Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Ann Oncol ; 25(8): 1603-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24827131

ABSTRACT

BACKGROUND: Cabozantinib targets tyrosine kinases including the hepatocyte growth factor receptor (MET) and vascular endothelial growth factor (VEGF) receptor 2, which are important drug targets in renal cell carcinoma (RCC). PATIENTS AND METHODS: This single-arm open-label phase I trial evaluated the safety and tolerability of cabozantinib in heavily pretreated patients with metastatic clear cell RCC. RESULTS: The study enrolled 25 RCC patients for whom standard therapy had failed. Patients received a median of two prior systemic agents, and most patients had previously received at least one VEGF pathway inhibiting therapy (22 patients [88%]). Common adverse events included fatigue, diarrhea, nausea, proteinuria, appetite decreased, palmar-plantar erythrodysesthesia, and vomiting. Partial response was reported in seven patients (28%). Median progression-free survival was 12.9 months, and median overall survival was 15.0 months. CONCLUSION: Cabozantinib demonstrates preliminary anti-tumor activity and a safety profile similar to that seen with other multitargeted VEGFR tyrosine kinase inhibitors in advanced RCC patients. Further evaluation of cabozantinib in RCC is warranted. ClinicalTrials.gov identifier: NCT01100619.


Subject(s)
Anilides/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Pyridines/therapeutic use , Adult , Aged , Carcinoma, Renal Cell/pathology , Female , Follow-Up Studies , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Treatment Outcome , Tumor Burden/drug effects
2.
Article in English | MEDLINE | ID: mdl-22089927

ABSTRACT

Hypoxia-inducible factor (HIF) deregulation contributes to the Warburg effect. HIF consists of an unstable α subunit and a stable ß subunit. In the presence of oxygen, HIFα becomes prolyl hydroxylated by members of the EglN (also called PHD) family, leading to its proteasomal degradation. Under hypoxic conditions, EglN activity is diminished and HIF levels rise. EglN1 is the primary HIF prolyl hydroxylase with EglN2 and EglN3 playing compensatory roles under certain conditions. EglN2 and EglN3 also appear to play HIF-independent roles in regulating cell proliferation and apoptosis, respectively. The EglNs belong to a large family of 2-oxoglutarate-dependent dioxygenases that includes the TET DNA hydroxymethylases and JmjC-containing histone demethylases. Members of this superfamily can be inhibited by endogenous metabolites, including fumarate and succinate, which accumulate in tumors that have fumarate hydratase (FH) or succinate dehydrogenase (SDH) mutations, respectively, as well as by the 2-hydroxyglutarate detected in isocitrate dehydrogenase (IDH) mutant tumors. 2-Oxoglutarate-dependent dioxygenases therefore provide a link between altered metabolism and cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Dioxygenases/metabolism , Ketoglutaric Acids/metabolism , Neoplasms/metabolism , Animals , Dioxygenases/antagonists & inhibitors , Glycolysis , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/therapy
3.
Neurobiol Aging ; 32(11): 2030-44, 2011 Nov.
Article in English | MEDLINE | ID: mdl-20018408

ABSTRACT

Microglia have neuroprotective capacities, yet chronic activation can promote neurotoxic inflammation. Neuronal fractalkine (FKN), acting on CX(3)CR1, has been shown to suppress excessive microglia activation. We found that disruption in FKN/CX(3)CR1 signaling in young adult rodents decreased survival and proliferation of neural progenitor cells through IL-1ß. Aged rats were found to have decreased levels of hippocampal FKN protein; moreover, interruption of CX(3)CR1 function in these animals did not affect neurogenesis. The age-related loss of FKN could be restored by exogenous FKN reversing the age-related decrease in hippocampal neurogenesis. There were no measureable changes in young animals by the addition of exogenous FKN. The results suggest that FKN/CX(3)CR1 signaling has a regulatory role in modulating hippocampal neurogenesis via mechanisms that involve indirect modification of the niche environment. As elevated neuroinflammation is associated with many age-related neurodegenerative diseases, enhancing FKN/CX(3)CR1 interactions could provide an alternative therapeutic approach to slow age-related neurodegeneration.


