Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Rev Cancer ; 1879(5): 189149, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38971209

ABSTRACT

Flavins and their associated proteins have recently emerged as compelling players in the landscape of cancer biology. Flavins, encompassing flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), serve as coenzymes in a multitude of cellular processes, such as metabolism, apoptosis, and cell proliferation. Their involvement in oxidative phosphorylation, redox homeostasis, and enzymatic reactions has long been recognized. However, recent research has unveiled an extended role for flavins in the context of cancer. In parallel, riboflavin transporters (RFVTs), FAD synthase (FADS), and riboflavin kinase (RFK) have gained prominence in cancer research. These proteins, responsible for riboflavin uptake, FAD biosynthesis, and FMN generation, are integral components of the cellular machinery that governs flavin homeostasis. Dysregulation in the expression/function of these proteins has been associated with various cancers, underscoring their potential as diagnostic markers, therapeutic targets, and key determinants of cancer cell behavior. This review embarks on a comprehensive exploration of the multifaceted role of flavins and of the flavoproteins involved in nucleus-mitochondria crosstalk in cancer. We journey through the influence of flavins on cancer cell energetics, the modulation of RFVTs in malignant transformation, the diagnostic and prognostic significance of FADS, and the implications of RFK in drug resistance and apoptosis. This review also underscores the potential of these molecules and processes as targets for novel diagnostic and therapeutic strategies, offering new avenues for the battle against this relentless disease.

2.
mSphere ; 8(2): e0005623, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36883841

ABSTRACT

Tumor suppressor p53 and its related proteins, p63 and p73, can be synthesized as multiple isoforms lacking part of the N- or C-terminal regions. Specifically, high expression of the ΔNp73α isoform is notoriously associated with various human malignancies characterized by poor prognosis. This isoform is also accumulated by oncogenic viruses, such as Epstein-Barr virus (EBV), as well as genus beta human papillomaviruses (HPV) that appear to be involved in carcinogenesis. To gain additional insight into ΔNp73α mechanisms, we have performed proteomics analyses using human keratinocytes transformed by the E6 and E7 proteins of the beta-HPV type 38 virus as an experimental model (38HK). We find that ΔNp73α associates with the E2F4/p130 repressor complex through a direct interaction with E2F4. This interaction is favored by the N-terminal truncation of p73 characteristic of ΔNp73 isoforms. Moreover, it is independent of the C-terminal splicing status, suggesting that it could represent a general feature of ΔNp73 isoforms (α, ß, γ, δ, ε, ζ, θ, η, and η1). We show that the ΔNp73α-E2F4/p130 complex inhibits the expression of specific genes, including genes encoding for negative regulators of proliferation, both in 38HK and in HPV-negative cancer-derived cell lines. Such genes are not inhibited by E2F4/p130 in primary keratinocytes lacking ΔNp73α, indicating that the interaction with ΔNp73α rewires the E2F4 transcriptional program. In conclusion, we have identified and characterized a novel transcriptional regulatory complex with potential implications in oncogenesis. IMPORTANCE The TP53 gene is mutated in about 50% of human cancers. In contrast, the TP63 and TP73 genes are rarely mutated but rather expressed as ΔNp63 and ΔNp73 isoforms in a wide range of malignancies, where they act as p53 antagonists. Accumulation of ΔNp63 and ΔNp73, which is associated with chemoresistance, can result from infection by oncogenic viruses such as EBV or HPV. Our study focuses on the highly carcinogenic ΔNp73α isoform and uses a viral model of cellular transformation. We unveil a physical interaction between ΔNp73α and the E2F4/p130 complex involved in cell cycle control, which rewires the E2F4/p130 transcriptional program. Our work shows that ΔNp73 isoforms can establish interactions with proteins that do not bind to the TAp73α tumor suppressor. This situation is analogous to the gain-of-function interactions of p53 mutants supporting cellular proliferation.


Subject(s)
Epstein-Barr Virus Infections , Papillomavirus Infections , Humans , Cell Transformation, Neoplastic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F4 Transcription Factor/genetics , E2F4 Transcription Factor/metabolism , Gene Expression , Herpesvirus 4, Human/genetics , Human Papillomavirus Viruses , Keratinocytes , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Crk-Associated Substrate Protein/metabolism , Neoplasms/metabolism
3.
mSphere ; 5(4)2020 07 15.
Article in English | MEDLINE | ID: mdl-32669468

