Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 333
Filter
1.
Cell Death Differ ; 14(11): 1948-57, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17690712

ABSTRACT

Avicins, a family of plant triterpene electrophiles, can trigger apoptosis-associated tumor cell death, and suppress chemical-induced carcinogenesis by its anti-inflammatory, anti-mutagenic, and antioxidant properties. Here, we show that tumor cells treated with benzyloxycarbonylvalyl-alanyl-aspartic acid (O-methyl)-fluoro-methylketone, an apoptosis inhibitor, and Bax(-/-)Bak(-/-) apoptosis-resistant cells can still undergo cell death in response to avicin D treatment. We demonstrate that this non-apoptotic cell death is mediated by autophagy, which can be suppressed by chloroquine, an autophagy inhibitor, and by specific knockdown of autophagy-related gene-5 (Atg5) and Atg7. Avicin D decreases cellular ATP levels, stimulates the activation of AMP-activated protein kinase (AMPK), and inhibits mammalian target of rapamycin (mTOR) and S6 kinase activity. Suppression of AMPK by compound C and dominant-negative AMPK decreases avicin D-induced autophagic cell death. Furthermore, avicin D-induced autophagic cell death can be abrogated by knockdown of tuberous sclerosis complex 2 (TSC2), a key mediator linking AMPK to mTOR inhibition, suggesting that AMPK activation is a crucial event targeted by avicin D. These findings indicate the therapeutic potential of avicins by triggering autophagic cell death.


Subject(s)
Autophagy , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Saponins/pharmacology , Ubiquitin-Activating Enzymes/metabolism , AMP-Activated Protein Kinases , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Cell Line, Tumor , Chloroquine/pharmacology , Enzyme Activation , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Sirolimus/pharmacology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Activating Enzymes/genetics
2.
Proc Natl Acad Sci U S A ; 98(20): 11551-6, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11572997

ABSTRACT

We tested the ability of avicins, a family of triterpenoid saponins obtained from Acacia victoriae (Bentham) (Leguminosae: Mimosoideae), to inhibit chemically induced mouse skin carcinogenesis. Varying doses of avicins were applied to shaved dorsal skin of SENCAR mice 15 min before application of 100 nmol of 7,12-dimethylbenz[a]anthracene (DMBA) twice a week for 4 weeks (complete carcinogenesis model). The dorsal skin of a second group of mice was treated with one dose of 10 nmol of DMBA. Avicins were then applied 15 min before repetitive doses of 2 microg of phorbol 12-tetradecanoate 13-acetate (TPA) twice a week for 8 weeks (initiation/promotion model). At 12 weeks, avicins produced a 70% decrease in the number of mice with papillomas and a greater than 90% reduction in the number of papillomas per mouse in both protocols. We also observed a 62% and 74% reduction by avicins in H-ras mutations at codon 61 in the DMBA and DMBA/TPA models, respectively, as well as a significant inhibition of the modified DNA base formation (8-OH-dG) in both protocols. Marked suppression of aneuploidy occurred with treatment at 16 weeks in the initiation/promotion experiment. These findings, when combined with the proapoptotic property of these compounds and their ability to inhibit hydrogen peroxide (H(2)O(2)) generation, nuclear factor-kappaB (NF-kappaB) activation, and inducible nitric oxide synthase (iNOS) induction reported elsewhere, suggest that avicins could prove exciting in reducing oxidative and nitrosative stress and thereby suppressing the development of human skin cancer and other epithelial malignancies.


Subject(s)
Acacia/therapeutic use , Dioxoles/therapeutic use , Genes, ras , Phenanthridines/therapeutic use , Phytotherapy , Saponins/therapeutic use , Skin Neoplasms/prevention & control , Triterpenes/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Aneuploidy , Animals , Disease Models, Animal , Female , Humans , Jurkat Cells , Mice , Mice, Inbred SENCAR , Papilloma/chemically induced , Papilloma/prevention & control , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate/therapeutic use
3.
Proc Natl Acad Sci U S A ; 98(20): 11557-62, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11572998