Subject(s)
Aging/physiology , Chemokine CX3CL1/metabolism , Hippocampus/metabolism , Neurogenesis/physiology , Receptors, Chemokine/metabolism , Age Factors , Animals , CX3C Chemokine Receptor 1 , Interleukin-1beta/metabolism , Microglia/metabolism , Neurons/metabolism , Rats , Signal Transduction/physiology
4.
Clin Genet ; 79(6): 539-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20560986

ABSTRACT

This report describes clinical characteristics in families with a Type 2A phenotype and functional properties of a novel von Hippel Lindau variant (X214L). Pedigrees were analyzed. Analysis of von Hippel Lindau (VHL) coding exons and flanking intronic sequences in DNA from a proband with pheochromocytoma and islet cell tumor was performed. Western blot assays for VHL protein (pVHL), HIFα, and Jun B were conducted using VHL null renal clear carcinoma cell lines that were engineered to produce wild-type or X214L mutant pVHL. Pedigree analysis indicated that the variant tracked with disease and the same or similar VHL point mutations were identified in several Type 2A families. The predicted 14 amino acid extended pVHL variant, when reintroduced into VHL null cells, was stable and retained the ability to downregulate HIFα in a hydroxylationdependent manner. In contrast, the variant was defective with respect to downregulation of JunB. pVHL X214L, like other pVHL variants associated with a low risk of clear cell renal carcinoma, largely preserves the ability to downregulate HIF. In contrast, this variant, like other pVHL variants linked to Type 2A disease, fails to suppress JunB. This underscores that JunB may play a role in the pathogenesis of Type 2A VHL disease.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Point Mutation , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics , Adult , Cell Line, Tumor , Female , Genetic Association Studies , Genotype , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Middle Aged , Pedigree , Phenotype , Proto-Oncogene Proteins c-jun/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Young Adult , von Hippel-Lindau Disease/diagnosis , von Hippel-Lindau Disease/metabolism
5.
Article in English | MEDLINE | ID: mdl-16869749

ABSTRACT

Biallelic inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene is a common event in hereditary (von Hippel- Lindau disease) and sporadic hemangioblastomas and clear-cell renal carcinomas. Germ-line VHL mutations are also linked to some hereditary pheochromocytoma families. The VHL gene product, pVHL, interacts with a number of cellular proteins and is implicated in the control of angiogenesis, extracellular matrix formation, cell metabolism, and mitogenesis. The best understood function of pVHL relates to its role as the substrate recognition unit of an E3 ligase that targets the heterodimeric transcription factor HIF (hypoxia-inducible factor) for destruction in the presence of oxygen. Down-regulation of HIF appears to be both necessary and sufficient for renal tumor suppression by pVHL, and HIF is strongly suspected of contributing to hemangioblastoma development as well. Recent work suggests that pVHL's role in pheochromocytoma is not related to HIF but rather to the ability of pVHL to regulate neuronal apoptosis, which is mediated by c-Jun, when growth factors such as NGF become limiting. Loss of pVHL leads to up-regulation of JunB, which antagonizes c-Jun and blunts apoptosis.


Subject(s)
Neoplasms/etiology , Oxygen/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Adrenal Gland Neoplasms/etiology , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/physiopathology , Animals , Disease Models, Animal , Genes, Tumor Suppressor , Hemangioblastoma/etiology , Hemangioblastoma/genetics , Hemangioblastoma/physiopathology , Humans , Kidney Neoplasms/etiology , Kidney Neoplasms/genetics , Kidney Neoplasms/physiopathology , Models, Biological , Mutation , Neoplasms/genetics , Neoplasms/physiopathology , Pheochromocytoma/etiology , Pheochromocytoma/genetics , Pheochromocytoma/physiopathology , von Hippel-Lindau Disease/etiology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/physiopathology
6.
Genes Dev ; 15(23): 3104-17, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731475

