Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Nanotechnology ; 32(2): 025708, 2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33055385

ABSTRACT

During neuronal development, neuronal cells read extracellular stimuli from the micro/nano-environment within which they exist, retrieving essential directionality and wiring information. Here, focal adhesions (FAs-protein clusters anchoring integrins to cytoskeleton) act as sensors, by integrating signals from both the extracellular matrix environment and chemotactic factors, contributing to the final neuronal pathfinding and migration. In the processes that orchestrate neuronal development, the important function of ubiquitin E3A ligase (UBE3A) is emerging. UBE3A has crucial functions in the brain and changes in its expression levels lead to neurodevelopmental disorders: the lack of UBE3A leads to Angelman syndrome (AS, OMIN 105830), while its increase causes autisms (Dup15q-autism). By using nano/micro-structured anisotropic substrates we previously showed that UBE3A-deficient neurons have deficits in contact guidance (Tonazzini et al, Mol Autism 2019). Here, we investigate the adhesion and migration dynamics of UBE3A-silenced SH-SY5Y neuroblastoma cells in vitro by exploiting nano/micro-grooved substrates. We analyze the molecular processes regulating the development of FAs by transfection with EGFP-vector encoding for paxillin, a protein of FA clusters, and by live-cell total-internal-reflection-fluorescence microscopy. We show that UBE3A-silenced SH-SY5Y cells have impaired FA morphological development and pathway activation, which lead to a delayed adhesion and also explain the defective contact guidance in response to directional topographical stimuli. However, UBE3A-silenced SH-SY5Y cells show an overall normal migration behavior, in terms of speed and ability to follow the GRs directional stimulus. Only the collective cell migration upon cell gaps was slightly delayed for UBE3Ash SHs. Overall, the deficits of UBE3Ash SHS-SY5Y cells in FA maturation/sensing and in collective migration may have patho-physiological implications, in AS condition, considering the much more complex stimuli that neurons find in vivo during the neurodevelopment.


Subject(s)
Cell Adhesion , Cell Movement , Gene Silencing , Neurons/cytology , Ubiquitin-Protein Ligases/genetics , Cell Line, Tumor , Focal Adhesions/genetics , Focal Adhesions/metabolism , Humans , Nanostructures/chemistry , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neurons/metabolism , Surface Properties , Ubiquitin-Protein Ligases/metabolism
2.
Cell Death Differ ; 16(8): 1156-66, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19325566

ABSTRACT

The promyelocytic leukemia (PML) tumor suppressor is essential for the formation of PML nuclear bodies (NBs). PML and PML-NBs have been implicated in the regulation of growth inhibition, senescence and apoptosis. PML is activated in response to stress signals and is downregulated in certain human cancers. However, the factors mediating PML stability are incompletely understood. Here we demonstrate that a catalytically active form of the mammalian E3 ligase E6AP (HPV E6-associated protein) acts to reduce the half-life of the PML protein by promoting its degradation in the proteasome. E6AP mediates the ubiquitination of PML in an in vitro ubiquitination assay. E6AP and PML interact at physiological levels and colocalize in PML-NBs. Importantly, PML protein expression is elevated in multiple organs and cell types from E6AP null mice and in lymphoid cells is associated with increased number and intensity of PML-NBs. This PML elevation is enhanced in response to DNA damage. Our results identify E6AP as an important regulator of PML and PML-NBs.


Subject(s)
Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Down-Regulation , Humans , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Transcription Factors/deficiency , Transcription Factors/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
4.
Proc Natl Acad Sci U S A ; 98(3): 1218-23, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11158620