ABSTRACT

Triterpenoid saponins, which are present in leguminous plants and some marine animals, possess a broad range of biological actions. We have earlier reported the extraction of avicins, a family of triterpenoid saponins obtained from the Australian desert tree Acacia victoriae (Leguminosae: Mimosoideae) that inhibit tumor cell growth and induce apoptosis, in part, by perturbing mitochondrial function. These saponins have also been found to prevent chemical-induced carcinogenesis in mice. This study examines the effect of a triterpene mixture (F094) and a single molecular species (avicin G) isolated from the mixture on tumor necrosis factor (TNF)-induced activation of nuclear transcription factor-kappaB (NF-kappaB) in Jurkat cells (human T cell leukemia). Both F094 and avicin G were found to be potent inhibitors of TNF-induced NF-kappaB. Treatment of Jurkat cells with avicin G resulted in a much slower accumulation of the p65 subunit of NF-kappaB into the nucleus whereas the degradation of IkappaBalpha was unaffected. Avicin G also impaired the binding of NF-kappaB to DNA in in vitro binding assays. Treatment of cells with DTT totally reversed the avicin G-induced inhibition of NF-kappaB activity, suggesting that sulfhydryl groups critical for NF-kappaB activation were being affected. Avicin G treatment resulted in decreased expression of NF-kappaB-regulated proteins such as inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX-2). Thus, the avicins may prove important for reducing both oxidative and nitrosative cellular stress and thereby suppressing the development of malignancies and related diseases.


Subject(s)
Acacia/therapeutic use , NF-kappa B/antagonists & inhibitors , Phytotherapy , Saponins/therapeutic use , Animals , Cell Line , Cell Nucleus/metabolism , Cyclooxygenase 2 , DNA, Neoplasm/metabolism , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/genetics , Jurkat Cells , Luciferases/genetics , Membrane Proteins , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Prostaglandin-Endoperoxide Synthases/genetics , Tumor Necrosis Factor-alpha/pharmacology
4.
Cancer Res ; 61(14): 5486-90, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454696

ABSTRACT

This report describes the isolation and partial purification of novel triterpenoid saponins [Fraction 35 (F035)] and two pure biologically active derivatives (termed avicins D and G) from Acacia victoriae, an Australian desert tree of the Leguminosae family. F035 and the avicins markedly inhibited the growth of several tumor cell lines with minimum growth inhibition in human foreskin fibroblasts, mouse fibroblasts, and immortalized breast epithelial cells at similar concentrations. F035 and the avicins induced cell cycle (G1) arrest of the human MDA-MB-453 breast cancer cell line and apoptosis of the Jurkat (T-cell leukemia) and the MDA-MB-435 breast cancer cell line. The triterpenoid saponins also partially inhibited phosphatidylinositol 3-kinase activity in Jurkat T cells in a time-dependent manner and phosphorylation in the downstream protein Akt, whereas no affect was seen on the Ras/mitogen-activated protein kinase cascade. These observations as well as other work from our laboratory demonstrating mitochondrial perturbation, chemoprevention, and inhibition of nuclear factor kappaB suggest that triterpenoid saponins from A. victoriae have potential as novel anticancer agents. Recent work linking Akt signaling with glucose metabolism, stress resistance, and longevity suggests other potential applications of these compounds.


Subject(s)
Acacia/chemistry , Apoptosis/drug effects , Cell Division/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Cell Cycle/drug effects , Humans , Jurkat Cells , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Plant Extracts/chemistry , Plant Extracts/pharmacology , Signal Transduction/drug effects , Subcellular Fractions/chemistry , Tumor Cells, Cultured , U937 Cells
5.
Proc Natl Acad Sci U S A ; 98(10): 5821-6, 2001 May 08.
Article in English | MEDLINE | ID: mdl-11344312

ABSTRACT

Anticancer agents target various subcellular components and trigger apoptosis in chemosensitive cells. We have recently reported the tumor cell growth inhibitory properties of a mixture of triterpenoid saponins obtained from an Australian desert tree (Leguminosae) Acacia victoriae (Bentham). Here we report the purification of this mixture into two biologically pure components called avicins that contain an acacic acid core with two acyclic monoterpene units connected by a quinovose sugar. We demonstrate that the mixture of triterpenoid saponins and avicins induce apoptosis in the Jurkat human T cell line by affecting the mitochondrial function. Avicin G induced cytochrome c release within 30-120 min in whole cells and within a minute in the cell-free system. Caspase inhibitors DEVD or zVAD-fmk had no effect on cytochrome c release, suggesting the direct action of avicin G on the mitochondria. Activation of caspase-3 and total cleavage of poly(ADP-ribose) polymerase (PARP) occurred between 2 and 6 h posttreatment with avicins by zVAD-fmk. Interestingly, in the treated cells no significant changes in the membrane potential preceded or accompanied cytochrome c release. A small decrease in the generation of reactive oxygen species (ROS) was measured. The study of these evolutionarily ancient compounds may represent an interesting paradigm for the application of chemical ecology and chemical biology to human health.