ABSTRACT

Although MDM2 plays a major role in regulating the stability of the p53 tumor suppressor protein, other poorly understood MDM2-independent pathways also exist. Human adenoviruses have evolved strategies to regulate p53 function and stability to permit efficient viral replication. One mechanism involves adenovirus E1B55K and E4orf6 proteins, which collaborate to target p53 for degradation. To determine the mechanism of this process, a multiprotein E4orf6-associated complex was purified and shown to contain a novel Cullin-containing E3 ubiquitin ligase that is (1) composed of Cullin family member Cul5, Elongins B and C, and the RING-H2 finger protein Rbx1(ROC1); (2) remarkably similar to the von Hippel-Lindau tumor suppressor and SCF (Skp1-Cul1/Cdc53-F-box) E3 ubiquitin ligase complexes; and (3) capable of stimulating ubiquitination of p53 in vitro in the presence of E1/E2 ubiquitin-activating and -conjugating enzymes. Cullins are activated by NEDD8 modification; therefore, to determine whether Cullin complexes are required for adenovirus-induced p53 degradation, studies were conducted in ts41 Chinese hamster ovary cells that are temperature sensitive for the NEDD8 pathway. E4orf6/E1B55K failed to induce the degradation of p53 at the nonpermissive temperature. Thus, our results identify a novel role for the Cullin-based machinery in regulation of p53.


Subject(s)
Adenovirus E1B Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Cell Cycle Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Adenovirus E1B Proteins/chemistry , Animals , Blotting, Western , CHO Cells , Carrier Proteins/metabolism , Cell Line , Cricetinae , Elongin , Humans , Ligases/chemistry , Ligases/metabolism , Macromolecular Substances , Mice , Microscopy, Confocal , Models, Biological , Molecular Weight , Multiprotein Complexes , Protein Binding , Temperature , Transcription Factors/metabolism , Tumor Cells, Cultured , Ubiquitin/metabolism , Ubiquitin-Protein Ligases
8.
Mol Biol Cell ; 12(7): 2207-17, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11452014

ABSTRACT

Different cyclins mediate different cell-cycle transitions. Some cyclins, such as cyclin A and cyclin E, form stable complexes with proteins that bind directly or indirectly to DNA and thus might be recruited to certain regions of the genome at specific times in the cell cycle. Furthermore, cyclins contain structural motifs that are also present in known transcriptional modulators. We found that cyclin A is a potent transcriptional repressor and cyclin E is a potent transcriptional activator when bound to DNA via a heterologous DNA binding domain. The former activity was linked to the integrity of the cyclin A cyclin fold, whereas the latter activity related to the ability of cyclin E to activate cdk2 and recognize substrates. Furthermore, we found that cyclin E, but not cyclin A, activated transcription in a cell-cycle-dependent manner when present in physiological concentrations as an unfused protein. These results suggest that cyclin A and cyclin E intrinsically differ with respect to their ability to modulate transcription when tethered to DNA.


Subject(s)
CDC2-CDC28 Kinases , Cyclin A/metabolism , Cyclin E/metabolism , DNA/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , 3T3 Cells , Animals , Cyclin A/genetics , Cyclin E/genetics , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Doxycycline/pharmacology , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/genetics , Trans-Activators/genetics , Transcription, Genetic/drug effects , Transcriptional Activation , Tumor Cells, Cultured
10.
Dev Biol ; 233(2): 526-36, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11336512