ABSTRACT

The E6 oncoprotein of human papillomaviruses (HPVs) that are associated with cervical cancer utilizes the cellular ubiquitin-protein ligase E6-AP to target the tumor suppressor p53 for degradation. In normal cells (i.e., in the absence of E6), p53 is also a target of the ubiquitin-proteasome pathway. Under these conditions, however, p53 degradation is mediated by Mdm2 rather than by E6-AP. Here we show in a mutational analysis that, surprisingly, the structural requirements of p53 to serve as a proteolytic substrate differ between E6 proteins derived from different HPV types and, as expected, between Mdm2 and E6 proteins in vitro and in vivo. Stable expression of such mutants in HPV-negative and HPV-positive cell lines demonstrates that in HPV-positive cancer cells, the E6-dependent pathway of p53 degradation is not only active but, moreover, is required for degradation of p53, whereas the Mdm2-dependent pathway is inactive. Because the p53 pathway was reported to be functional in HPV-positive cancer cells, this finding indicates clearly that the ability of the E6 oncoprotein to target p53 for degradation is required for the growth of HPV-positive cancer cells.


Subject(s)
Nuclear Proteins , Oncogene Proteins, Viral/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Female , Half-Life , Humans , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Proto-Oncogene Proteins c-mdm2 , Recombinant Proteins/metabolism , Substrate Specificity , Transfection , Tumor Cells, Cultured , Ubiquitins/metabolism , Uterine Cervical Neoplasms
5.
Oncogene ; 19(51): 5944-50, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11127826

ABSTRACT

The E7 oncoprotein of the high risk human papillomavirus type 16 (HPV-16), which is etiologically associated with uterine cervical cancer, is a potent immortalizing and transforming agent. It probably exerts its oncogenic functions by interacting and altering the normal activity of cell cycle control proteins such as p21WAF1, p27KIP1 and pRb, transcriptional activators such as TBP and AP-1, and metabolic regulators such as M2-pyruvate kinase (M2-PK). Here we show that E7 is a short-lived protein and its degradation both in vitro and in vivo is mediated by the ubiquitin-proteasome pathway. Interestingly, ubiquitin does not attach to any of the two internal Lysine residues of E7. Substitution of these residues with Arg does not affect the ability of the protein to be conjugated and degraded; in contrast, addition of a Myc tag to the N-terminal but not to the C-terminal residue, stabilizes the protein. Also, deletion of the first 11 amino acid residues stabilizes the protein in cells. Taken together, these findings strongly suggest that, like MyoD and the Epstein Barr Virus (EBV) transforming Latent Membrane Protein 1 (LMPI), the first ubiquitin moiety is attached linearly to the free N-terminal residue of E7. Additional ubiquitin moieties are then attached to an internal Lys residue of the previously conjugated molecule. The involvement of E7 in many diverse and apparently unrelated processes requires tight regulation of its function and cellular level, which is controlled in this case by ubiquitin-mediated proteolysis.


Subject(s)
Multienzyme Complexes/metabolism , Oncogene Proteins, Viral/metabolism , Ubiquitin-Conjugating Enzymes , Ubiquitins/metabolism , Adenosine Triphosphate/metabolism , Animals , COS Cells , Cell-Free System , Chlorocebus aethiops , HeLa Cells , Humans , Ligases/metabolism , Lysine/genetics , Lysine/metabolism , Mutagenesis, Site-Directed , Papillomavirus E7 Proteins , Protein Structure, Tertiary , Rabbits , Ubiquitins/genetics
6.
Proc Natl Acad Sci U S A ; 97(12): 6693-7, 2000 Jun 06.
Article in English | MEDLINE | ID: mdl-10829072

ABSTRACT

Certain types of human papillomaviruses (HPVs) are closely linked to the development of human cancers. Herein, it is shown that intracellular targeting of the HPV16 E6 oncoprotein by E6-binding peptide aptamers resulted in the apoptotic elimination of HPV16-positive cancer cells, whereas HPV-negative cells were not affected. These results provide direct experimental evidence that the HPV E6 oncoprotein has antiapoptotic activity in HPV-positive tumor cells that is required for their survival. The E6-targeting molecules identified herein have implications for the development of therapeutic strategies for the treatment of HPV-associated dysplasias and cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Neoplasms/virology , Oncogene Proteins, Viral/metabolism , Papillomaviridae/drug effects , Peptides/pharmacology , Repressor Proteins , Amino Acid Sequence , HeLa Cells , Humans , Molecular Sequence Data , Peptides/metabolism , Peptides/therapeutic use
7.
Eur J Biochem ; 267(12): 3712-22, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848989