Subject(s)
Apoptosis/drug effects , Mitochondria/drug effects , Rosales/chemistry , Saponins/pharmacology , Triterpenes/chemistry , Carbohydrate Sequence , Caspases/metabolism , Cytochrome c Group/metabolism , Enzyme Activation , Humans , Jurkat Cells , Membrane Potentials/drug effects , Mitochondria/enzymology , Molecular Sequence Data , Reactive Oxygen Species , Saponins/chemistry , Saponins/isolation & purification
6.
Clin Cancer Res ; 7(5): 1198-203, 2001 May.
Article in English | MEDLINE | ID: mdl-11350884

ABSTRACT

Clinical and laboratory observations support the view that angiogenesis is necessary for prostate cancer progression. The angiogenesis inhibitor TNP-470 has demonstrated in vivo antitumor activity in a series of clinical models. To evaluate a possible therapeutic clinical value, we conducted a Phase I dose escalation trial of alternate-day i.v. TNP-470 in 33 patients with metastatic and androgen-independent prostate cancer. The patients were evaluated during therapy for evidence of neurological toxic effects. An assay of endothelial and vascular proliferation "markers" and a sequential assay of serum prostate-specific antigen concentration were performed. The effects of TNP-470 could be evaluated in 32 of the 33 patients. The maximum tolerated dose was 70.88 mg/m(2) of body surface area. The dose-limiting toxic effect was a characteristic neuropsychiatric symptom complex (anesthesia, gait disturbance, and agitation) that resolved upon cessation of therapy. The times to clinical recovery of neurological side effects were 6, 8, and 14 weeks. No definite antitumor activity of TNP-470 was observed; however, transient stimulation of the serum prostate-specific antigen concentration occurred in some of the patients treated. Additional studies of TNP-470 should be conducted using an alternate-day i.v. injection of 47.25 mg/m(2) body surface area and should focus on understanding and overcoming the neurological toxic effects. In addition, valid intermediate end points that reflect the status of tumor-associated neovascularity are needed to facilitate effective development of treatment strategies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Sesquiterpenes/therapeutic use , Aged , Androgens/metabolism , Angiogenesis Inhibitors/adverse effects , Blood Glucose/drug effects , Bone and Bones/drug effects , Communicable Diseases/etiology , Cyclohexanes , Digestive System/drug effects , Fibroblast Growth Factors/urine , Humans , Kinetics , Liver/drug effects , Male , Middle Aged , O-(Chloroacetylcarbamoyl)fumagillol , Pain/etiology , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/complications , Prostatic Neoplasms/metabolism , Sesquiterpenes/adverse effects , Thrombomodulin/blood
7.
Cell Growth Differ ; 10(2): 93-100, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10074903

ABSTRACT

p202 is an IFN-inducible, primarily nuclear, phosphoprotein (52-kDa) whose constitutive overexpression in transfected cells inhibits colony formation. To investigate the molecular mechanism(s) by which expression of p202 protein impairs colony formation, we established stable cell lines that inducibly express p202. Using this cell model, we demonstrate that the induced expression of p202 in asynchronous cultures of these cells was accompanied by: (a) an increase in steady-state levels of p21(WAF1/CIP1/SDI1) (p21) mRNA and protein; (b) a decrease in Cdk2 protein kinase activity; and (c) an increase in the functional form of retinoblastoma protein (pRb). Transient transfection of a p202-encoding plasmid in Saos-2 cells, which do not harbor a wild-type p53 protein, resulted in an increase in p21 protein, which indicated that p202 could regulate expression of p21 protein independent of p53 protein. Moreover, we demonstrate that expression of p202 in these cells increased cell doubling time without accumulation of cells in a particular phase of the cell cycle. Taken together, these results are consistent with the possibility that p202 protein contributes to the cell growth retardation activity of the IFNs, at least in part, by modulating p21 protein levels.