ABSTRACT

The von Hippel-Lindau tumor-suppressor protein (pVHL) regulates the stability of HIF1 alpha and HIF2 alpha and thus is pivotal in cellular responses to changes in oxygen tension. Paradoxically, human cytotrophoblasts proliferate under hypoxic conditions comparable to those measured in the early gestation placenta (2% O(2)), but differentiate into tumorlike invasive cells under well-oxygenated conditions such as those found in the uterus. We sought to explain this phenomenon in terms of pVHL expression. In situ, pVHL immunolocalized to villous cytotrophoblast stem cells, and expression was enhanced at sites of cell column initiation; in both of these relatively hypoxic locations, cytoplasmic staining for HIF2 alpha was also detected. As cytotrophoblasts attached to and invaded the uterus, which results in their increased exposure to oxygen, pVHL staining was abruptly downregulated concordant with localization of HIF2 alpha to the nucleus. In vitro, hypoxia (2% O(2)) upregulated cytotrophoblast pVHL expression together with HIF2 alpha, which localized to the cytoplasm; culture under well-oxygenated conditions greatly reduced levels of both molecules. These results, together with the placental defects previously observed in VHL(-/-) mice, suggest that pVHL is a component of the mechanism that transduces local differences in oxygen tension at the maternal-fetal interface to changes in the biological behavior of cytotrophoblasts. Furthermore, these data provide the first example of oxygen-dependent changes in pVHL abundance.


Subject(s)
Ligases , Proteins/metabolism , Trophoblasts/metabolism , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Basic Helix-Loop-Helix Transcription Factors , Down-Regulation , Female , Fibronectins/metabolism , Humans , Hypoxia/metabolism , Pregnancy , Trans-Activators/metabolism , Up-Regulation , Uterus/metabolism , Von Hippel-Lindau Tumor Suppressor Protein
11.
Hum Mol Genet ; 10(10): 1019-27, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11331612

ABSTRACT

von Hippel-Lindau (VHL) disease is a hereditary cancer syndrome caused by germ line mutation of the von Hippel-Lindau tumor suppressor gene (VHL). Tumors observed in this disorder include retinal and central nervous system hemangioblastomas, clear cell renal carcinomas and pheochromocytomas. The VHL gene product, pVHL, is a component of a ubiquitin ligase which targets the transcription factor known as hypoxia-inducible factor (HIF) for degradation in the presence of oxygen. pVHL also plays roles in the control of extracellular matrix formation and cell-cycle exit. Different VHL mutations confer different site-specific risks of cancer. Type 2C VHL mutations confer an increased risk of pheochromocytoma without the other stigmata of VHL disease. Here we report that the products of such type 2C VHL alleles retain the ability to down regulate HIF but are defective for promotion of fibronectin matrix assembly. Furthermore, pVHL L188V, a well studied type 2C mutant, retained the ability to suppress renal carcinoma growth in vivo. These studies strengthen the notion that HIF deregulation plays a causal role in hemangioblastoma and renal carcinoma, and raises the possibility that abnormal fibronectin matrix assembly contributes to pheochromocytoma pathogenesis in the setting of VHL disease.


Subject(s)
DNA-Binding Proteins/genetics , Down-Regulation , Genes, Tumor Suppressor , Ligases , Nuclear Proteins/genetics , Proteins/physiology , Transcription Factors , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , von Hippel-Lindau Disease/genetics , Alleles , Animals , Carcinoma, Renal Cell/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Fibronectins/genetics , Fibronectins/metabolism , Genotype , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Mice, Nude , Mutation , Nuclear Proteins/metabolism , Phenotype , Proteins/genetics , Proteins/metabolism , Transfection , Tumor Cells, Cultured , Von Hippel-Lindau Tumor Suppressor Protein , von Hippel-Lindau Disease/diagnosis
12.
Apoptosis ; 6(1-2): 17-29, 2001.
Article in English | MEDLINE | ID: mdl-11321038