ABSTRACT

According to our current knowledge, protein ubiquitination involves three steps: activation of ubiquitin through formation of an energy-rich bond with an E1 ubiquitin-activating enzyme; and transfer of activated ubiquitin onto E2 ubiquitin-conjugating enzymes, which, in turn, alone, or in combination with E3 ubiquitin-protein ligase enzymes, transfer ubiquitin onto target proteins. A31N-ts20 cells are mouse embryo fibroblasts, thermosensitive for E1. We show here that: (a) the enzymatic activity of the enzyme is heat-inactivatable in vitro; and (b) a major mechanism responsible for E1 inactivation in vivo consists of accelerated destruction. Surprisingly, a >90% reduction in E1 abundance little alters the formation of the bulk of protein-ubiquitin conjugates when A31N-ts20 cells are grown at the nonpermissive temperature, indicating that cautious interpretation of results is required when studying ubiquitination of specific substrates using this cell line. Surprisingly, our data also indicate that, in vivo, ubiquitination of the various protein substrates in A31N-ts20 cells requires different amounts of E1, indicating that this mutant cell line can be used for unveiling the existence of differences in the intimate mechanisms responsible for the ubiquitination of the various cell proteins in vivo, and for providing criteria of reliability when developing in vitro ubiquitination assays for specific proteins.


Subject(s)
Ligases/genetics , Ligases/metabolism , Animals , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Fibroblasts , Histones/metabolism , Hot Temperature , Leupeptins/pharmacology , Ligases/antagonists & inhibitors , Mice , Mutation , Proto-Oncogene Proteins c-jun/metabolism , Temperature , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Activating Enzymes , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
8.
EMBO J ; 18(22): 6462-71, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10562558

ABSTRACT

The growth-suppressive properties of p53 are controlled by posttranslational modifications and by regulation of its turnover rate. Here we show that p53 can be modified in vitro and in vivo by conjugation to the small ubiquitin-like protein SUMO-1. A lysine residue at amino acid position 386 of p53 is required for this previously undescribed modification, strongly suggesting that this lysine residue serves as the major attachment site for SUMO-1. Unlike ubiquitin, attachment of SUMO-1 does not appear to target proteins for rapid degradation but rather, has been proposed to change the ability of the modified protein to interact with other cellular proteins. Accordingly, we provide evidence that conjugation of SUMO-1 to wild-type p53 results in an increased transactivation ability of p53. We suggest that posttranslational modification of p53 by SUMO-1 conjugation provides a novel mechanism to regulate p53 activity.


Subject(s)
Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Humans , Kinetics , Lysine , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , SUMO-1 Protein , Transfection , Tumor Cells, Cultured , Ubiquitins/chemistry
9.
Nat Cell Biol ; 1(1): 14-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10559858

ABSTRACT

The transcription factor E2F-1 is important in the control of cell proliferation. Its activity must be tightly regulated in a cell-cycle-dependent manner to enable programs of gene expression to be coupled closely with cell-cycle position. Here we show that, following its accumulation in the late G1 phase of the cell cycle, E2F-1 is rapidly degraded in S/G2 phase. This event is linked to a specific interaction of E2F-1 with the F-box-containing protein p45SKP2, which is the cell-cycle-regulated component of the ubiquitin-protein ligase SCFSKP2 that recognizes substrates for this ligase. Disruption of the interaction between E2F-1 and p45SKP2 results in a reduction in ubiquitination of E2F-1 and the stabilization and accumulation of transcriptionally active E2F-1 protein. These results indicate that an SCFSKP2-dependent ubiquitination pathway may be involved in the downregulation of E2F-1 activity in the S/G2 phase of the cell cycle, and suggest a link between SCFSKP2 and cell-cycle-dependent gene control.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , DNA-Binding Proteins , Ligases/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , G1 Phase , G2 Phase , HeLa Cells , Homeostasis , Humans , Kinetics , Mice , Retinoblastoma-Binding Protein 1 , S Phase , S-Phase Kinase-Associated Proteins , Substrate Specificity , Transcription Factor DP1 , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
10.
J Biol Chem ; 274(43): 30963-8, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10521492