Subject(s)
Cell Division , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Intracellular Signaling Peptides and Proteins , Phosphoproteins/metabolism , Animals , Blotting, Northern , Carrier Proteins , Cell Cycle/genetics , Cell Line , Chromosomal Proteins, Non-Histone , Cyclin A/immunology , Cyclin D1/immunology , Cyclin E/immunology , Cyclin-Dependent Kinase Inhibitor p21 , DNA-Binding Proteins , Fibroblasts/metabolism , Flow Cytometry , Histones/metabolism , Humans , Interferon-alpha/metabolism , Mice , Microscopy, Fluorescence , Retinoblastoma Protein/immunology , Retinoblastoma Protein/metabolism , Tetracycline/pharmacology , Time Factors , Transfection , Tumor Suppressor p53-Binding Protein 1
8.
Oncogene ; 18(3): 807-11, 1999 Jan 21.
Article in English | MEDLINE | ID: mdl-9989832

ABSTRACT

Interferons (IFNs) can exert cytostatic and immunomodulatory effects on carcinoma cells. In particular, growth inhibition of human prostate carcinoma by IFNs has been demonstrated both in vitro and in vivo. p202 is a 52 kd nuclear phosphoprotein known to be induced by IFNs. In this report, we showed that the expression of p202 was associated with an anti-proliferative effect on human prostate cancer cells. More importantly, cells that expressed p202 showed reduced ability to grow in soft-agar, indicating a loss of transformation phenotype. Our data suggest that p202 is a growth inhibitor gene in prostate cancer cells and its expression may also suppress transformation phenotype of prostate cancer cells.


Subject(s)
Carrier Proteins/biosynthesis , Cell Transformation, Neoplastic , Growth Inhibitors/biosynthesis , Interferon-alpha/pharmacology , Intracellular Signaling Peptides and Proteins , Phosphoproteins/biosynthesis , Prostatic Neoplasms , Cell Division , Gene Expression , Humans , Male , Phenotype , Tumor Cells, Cultured , Tumor Suppressor p53-Binding Protein 1
9.
FEBS Lett ; 438(1-2): 21-4, 1998 Oct 30.
Article in English | MEDLINE | ID: mdl-9821952

ABSTRACT

Murine p202 is an interferon-inducible primarily nuclear phosphoprotein (52 kDa) whose expression in transfected cells inhibits colony formation. p202-binding proteins include the pocket proteins (pRb, p107 and p130), a p53-binding protein (sm53BP1), and transcription factors (e.g. NF-kappaB (p50 and p65), AP-1 (c-Fos and c-Jun), E2F-1, E2F-4, MyoD, and myogenin). p202 modulates the transcriptional activity of these factors in transfected cells. Here we demonstrate that p202 self-associates directly and a sequence in p202, which is conserved among the members of the 200-family proteins, was sufficient for self-association in vitro. Our observations reported herein raise the possibility that self-association of p202 may provide a mechanism for the regulation of its activity.


Subject(s)
Carrier Proteins/metabolism , Conserved Sequence , Intracellular Signaling Peptides and Proteins , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Carrier Proteins/chemistry , Chromatography, Affinity , Chromosomal Proteins, Non-Histone , Cross-Linking Reagents , DNA-Binding Proteins , Dimerization , Dimethyl Suberimidate , Glutathione Transferase/metabolism , Mice , Multigene Family , Nuclear Proteins/metabolism , Peptides/metabolism , Phosphoproteins/chemistry , Protein Binding , Protein Biosynthesis , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Tumor Suppressor p53-Binding Protein 1
10.
Biochem Biophys Res Commun ; 247(2): 379-82, 1998 Jun 18.
Article in English | MEDLINE | ID: mdl-9642135