ABSTRACT

The most recently identified members of the p53 family, p63 and p73, share certain structural and functional similarities with p53. Both p63 and p73 can bind to canonical p53-DNA-binding sites, transactivate the promoters of known p53 target genes and induce apoptosis. Despite these similarities there are many important differences. In contrast to p53, p63 and p73 give rise to multiple distinct protein isoforms that have different functional properties. Upstream signaling pathways involved in the activation of p63 and p73 differ from those involved in p53 activation. Only a subset of the DNA damaging agents that induce p53 can induce p73. Cellular and viral oncoproteins can discriminate between p53 and the newer family members. In addition, the levels of p63 and p73 are affected by certain states of cellular differentiation. Finally, it is becoming clear that the newest members of the p53 family are not classical tumor suppressor genes. In contrast to the high prevalence of p53 mutations in human cancers, p63 and p73 mutations are rare. Indeed, levels of p73 increase during malignant progression. In addition, unlike p53-/- mice, mice lacking p63 and p73 do not develop tumors, but instead have significant developmental abnormalities. Mutations in p63 have also been detected in humans with the ectodermal dysplastic syndrome EEC. Further studies are required to determine whether qualitative or quantitative differences in the expression of p63 and p73 isoforms are important in the development of human cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, p53 , Neoplasms/genetics , Animals , Genetic Predisposition to Disease , Humans , Mutation , Neoplasms/etiology , Protein Isoforms/genetics , Tumor Suppressor Protein p53/genetics
13.
Exp Cell Res ; 264(1): 117-25, 2001 Mar 10.
Article in English | MEDLINE | ID: mdl-11237528

ABSTRACT

Germline mutations of the von Hippel-Lindau tumor suppressor gene (VHL) in humans causes a hereditary cancer syndrome characterized by the development of retinal and central nervous system hemangioblastomas. Other tumors associated with von Hippel-Lindau disease include clear cell renal carcinomas and pheochromocytomas. Tumor development in this setting is due to functional loss of the remaining wild-type VHL allele. Biallelic VHL inactivation is also common in nonhereditary hemangioblastomas and clear cell renal carcinomas, in keeping with Knudson's 2-Hit Model of carcinogenesis. The VHL gene product, pVHL, is a component of an E3 ubiquitin ligase that targets the alpha subunits of the HIF (hypoxia-inducible factor) transcription factor for destruction in the presence of oxygen. Consequently, tumor cells lacking pVHL overproduce the products of HIF target genes such as vascular endothelial growth factor and transforming growth factor alpha. pVHL has been implicated in a variety of processes that are central to carcinogenesis including cell-cycle control, differentiation, extracellular matrix formation and turnover, and angiogenesis.


Subject(s)
Genes, Tumor Suppressor , Ligases , Neoplasms/genetics , Proteins/genetics , Proteins/physiology , Transcription Factors , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , von Hippel-Lindau Disease/genetics , Animals , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Gene Deletion , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Macromolecular Substances , Male , Mice , Models, Biological , Mutation , Nuclear Proteins/metabolism , Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein , von Hippel-Lindau Disease/diagnosis
14.
Curr Opin Genet Dev ; 11(1): 27-34, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11163147
15.
Nat Med ; 6(12): 1335-40, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11100117

ABSTRACT

Chronic hypoxia, a hallmark of many tumors, is associated with angiogenesis and tumor progression. Strategies to treat tumors have been developed in which tumor cells are targeted with drugs or gene-therapy vectors specifically activated under hypoxic conditions. Here we report a different approach, in which the normal transcriptional response to hypoxia is selectively disrupted. Our data indicate that specific blockade of the interaction of hypoxia-inducible factor with the CH1 domain of its p300 and CREB binding protein transcriptional coactivators leads to attenuation of hypoxia-inducible gene expression and diminution of tumor growth. Thus, disrupting the normal co-activational response to hypoxia may be a new and useful therapeutic strategy.