ABSTRACT

Ubiquitinylation of proteins appears to be mediated by the specific interplay between ubiquitin-conjugating enzymes (E2s) and ubiquitin-protein ligases (E3s). However, cognate E3s and/or substrate proteins have been identified for only a few E2s. To identify proteins that can interact with the human E2 UbcH7, a yeast two-hybrid screen was performed. Two proteins were identified and termed human homologue of Drosophila ariadne (HHARI) and UbcH7-associated protein (H7-AP1). Both proteins, which are widely expressed, are characterized by the presence of RING finger and in between RING fingers (IBR) domains. No other overt structural similarity was observed between the two proteins. In vitro binding studies revealed that an N-terminal RING finger motif (HHARI) and the IBR domain (HHARI and H7-AP1) are involved in the interaction of these proteins with UbcH7. Furthermore, binding of these two proteins to UbcH7 is specific insofar that both HHARI and H7-AP1 can bind to the closely related E2, UbcH8, but not to the unrelated E2s UbcH5 and UbcH1. Although it is not clear at present whether HHARI and H7-AP1 serve, for instance, as substrates for UbcH7 or represent proteins with E3 activity, our data suggests that a subset of RING finger/IBR proteins are functionally linked to the ubiquitin/proteasome pathway.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/chemistry , Cloning, Molecular , Drosophila , Humans , Insect Proteins/chemistry , Insect Proteins/metabolism , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Sequence Alignment , Sequence Homology, Amino Acid , Transferases , Ubiquitin-Protein Ligases
11.
Mamm Genome ; 10(10): 977-82, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10501966

ABSTRACT

The ubiquitination pathway targets not only normal (short-lived) intracellular eukaryotic proteins for degradation when appropriate, but also serves to eliminate mutant/misfolded proteins from the cell. An understanding of the molecular basis of the interaction between the ubiquitin-conjugating enzymes (E2s), ubiquitin protein ligases (E3s), and target proteins is essential to explain the process in normal cellular function and in disease. UbcM4 is the mouse ortholog of the human E2, UbcH7, which can participate in the in vitro degradation of many proteins including p53. We describe the characterization of the mouse UbcM4 gene and the identification of a UbcM4 pseudogene. Four UbcM4 transcripts of approximately 0.7, 1.5, 2.1, and 2.6 kb, observed on Northern blots, are differentiated by their utilization of alternative UbcM4 polyadenylation sites. A single alternative splice variant cDNA, termed UbcM4Deltaex2, was also identified. The polypeptide encoded by UbcM4Deltaex2 is incapable of forming an ubiquitin-thioester in contrast to UbcM4, despite retaining the key cysteine residue essential for ubiquitin thioester formation and the active site consensus sequence that defines the ubiquitin-conjugating enzyme class. These observations are of particular relevance for analysis of UbcM4 function in vivo as our studies indicate that the targeted deletion of the coding exon absent in UbcM4Deltaex2 would produce an inactive UbcM4 protein and presents an alternative to disruption of its transcriptional initiation site/promoter region. Furthermore, it suggests that a similar strategy may be applicable to disrupt the function of other ubiquitin-conjugating enzymes in vivo.


Subject(s)
Ligases/genetics , Ubiquitin-Conjugating Enzymes , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Mice , Molecular Sequence Data , Pseudogenes , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins , Ubiquitins/metabolism
12.
J Biol Chem ; 274(28): 19581-6, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10391892