ABSTRACT

p202 is an interferon (IFN)-inducible, primarily nuclear, phosphoprotein (52-kDa) whose overexpression in transfected cells inhibits colony formation. p202 binds to the retinoblastoma tumor suppressor protein and two other members of the pocket family proteins (p107 and p130). Moreover, overexpression of p202 in transfected cells inhibits the transcriptional activity of E2Fs (E2F-1/DP-1 and E2F-4/DP-1), p53, AP-1 c-Fos and c-Jun, NF-kappaB p50 and p65. Here we demonstrate that inhibition of endogenous p202 production in murine AKR-2B fibroblasts did not result in an increase in cell proliferation. Instead, these cells exhibited increased susceptibility to apoptosis in response to decrease in serum concentrations in the growth medium. These observations are consistent with the notion that normal levels of p202 may be needed for the regulation of cell proliferation.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/physiology , Phosphoproteins/physiology , Animals , Carrier Proteins/genetics , Cell Division/physiology , Cell Line , Chromosomal Proteins, Non-Histone , Culture Media , DNA-Binding Proteins , Fibroblasts , Gene Expression , Mice , Nuclear Proteins/genetics , Phosphoproteins/genetics , RNA, Antisense/genetics , Transfection , Tumor Suppressor p53-Binding Protein 1
11.
Oncogene ; 15(3): 291-301, 1997 Jul 17.
Article in English | MEDLINE | ID: mdl-9233764

ABSTRACT

The interferon (IFN)-inducible proteins mediate activities of the interferons including the cell growth-regulatory activity. We have shown that p202, an IFN-inducible 52kDa primarily nuclear phosphoprotein whose expression in transfected cells inhibits cell proliferation, interacts with the retinoblastoma tumor suppressor protein (pRb) and the transcription factor E2F (E2F-1/ DP-1) in vitro and in vivo. p202 was shown to inhibit E2F-1/DP-1-stimulated transcription of a reporter gene and of endogenous genes. Here we report that expression of p202 inhibited E2F-4/DP-1-stimulated transcription of a reporter gene in transfected cells. Furthermore, this inhibition was associated with the inhibition of the sequence-specific DNA-binding of E2F-4 both in complex with the pocket proteins p107 or p130 and in its 'free' form in vitro. p202 bound to p107 and p130 in vitro and in vivo and also associated with E2F-4, supporting the notion that complexes containing p107/E2F-4 or p130/ E2F-4 and p202 exist in vivo. Moreover, cotransfection of E2F-4-encoding plasmid in AKR-2B cells overcame p202-mediated inhibition of cell growth, raising the possibility that p202 contributes to cell growth inhibition by the interferons, at least in part, by modulating E2F-4-mediated transcription.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Cycle , Cell Division , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , Female , Genes, Reporter , Humans , Mice , Protein Binding , Recombinant Fusion Proteins/biosynthesis , Reticulocytes/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/antagonists & inhibitors , Transfection , Tumor Cells, Cultured , Tumor Suppressor p53-Binding Protein 1 , Uterine Cervical Neoplasms
14.
Clin Cancer Res ; 3(9): 1501-5, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9815836

ABSTRACT

A Phase I study of the novel angiogenesis inhibitor TNP-470 was performed. Patients with inoperable recurring or metastatic squamous cell cancer of the cervix with evaluable disease, no coagulopathy, and adequate renal, hepatic, and hematological function were eligible. One course of treatment consisted of an i.v. infusion of TNP-470 over 60 min every other day for 28 days, followed by a 14-day rest period. The starting dose was 9.3 mg/m2. Eighteen evaluable patients were treated, with a median age of 48 years (range 27-55) and performance status Zubrod 1 (range 0-2). Grade 3 neurotoxicities consisting of weakness, nystagmus, diplopia, and ataxia were encountered in two patients receiving the 71.2 mg/m2 dose. An intermediate dose level of 60 mg/m2 was evaluated and found to be well tolerated by three patients. Only one patient experienced grade 3 nausea on the 60 mg/m2 dose level. No myelosuppression, retinal hemorrhage, weight loss, or significant alopecia were observed. One patient had a complete response, which continues for 26 months, and three patients with initially progressive disease stage had stable disease for 5, 7.7, and 19+ months. Other Phase I studies, including over 200 patients, were performed concurrently with this study. Based on this experience, the dose of TNP-470 recommended for further studies is 60 mg/m2 as a 60-min i.v. infusion every Monday, Wednesday, and Friday. Neurotoxicity was dose limiting, but appears to be reversible. Otherwise, the treatment was well tolerated. The drug may be active in squamous cell cancer of the cervix. Further studies of TNP-470 in squamous cell cancer of the cervix are warranted.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Neovascularization, Pathologic/drug therapy , Sesquiterpenes/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Adult , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Combined Modality Therapy , Cyclohexanes , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Middle Aged , Nausea/chemically induced , Nervous System Diseases/chemically induced , O-(Chloroacetylcarbamoyl)fumagillol , Salvage Therapy , Sesquiterpenes/administration & dosage , Sesquiterpenes/adverse effects , Treatment Outcome , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/therapy
15.
EMBO J ; 15(20): 5668-78, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8896460