Subject(s)
Cell Hypoxia/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms, Experimental/therapy , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors , Animals , Binding Sites , CREB-Binding Protein , E1A-Associated p300 Protein , Genetic Therapy/methods , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Mice, Nude , Neovascularization, Pathologic , Protein Binding , Transcription, Genetic
16.
Oncogene ; 19(50): 5703-11, 2000 Nov 23.
Article in English | MEDLINE | ID: mdl-11126356

ABSTRACT

The retinoblastoma tumor suppressor protein has been shown to bind directly and inhibit a transcriptionally-important amino-terminal kinase domain of TATA-binding protein-associated factor TAFII250. Cyclin D1 also is able to associate with the amino terminus of TAFII250 in a region very similar to or overlapping the Rb-binding site. In this study, we have examined whether cyclin D1 affects the functional interaction between Rb and TAFII250. We observed that when cyclin D1 is coincubated with Rb and TAFII250, the ability of Rb to inhibit TAFII250 kinase activity is effectively blocked. However, cyclin D1 by itself has no apparent effect on TAFII250 kinase activity. We further found that the Rb-related protein p107 can inhibit TAFII250 kinase activity, and this inhibition is likewise alleviated by cyclin D1. Cyclin D1 prevents the kinase-inhibitory effect of an Rb mutant unable to bind to D-type cyclins, indicating that it is acting through its association with TAFII250 and not with Rb. However, we found no evidence of TAFII250-binding competition between Rb and cyclin D1 in vitro. The adenovirus E1A protein, which also binds to both Rb and TAFII250, exhibited a suppressive effect on Rb-mediated kinase inhibition similar to that seen with cyclin D1. Our results suggest a novel means by which cyclin D1 may be able to independently regulate the activity of Rb.


Subject(s)
Cyclin D1/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors , Retinoblastoma Protein/metabolism , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Binding, Competitive , Cyclin D1/genetics , Cyclin D1/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Acetyltransferases , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107 , TATA Box
17.
Mol Cell Biol ; 20(23): 8889-902, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11073989

ABSTRACT

Control of proliferation and differentiation by the retinoblastoma tumor suppressor protein (pRB) and related family members depends upon their interactions with key cellular substrates. Efforts to identify such cellular targets led to the isolation of a novel protein, EID-1 (for E1A-like inhibitor of differentiation 1). Here, we show that EID-1 is a potent inhibitor of differentiation and link this activity to its ability to inhibit p300 (and the highly related molecule, CREB-binding protein, or CBP) histone acetylation activity. EID-1 is rapidly degraded by the proteasome as cells exit the cell cycle. Ubiquitination of EID-1 requires an intact C-terminal region that is also necessary for stable binding to p300 and pRB, two proteins that bind to the ubiquitin ligase MDM2. A pRB variant that can bind to EID1, but not MDM2, stabilizes EID-1 in cells. Thus, EID-1 may act at a nodal point that couples cell cycle exit to the transcriptional activation of genes required for differentiation.


Subject(s)
Adenovirus E1A Proteins/metabolism , Cell Cycle/physiology , Saccharomyces cerevisiae Proteins , Acetyltransferases/antagonists & inhibitors , Amino Acid Sequence , CREB-Binding Protein , Cell Cycle Proteins , Cell Differentiation , Cloning, Molecular , Down-Regulation , Histone Acetyltransferases , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Repressor Proteins , Retinoblastoma Protein/metabolism , Trans-Activators/antagonists & inhibitors , Transcriptional Activation , Two-Hybrid System Techniques , Ubiquitins/metabolism
18.
Nature ; 407(6804): 642-5, 2000 Oct 05.
Article in English | MEDLINE | ID: mdl-11034214

ABSTRACT

Strong stimulation of the T-cell receptor (TCR) on cycling peripheral T cells causes their apoptosis by a process called TCR-activation-induced cell death (TCR-AICD). TCR-AICD occurs from a late G1 phase cell-cycle check point independently of the 'tumour suppressor' protein p53. Disruption of the gene for the E2F-1 transcription factor, an inducer of apoptosis, causes significant increases in T-cell number and splenomegaly. Here we show that T cells undergoing TCR-AICD induce the p53-related gene p73, another mediator of apoptosis, which is hypermethylated in lymphomas. Introducing a dominant-negative E2F-1 protein or a dominant-negative p73 protein into T cells protects them from TCR-mediated apoptosis, whereas dominant-negative E2F-2, E2F-4 or p53 does not. Furthermore, E2F-1-null or p73-null primary T cells do not undergo TCR-mediated apoptosis either. We conclude that TCR-AICD occurs from a late G1 cell-cycle checkpoint that is dependent on both E2F-1 and p73 activities. These observations indicate that, unlike p53, p73 serves to integrate receptor-mediated apoptotic stimuli.


