower due to mRNA destabilization by AU-rich elements (AREs). Thus, we found no benefit from the inclusion of the 3' UTR in our vectors. Of all the vectors tested, the combination of 5HRE and a CMV minimal promoter exhibited hypoxia responsiveness (over 500-fold) to the similar level to the intact CMV promoter. We propose that this vector would be useful for tumor selective gene therapy.


Subject(s)
Endothelial Growth Factors/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Lymphokines/genetics , Neoplasms/therapy , Transcription Factors , 5' Untranslated Regions/genetics , Cell Hypoxia , Cytomegalovirus/genetics , DNA-Binding Proteins/genetics , Gene Expression , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Luciferases/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Cancer Res ; 60(4): 883-7, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706099

ABSTRACT

In this study, we have analyzed changes induced by hypoxia at the transcriptional level of genes that could be responsible for a more aggressive phenotype. Using a series of DNA array membranes, we identified a group of hypoxia-induced genes that included plasminogen activator inhibitor-1 (PAI-1), insulin-like growth factor-binding protein 3 (IGFBP-3), endothelin-2, low-density lipoprotein receptor-related protein (LRP), BCL2-interacting killer (BIK), migration-inhibitory factor (MIF), matrix metalloproteinase-13 (MMP-13), fibroblast growth factor-3 (FGF-3), GADD45, and vascular endothelial growth factor (VEGF). The induction of each gene was confirmed by Northern blot analysis in two different squamous cell carcinoma-derived cell lines. We also analyzed the kinetics of PAI-1 induction by hypoxia in more detail because it is a secreted protein that may serve as a useful molecular marker of hypoxia. On exposure to hypoxia, there was a gradual increase in PAI-1 mRNA between 2 and 24 h of hypoxia followed by a rapid decay after 2 h of reoxygenation. PAI-1 levels were also measured in the serum of a small group of head and neck cancer patients and were found to correlate with the degree of tumor hypoxia found in these patients.


Subject(s)
Cell Hypoxia , Membrane Proteins , Neoplasms/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins , Endothelial Growth Factors/genetics , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Lymphokines/genetics , Mice , Mitochondrial Proteins , Neoplasms/pathology , Phenotype , Plasminogen Activator Inhibitor 1/genetics , Proteins/genetics , RNA, Messenger/analysis , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Genes Dev ; 14(4): 391-6, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10691731

ABSTRACT

In glioblastoma-derived cell lines, PTEN does not significantly alter apoptotic sensitivity or cause complete inhibition of DNA synthesis. However, in these cell lines PTEN regulates hypoxia- and IGF-1-induced angiogenic gene expression by regulating Akt activation of HIF-1 activity. Restoration of wild-type PTEN to glioblastoma cell lines lacking functional PTEN ablates hypoxia and IGF-1 induction of HIF-1-regulated genes. In addition, Akt activation leads to HIF-1alpha stabilization, whereas PTEN attenuates hypoxia-mediated HIF-1alpha stabilization. We propose that loss of PTEN during malignant progression contributes to tumor expansion through the deregulation of Akt activity and HIF-1-regulated gene expression.


Subject(s)
Brain Neoplasms/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/genetics , Insulin-Like Growth Factor I/pharmacology , Neoplasm Proteins/physiology , Nuclear Proteins/physiology , Phosphoric Monoester Hydrolases/deficiency , Protein Serine-Threonine Kinases , Transcription Factors , Tumor Suppressor Proteins , Apoptosis , Brain Neoplasms/pathology , Cell Hypoxia/genetics , Culture Media, Serum-Free/pharmacology , Cyclooxygenase 1 , Disease Progression , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Gene Deletion , Genetic Complementation Test , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Isoenzymes/biosynthesis , Isoenzymes/genetics , Lymphokines/biosynthesis , Lymphokines/genetics , Membrane Proteins , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , PTEN Phosphohydrolase , Phosphofructokinase-1/biosynthesis , Phosphofructokinase-1/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Methods Mol Med ; 35: 413-8, 2000.
Article in English | MEDLINE | ID: mdl-21390820

ABSTRACT

Many murine and human tumors contain hypoxic or necrotic regions in which the oxygen tension is abnormally low. For example,>50% of primary tumors of the breast, cervix, and head and neck contain areas that are hypoxic. Because hypoxic regions are not present in normal tissue, this provides the potential for selectively targeting gene therapy to tumor cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...