ABSTRACT

The ubiquitin/proteasome pathway mediates the degradation of many short-lived proteins that are critically involved in the regulation of cell proliferation and cell death, including the tumor suppressor protein p53. Accumulation of p53 and induction of apoptosis in RAW 264.7 macrophages in response to nitric oxide are well established. However, the molecular mechanisms involved in nitric oxide-induced p53 accumulation are unknown. Here we show that, similar to nitric oxide, treatment of macrophages with specific proteasome inhibitors, including clastolactacystin-beta-lactone, induces p53 accumulation and apoptosis, suggesting that nitric oxide may affect the activity of the proteasome. In support of this hypothesis, both exposure of cells to S-nitrosoglutathione and stimulation of endogenous nitric oxide production by lipopolysaccharide/interferon-gamma treatment result in inhibition of proteasome activity as measured in vitro by the degradation of the proteasome-specific substrate succinyl-Leu-Leu-Val-Tyr-4-methylcoumarin-7-amide. Moreover, chemically diverse nitric oxide donors interfere with proteasome-mediated degradation of polyubiquitinated p53 in vitro. These data imply that nitric oxide-induced apoptosis and accumulation of p53 are, at least in part, mediated by inhibition of the proteasome.


Subject(s)
Apoptosis/drug effects , Cysteine Endopeptidases/drug effects , Multienzyme Complexes/drug effects , Nitric Oxide/pharmacology , Animals , Coumarins/metabolism , Cysteine Endopeptidases/metabolism , DNA Fragmentation , Glutathione/analogs & derivatives , Glutathione/pharmacology , Macrophages/drug effects , Mice , Multienzyme Complexes/metabolism , Nitroso Compounds/pharmacology , Oligopeptides/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex , S-Nitrosoglutathione , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Ubiquitins/metabolism
14.
J Biol Chem ; 274(11): 7576-82, 1999 Mar 12.
Article in English | MEDLINE | ID: mdl-10066826

ABSTRACT

Members of the hect domain protein family are characterized by sequence similarity of their C-terminal regions to the C terminus of E6-AP, an E3 ubiquitin-protein ligase. An essential intermediate step in E6-AP-dependent ubiquitination is the formation of a thioester complex between E6-AP and ubiquitin in the presence of distinct E2 ubiquitin-conjugating enzymes including human UbcH5, a member of the UBC4/UBC5 subfamily of E2s. Similarly, several hect domain proteins, including Saccharomyces cerevisiae RSP5, form ubiquitin thioester complexes, indicating that hect domain proteins in general have E3 activity. We show here, by the use of chimeric E2s generated between UbcH5 and other E2s, that a region of UbcH5 encompassing the catalytic site cysteine residue is critical for its ability to interact with E6-AP and RSP5. Of particular importance is a phenylalanine residue at position 62 of UbcH5 that is conserved among the members of the UBC4/UBC5 subfamily but is not present in any of the other known E2s, whereas the N-terminal 60 amino acids do not contribute significantly to the specificity of these interactions. The conservation of this phenylalanine residue throughout evolution underlines the importance of the ability to interact with hect domain proteins for the cellular function of UBC4/UBC5 subfamily members.


Subject(s)
Fungal Proteins/metabolism , Ligases/metabolism , Saccharomyces cerevisiae Proteins , Ubiquitin-Protein Ligase Complexes , Amino Acid Sequence , Conserved Sequence , Endosomal Sorting Complexes Required for Transport , Ligases/chemistry , Molecular Sequence Data , Phenylalanine/chemistry , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases
15.
J Virol ; 72(12): 9470-8, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9811680

ABSTRACT

The 55-kDa E1B protein of adenovirus, which binds to and inactivates the tumor suppressor protein p53, is not expressed in the adenoviral mutant termed ONYX-015 (i.e., dl1520). It was reported that the mutant virus due to a deletion in E1B is able to replicate only in cells deficient for wild-type p53. Accordingly, dl1520 is currently being evaluated as a potential tool in the therapy of p53 deficient cancers. In contrast, we report here that dl1520 replicates independently of the p53 status in various tumor cell lines (U87, RKO, A549, H1299, and U373). In addition, the inhibition of p53-mediated transcriptional activation in wild-type p53 containing U2OS cells, by overexpression of a transdominant negative p53 mutant, did not render the cells permissive for dl1520 replication. Finally, we show that, depending on the multiplicity of infection, the deleted virus is able to replicate in and to kill primary human cells. Thus, the molecular basis for the growth differences of dl1520 within different cell types remains to be determined.