Subject(s)
Apoptosis , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/physiology , Transcription Factors/physiology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F4 Transcription Factor , G1 Phase , Gene Products, tat/genetics , Gene Products, tat/metabolism , Genes, Tumor Suppressor , Humans , In Situ Nick-End Labeling , Jurkat Cells , Metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Recombinant Fusion Proteins , Retinoblastoma-Binding Protein 1 , T-Lymphocytes/metabolism , Transcription Factor DP1 , Transcription Factors/metabolism , Tumor Protein p73 , Tumor Suppressor Proteins
19.
Nature ; 407(6804): 645-8, 2000 Oct 05.
Article in English | MEDLINE | ID: mdl-11034215

ABSTRACT

The transcription factor E2F-1 induces both cell-cycle progression and, in certain settings, apoptosis. E2F-1 uses both p53-dependent and p53-independent pathways to kill cells. The p53-dependent pathway involves the induction by E2F-1 of the human tumour-suppressor protein p14ARF, which neutralizes HDM2 (human homologue of MDM2) and thereby stabilizes the p53 protein. Here we show that E2F-1 induces the transcription of the p53 homologue p73. Disruption of p73 function inhibited E2F-1-induced apoptosis in p53-defective tumour cells and in p53-/- mouse embryo fibroblasts. We conclude that activation of p73 provides a means for E2F-1 to induce death in the absence of p53.


Subject(s)
Apoptosis , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Cell Line , DNA/metabolism , DNA-Binding Proteins/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation , Genes, Tumor Suppressor , Mice , Mutation , Nuclear Proteins/genetics , Protein Binding , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription, Genetic , Tumor Protein p73 , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins
20.
Genes Dev ; 14(17): 2185-91, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10970882

ABSTRACT

The c-Myc oncoprotein plays an important role in the growth and proliferation of normal and neoplastic cells. To execute these actions, c-Myc is thought to regulate functionally diverse sets of genes that directly govern cellular mass and progression through critical cell cycle transitions. Here, we provide several lines of evidence that c-Myc promotes ubiquitin-dependent proteolysis by directly activating expression of the Cul1 gene, encoding a critical component of the ubiquitin ligase SCF(SKP2). The cell cycle inhibitor p27(kip1) is a known target of the SCF(SKP2) complex, and Myc-induced Cul1 expression matched well with the kinetics of declining p27(kip1) protein. Enforced Cul1 expression or antisense neutralization of p27(kip1) was capable of overcoming the slow-growth phenotype of c-Myc null primary mouse embryonic fibroblasts (MEFs). In reconstitution assays, the addition of in vitro translated Cul1 protein alone was able to restore p27(kip1) ubiquitination and degradation in lysates derived from c-myc(-/-) MEFs or density-arrested human fibroblasts. These functional and biochemical data provide a direct link between c-Myc transcriptional regulation and ubiquitin-mediated proteolysis and together support the view that c-Myc promotes G(1) exit in part via Cul1-dependent ubiquitination and degradation of the CDK inhibitor, p27(kip1).


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins/genetics , Cell Cycle , Cullin Proteins , Gene Expression Regulation, Developmental , Peptide Synthases/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins , Ubiquitins/metabolism , 3T3 Cells , Animals , Blotting, Northern , Cell Cycle Proteins/metabolism , Cell Division , Cyclin A/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Humans , Immunoblotting , Luciferases/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Peptide Synthases/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Retroviridae/genetics , S Phase , SKP Cullin F-Box Protein Ligases , Time Factors , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...