ABSTRACT

The beta human papillomaviruses (HPVs) are subdivided into 5 species (beta-1 to beta-5), and they were first identified in the skin. However, the beta-3 species appears to be more highly represented in the mucosal epithelia than in the skin. Functional studies have also highlighted that beta-3 HPV49 shares some functional similarities with mucosal high-risk (HR) HPV16. Here, we describe the characterization of the in vitro transforming properties of the entire beta-3 species, which includes three additional HPV types: HPV75, HPV76, and HPV115. HPV49, HPV75, and HPV76 E6 and E7 (E6/E7), but not HPV115 E6 and E7, efficiently inactivate the p53 and pRb pathways and immortalize or extend the life span of human foreskin keratinocytes (HFKs). As observed for HR HPV16, cell cycle deregulation mediated by beta-3 HPV E6/E7 expression leads to p16INK4a accumulation, whereas no p16INK4a was detected in beta-2 HPV38 E6/E7 HFKs. As shown for HPV49 E6, HPV75 and HPV76 E6s degrade p53 by an E6AP/proteasome-mediated mechanism. Comparative analysis of cellular gene expression patterns of HFKs containing E6 and E7 from HR HPV16, beta-3 HPV types, and beta-2 HPV38 further highlights the functional similarities of HR HPV16 and beta-3 HPV49, HPV75, and HPV76. The expression profiles of these four HPV HFKs show some similarities and diverge substantially from those of beta-3 HPV115 E6/E7 and beta-2 HPV38 E6/E7 HFKs. In summary, our data show that beta-3 HPV types share some mechanisms with HR HPV types and pave the way for additional studies aiming to evaluate their potential role in human pathologies.IMPORTANCE Human papillomaviruses are currently classified in different genera. Mucosal HPVs belonging to the alpha genus have been clearly associated with carcinogenesis of the mucosal epithelium at different sites. Beta HPV types have been classified as cutaneous. Although findings indicate that some beta HPVs from species 1 and 2 play a role, together with UV irradiation, in skin cancer, very little is known about the transforming properties of most of the beta HPVs. This report shows the transforming activity of E6 and E7 from beta-3 HPV types. Moreover, it highlights that beta-3 HPVs share some biological properties more extensively with mucosal high-risk HPV16 than with beta-2 HPV38. This report provides new paradigms for a better understanding of the biology of the different HPV types and their possible association with lesions at mucosal and/or cutaneous epithelia.


Subject(s)
Alphapapillomavirus/genetics , Alphapapillomavirus/pathogenicity , Epithelial Cells/virology , Mucous Membrane/virology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Alphapapillomavirus/classification , Animals , Cells, Cultured , Human papillomavirus 16/genetics , Humans , Keratinocytes/virology , Male , Mice , Mucous Membrane/cytology , NIH 3T3 Cells , Skin/virology
4.
PLoS Pathog ; 14(8): e1007158, 2018 08.
Article in English | MEDLINE | ID: mdl-30089163

ABSTRACT

Human papillomavirus type 16 (HPV16) and other oncoviruses have been shown to block innate immune responses and to persist in the host. However, to avoid viral persistence, the immune response attempts to clear the infection. IL-1ß is a powerful cytokine produced when viral motifs are sensed by innate receptors that are members of the inflammasome family. Whether oncoviruses such as HPV16 can activate the inflammasome pathway remains unknown. Here, we show that infection of human keratinocytes with HPV16 induced the secretion of IL-1ß. Yet, upon expression of the viral early genes, IL-1ß transcription was blocked. We went on to show that expression of the viral oncoprotein E6 in human keratinocytes inhibited IRF6 transcription which we revealed regulated IL-1ß promoter activity. Preventing E6 expression using siRNA, or using E6 mutants that prevented degradation of p53, showed that p53 regulated IRF6 transcription. HPV16 abrogation of p53 binding to the IRF6 promoter was shown by ChIP in tissues from patients with cervical cancer. Thus E6 inhibition of IRF6 is an escape strategy used by HPV16 to block the production IL-1ß. Our findings reveal a struggle between oncoviral persistence and host immunity; which is centered on IL-1ß regulation.


Subject(s)
Gene Expression Regulation/immunology , Immune Evasion/immunology , Interferon Regulatory Factors/metabolism , Interleukin-1beta/biosynthesis , Papillomavirus Infections/immunology , Human papillomavirus 16/immunology , Humans , Interferon Regulatory Factors/immunology , Interleukin-1beta/immunology , Keratinocytes/immunology , Keratinocytes/metabolism , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/metabolism , Repressor Proteins/metabolism
5.
FEBS J ; 284(19): 3171-3201, 2017 10.
Article in English | MEDLINE | ID: mdl-28786561