Subject(s)
Adenoviruses, Human/physiology , Tumor Suppressor Protein p53/metabolism , Virus Replication , Adenoviruses, Human/genetics , Cells, Cultured , Cytopathogenic Effect, Viral , Defective Viruses/genetics , Defective Viruses/physiology , Gene Expression , Genetic Therapy , Genetic Vectors , Humans , Mutation , Neoplasms/metabolism , Neoplasms/therapy , Neoplasms/virology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
16.
Eur J Biochem ; 254(3): 643-9, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9688277

ABSTRACT

Recognition of substrate proteins by the ubiquitin-conjugation system is a highly specific and regulated event and involves the action of ubiquitin-conjugating enzymes (E2) and ubiquitin-protein ligases (E3). However, the E2 and E3 involved in the recognition of particular substrates have been identified in only a few cases. The ubiquitin-protein ligase E6-associated protein (E6-AP) was originally identified as a protein involved in the human papillomavirus E6-oncoprotein-induced degradation of p53. The substrate proteins of E6-AP in the absence of the E6 oncoprotein, however, have not been identified. We show here that E6-AP can target itself for ubiquitination in vitro and provide evidence that, under conditions of overexpression, E6-AP efficiently promotes its own degradation in vivo. Autoubiquitination of E6-AP is mediated mainly by intermolecular transfer of ubiquitin. In addition, highly ubiquitinated forms of E6-AP cannot bind to p53 in the presence of the E6 oncoprotein and, conversely, binding of E6-AP to p53 interferes with ubiquitination of E6-AP. These results suggest that autoubiquitination and subsequent degradation of E6-AP represents a mechanism to control intracellular E6-AP levels by inactivating E6-AP molecules that are not bound to substrate proteins.


Subject(s)
Ligases/metabolism , Ubiquitins/metabolism , Plasmids , Recombinant Proteins/metabolism , Substrate Specificity , Ubiquitin-Protein Ligases
17.
Pharmacol Ther ; 78(3): 129-39, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9690814

ABSTRACT

Inactivation of the tumor suppressor protein p53 plays a key role in human carcinogenesis. Activation of the growth-suppressive properties of p53 by appropriate stress signals, including genotoxic stress, is generally accompanied by intracellular accumulation of the protein. This suggests that stabilization of the otherwise short-lived protein is an intrinsic feature of p53 activation. The ubiquitin/proteasome system is believed to be a major proteolytic system involved in selective degradation of cell regulatory proteins. In this review, the potential role of the ubiquitin/proteasome system in p53 degradation and possible mechanisms involved in p53 stability regulation are discussed.


Subject(s)
Ligases/physiology , Tumor Suppressor Protein p53/physiology , Ubiquitins/physiology , Cysteine Endopeptidases/metabolism , Humans , Multienzyme Complexes/metabolism , Neoplasms/metabolism , Proteasome Endopeptidase Complex , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases
18.
J Biol Chem ; 273(20): 12148-54, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9575161

ABSTRACT

The hect domain protein family was originally identified by sequence similarity of its members to the C-terminal region of E6-AP, an E3 ubiquitin-protein ligase. Since the C terminus of E6-AP mediates thioester complex formation with ubiquitin, a necessary intermediate step in E6-AP-dependent ubiquitination, it was proposed that members of the hect domain family in general have E3 activity. The hect domain is approximately 350 amino acids in length, and we show here that the hect domain of E6-AP is necessary and sufficient for ubiquitin thioester adduct formation. Furthermore, the human genome encodes at least 20 different hect domain proteins, and in further support of the hypothesis that hect domain proteins represent a family of E3s, several of these are shown to form thioester complexes with ubiquitin. In addition, some hect domain proteins interact preferentially with UbcH5, whereas others interact with UbcH7, indicating that human hect domain proteins can be grouped into at least two classes based on their E2 specificity. Since E3s are thought to play a major role in substrate recognition, the presence of a large family of E3s should contribute to ensure the specificity and selectivity of ubiquitin-dependent proteolytic pathways.