ABSTRACT

Many of the antimicrobial, immunomodulatory and cell growth inhibitory activities of the interferons are mediated by interferon-inducible proteins. Earlier we characterized an interferon-inducible murine protein, p202, whose expression in transfected cells inhibits cell proliferation and which can form a complex with retinoblastoma protein (pRb). Here we report that in transfected cells expression of p202 inhibits E2F-stimulated transcription of a reporter gene and of endogenous genes. Inhibition of the transcriptional activity of E2F by p202 does not depend on fully functional pRb and is correlated with inhibition of the sequence-specific DNA binding of E2F. p202 interacts with the transcription factor E2F (E2F-1/DP-1) in vitro and in vivo. Inhibition of E2F activity by p202 may contribute to growth inhibition by the interferons.


Subject(s)
Carrier Proteins/pharmacology , Cell Cycle Proteins , DNA-Binding Proteins , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/pharmacology , Phosphoproteins/pharmacology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Animals , Cell Division/drug effects , DNA/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Electrophoresis, Polyacrylamide Gel , Female , Genes, Reporter , HeLa Cells , Humans , Interferons/physiology , Mice , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Tumor Cells, Cultured , Tumor Suppressor p53-Binding Protein 1
16.
J Interferon Cytokine Res ; 16(9): 709-15, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8887055

ABSTRACT

Previous work showed that IFN-alpha induced the autoimmune-associated lupus inclusions (LI) in all 16 umbilical cord mononuclear cell samples from healthy mothers. In contrast, IFN-alpha induced LI and the LI-associated protein, p36, in only 2 of 16 human B lymphoblastoid cell lines. Resistance of these 14 cell lines to form LI and p36 may be due to their stage of development or differentiation or their transformed state. We sought to determine whether aging, neoplastic transformation, and HIV infection affected the observed IFN-alpha induction of LI in cord blood mononuclear cells. Expression of LI and p36 was investigated in PBMC on IFN-alpha chemotherapy and on culturing IFN-alpha with PBMC samples prepared from healthy adults and AIDS patients. The IFN-alpha induction of LI (detected by electron microscopy) or p36 (detected by two-dimensional gels) in all of the PBMC samples from these individuals was indistinguishable from the cord blood mononuclear cell response. Furthermore, induction of p36 and LI was not a good indicator of effective IFN-alpha chemotherapy. It may be consequential for autoimmunity induced by IFN-alpha in cancer, AIDS, and systemic lupus erythematosus (SLE). An essential biologic role for p36 and LI is suggested by a highly homologous p36 gene in the invertebrate Caenorhabditis elegans.


Subject(s)
Antineoplastic Agents/adverse effects , Antiviral Agents/adverse effects , Inclusion Bodies/drug effects , Interferon Type I/adverse effects , Interferon-alpha/biosynthesis , Lupus Vulgaris/pathology , Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/metabolism , Acquired Immunodeficiency Syndrome/pathology , Adult , Annexin A2/biosynthesis , Case-Control Studies , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Neoplasm Proteins/biosynthesis , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Proteins , Reference Values
17.
Biochem Biophys Res Commun ; 221(2): 396-401, 1996 Apr 16.
Article in English | MEDLINE | ID: mdl-8619867

ABSTRACT

p202 is an interferon-inducible protein whose expression in transfected cells inhibits proliferation. p202 binds to the retinoblastoma tumor suppressor protein in vitro and in vivo and the transcription factors AP-1 c-Fos and c-Jun, NF-kappaB p50 and p65, and inhibits the transcriptional activity of these factors in vivo. Here we report that p202 nonspecifically binds to double-stranded DNA and to single-stranded DNA in vitro. Analysis with recombinant p202 revealed that DNA binding activity is intrinsic to p202. A C-terminal deletion mutant of p202 exhibited DNA-binding properties, indicating that the C-terminus is dispensable for DNA binding. We also found that underphosphorylated p202 efficiently binds to DNA. Our data suggest that DNA binding activity of p202 may contribute to its functions.