Subject(s)
Fungal Proteins/metabolism , Ligases/metabolism , Saccharomyces cerevisiae Proteins , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligase Complexes , Amino Acid Sequence , Endosomal Sorting Complexes Required for Transport , Fungal Proteins/chemistry , Humans , Ligases/chemistry , Molecular Sequence Data , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
19.
J Clin Microbiol ; 36(2): 475-80, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9466762

ABSTRACT

Cervical cancer is the most prevalent tumor in developing countries and the second most frequent cancer among females worldwide. Specific human papillomaviruses (HPVs) and, most notably, HPV types 16 and 18 are recognized as being causally associated with this malignancy. Antibodies against early HPV proteins E6 and E7 have been found more often in patients with tumors than in controls. Existing peptide enzyme-linked immunosorbent assays (ELISAs) for the detection of anti-E6 and anti-E7 antibodies in human sera have low levels of sensitivity and specificity and thus are not suitable for use as diagnostic tools. Based on highly purified recombinant native proteins, we developed four sandwich ELISAs for the detection of antibodies against HPV type 16 and 18 E6 and E7 proteins. We demonstrate their sensitivities and high degrees of specificity for cervical cancer. Among a total of 501 serum specimens from unselected patients with invasive cervical cancer, 52.9% reacted positively in at least one of the four assays. In contrast, among 244 serum specimens from control subjects without cervical cancer, only 2 reactive serum specimens (0.8%) were found. For 19 of 19 antibody-positive patients, the HPV type indicated by seroreactivity was identical to the HPV DNA type found in the tumor, which also indicates a high degree of specificity for antibody detection with respect to HPV type. In a direct comparison of 72 serum specimens from patients with cervical cancer, 56% of the specimens reacted in at least one of the four protein ELISAs, whereas 40% reacted in at least one of seven peptide ELISAs covering the four antigens. These assays could be of value for the detection of invasive cervical cancer in settings in which cytology-based early tumor screening is not available, for the clinical management of patients diagnosed with cervical cancer, and for the immunological monitoring of E6 and E7 vaccination trials.


Subject(s)
Antibodies, Viral/immunology , DNA-Binding Proteins , Enzyme-Linked Immunosorbent Assay/methods , Papillomaviridae/immunology , Papillomavirus Infections/diagnosis , Repressor Proteins , Tumor Virus Infections/diagnosis , Uterine Cervical Neoplasms/virology , Adult , Aged , Antigens, Viral/genetics , Antigens, Viral/immunology , Female , Humans , Middle Aged , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Papillomavirus Infections/blood , Papillomavirus Infections/immunology , Recombinant Proteins/immunology , Sensitivity and Specificity , Tumor Virus Infections/blood , Tumor Virus Infections/immunology , Uterine Cervical Neoplasms/blood
20.
Proc Natl Acad Sci U S A ; 95(2): 560-4, 1998 Jan 20.
Article in English | MEDLINE | ID: mdl-9435231

ABSTRACT

The ubiquitin-like protein SMT3 from Saccharomyces cerevisiae and SUMO-1, its mammalian homolog, can be covalently attached to other proteins posttranslationally. Conjugation of ubiquitin requires the activities of ubiquitin-activating (E1) and -conjugating (E2) enzymes and proceeds via thioester-linked enzyme-ubiquitin intermediates. Herein we show that UBC9, one of the 13 different E2 enzymes from yeast, is required for SMT3 conjugation in vivo. Moreover, recombinant yeast and mammalian UBC9 enzymes were found to form thioester complexes with SMT3 and SUMO-1, respectively. This suggests that UBC9 functions as an E2 in a SMT3/SUMO-1 conjugation pathway analogous to ubiquitin-conjugating enzymes. The role of yeast UBC9 in cell cycle progression may thus be mediated through its SMT3 conjugation activity.


Subject(s)
Fungal Proteins/metabolism , Ligases/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins , Ubiquitin-Conjugating Enzymes , Ubiquitins/metabolism , Ligases/genetics , Mutation , SUMO-1 Protein , Saccharomyces cerevisiae , Small Ubiquitin-Related Modifier Proteins , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...