Subject(s)
Carrier Proteins/biosynthesis , DNA-Binding Proteins/biosynthesis , Interferons/physiology , Intracellular Signaling Peptides and Proteins , Phosphoproteins/biosynthesis , Animals , Carrier Proteins/metabolism , Cell Line , DNA-Binding Proteins/metabolism , Mice , Phosphoproteins/metabolism , Phosphorylation
18.
Oncogene ; 12(8): 1617-23, 1996 Apr 18.
Article in English | MEDLINE | ID: mdl-8622881

ABSTRACT

Mutations of p53 gene are reported in 50-60% of human cancers and reintroduction of wild-type p53 can suppress cell proliferation. In this study, replication deficient recombinant adenovirus encoding wild-type p53 (ACN53) under the control of the human cytomegalovirus (CMV) promoter was constructed. A specific incorporation of the p53 gene with ACN53 reduced 3 (deleted p53 gene) cells was observed. ACN53 reduced the colony-forming ability of SK-OV-3 cells 72-216 h after single infection. A highly aggressive ovarian xenograft model was established in which animals die between 25-45 days. A localization study with the adenovirus-containing beta galactosidase reporter gene showed effective gene transfer in the tumor tissues. Ex vivo treatment of SK-OV-3 cells with ACN53 followed by injection into nude mice, increased the survival of the p53 treated mice by more than 50% compared with control animals. Gene therapy with ACN53 in intraperitoneal model of SK-OV-3 cells in two independent experiments revealed that there were some long-term survivors in the group of mice [2/5 (66 and 120 days) and [2/8 (166 and 423 days)] treated with ACN53. These findings demonstrate the potential of the p53 tumor suppressor gene therapy in human ovarian carcinoma.


Subject(s)
Adenocarcinoma/therapy , Adenoviridae/genetics , Genes, p53 , Genetic Therapy , Ovarian Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/virology , Animals , Carcinogenicity Tests , Female , Genetic Vectors , Humans , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/virology , Transduction, Genetic , Tumor Cells, Cultured
19.
JAMA ; 274(13): 1061, 1995 Oct 04.
Article in English | MEDLINE | ID: mdl-7563459
20.
Cell Growth Differ ; 6(6): 673-80, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7669721

ABSTRACT

Aberrant expression of the tumor suppressor gene RB1 is associated with a variety of solid tumors and hematopoietic neoplasms. Certain cancer cell lines in which the protein encoded by RB1 (p110RB) is absent have been reported to show decreased growth rate, clonogenicity, or tumorigenicity following insertion of a transcriptionally active RB1 gene. We asked whether these RB-deficient cells could be growth inhibited by direct exposure to purified p110RB. We report a decrease in uptake of tritiated thymidine by 5637 bladder carcinoma cells (RB-negative) when purified recombinant p110RB is added to culture media. Internalization of the protein by cells and translocation to the nucleus are demonstrated by immunohistochemistry, FACS, and detection of radiolabeled protein in subcellular fractions. Next, we chose a well-described leukemia cell culture model to investigate the potential effect of recombinant p110RB in clinical disease. We observed dose-related decreases in cell number of colony formation in vitro in 8 of 20 acute myelogenous leukemia samples, 7 of which did show endogenous p110RB detectable by immunohistochemistry. Histological appearance following exposure to p110RB shows cytoplasmic vacuolization and nuclear lobulation of degenerating cells. We conclude that purified p110RB added to culture media is internalized by cells, translocated to the nucleus, and exerts a growth-inhibitory effect on certain cancer cell types.


Subject(s)
Carcinoma, Squamous Cell/pathology , Leukemia, Myeloid/pathology , Recombinant Fusion Proteins/pharmacology , Retinoblastoma Protein/pharmacology , Urinary Bladder Neoplasms/pathology , Acute Disease , Adult , Aged , Amino Acid Sequence , Cell Differentiation/drug effects , Cell Division/drug effects , DNA Replication/drug effects , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Neoplastic Stem Cells/drug effects , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...