ABSTRACT

Protein ubiquitination and its reverse reaction, deubiquitination, regulate protein stability, protein binding activity, and their subcellular localization. These reactions are catalyzed by the enzymes E1, E2, and E3 ubiquitin (Ub) ligases and deubiquitinases (DUBs). The Ub-proteasome system (UPS) is targeted by viruses for the sake of their replication and to escape host immune response. To identify novel partners of human papillomavirus 16 (HPV16) E6 and E7 proteins, we assembled and screened a library of 590 cDNAs related to the UPS by using the Gaussia princeps luciferase protein complementation assay. HPV16 E6 was found to bind to the homology to E6AP C terminus-type Ub ligase (E6AP), three really interesting new gene (RING)-type Ub ligases (MGRN1, LNX3, LNX4), and the DUB Ub-specific protease 15 (USP15). Except for E6AP, the binding of UPS factors did not require the LxxLL-binding pocket of HPV16 E6. LNX3 bound preferentially to all high-risk mucosal HPV E6 tested, whereas LNX4 bound specifically to HPV16 E6. HPV16 E7 was found to bind to several broad-complex tramtrack and bric-a-brac domain-containing proteins (such as TNFAIP1/KCTD13) that are potential substrate adaptors of Cullin 3-RING Ub ligases, to RING-type Ub ligases implicated in innate immunity (RNF135, TRIM32, TRAF2, TRAF5), to the substrate adaptor DCAF15 of Cullin 4-RING Ub ligase and to some DUBs (USP29, USP33). The binding to UPS factors did not require the LxCxE motif but rather the C-terminal region of HPV16 E7 protein. The identified UPS factors interacted with most of E7 proteins across different HPV types. This study establishes a strategy for the rapid identification of interactions between host or pathogen proteins and the human ubiquitination system.


Subject(s)
Host-Pathogen Interactions , Human papillomavirus 16/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin/genetics , Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Computational Biology , Gene Expression Regulation , Genes, Reporter , Human papillomavirus 16/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Annotation , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Peptide Library , Protein Binding , Protein Interaction Mapping , Proteins/genetics , Proteins/metabolism , Repressor Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Virus Replication
6.
Proc Natl Acad Sci U S A ; 113(46): E7169-E7175, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799531

ABSTRACT

An essential early step in the assembly of human spliceosomes onto pre-mRNA involves the recognition of regulatory RNA cis elements in the 3' splice site by the U2 auxiliary factor (U2AF). The large (U2AF65) and small (U2AF35) subunits of the U2AF heterodimer contact the polypyrimidine tract (Py-tract) and the AG-dinucleotide, respectively. The tandem RNA recognition motif domains (RRM1,2) of U2AF65 adopt closed/inactive and open/active conformations in the free form and when bound to bona fide Py-tract RNA ligands. To investigate the molecular mechanism and dynamics of 3' splice site recognition by U2AF65 and the role of U2AF35 in the U2AF heterodimer, we have combined single-pair FRET and NMR experiments. In the absence of RNA, the RRM1,2 domain arrangement is highly dynamic on a submillisecond time scale, switching between closed and open conformations. The addition of Py-tract RNA ligands with increasing binding affinity (strength) gradually shifts the equilibrium toward an open conformation. Notably, the protein-RNA complex is rigid in the presence of a strong Py-tract but exhibits internal motion with weak Py-tracts. Surprisingly, the presence of U2AF35, whose UHM domain interacts with U2AF65 RRM1, increases the population of the open arrangement of U2AF65 RRM1,2 in the absence and presence of a weak Py-tract. These data indicate that the U2AF heterodimer promotes spliceosome assembly by a dynamic population shift toward the open conformation of U2AF65 to facilitate the recognition of weak Py-tracts at the 3' splice site. The structure and RNA binding of the heterodimer was unaffected by cancer-linked myelodysplastic syndrome mutants.


Subject(s)
RNA Splice Sites , RNA/metabolism , Splicing Factor U2AF/metabolism , Dimerization , Humans , Mutation , Protein Binding , Protein Conformation , Protein Multimerization , RNA/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spliceosomes/metabolism , Splicing Factor U2AF/chemistry , Splicing Factor U2AF/genetics
8.
Nature ; 529(7587): 541-5, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26789255

ABSTRACT

The p53 pro-apoptotic tumour suppressor is mutated or functionally altered in most cancers. In epithelial tumours induced by 'high-risk' mucosal human papilloma viruses, including human cervical carcinoma and a growing number of head-and-neck cancers, p53 is degraded by the viral oncoprotein E6 (ref. 2). In this process, E6 binds to a short leucine (L)-rich LxxLL consensus sequence within the cellular ubiquitin ligase E6AP. Subsequently, the E6/E6AP heterodimer recruits and degrades p53 (ref. 4). Neither E6 nor E6AP are separately able to recruit p53 (refs 3, 5), and the precise mode of assembly of E6, E6AP and p53 is unknown. Here we solve the crystal structure of a ternary complex comprising full-length human papilloma virus type 16 (HPV-16) E6, the LxxLL motif of E6AP and the core domain of p53. The LxxLL motif of E6AP renders the conformation of E6 competent for interaction with p53 by structuring a p53-binding cleft on E6. Mutagenesis of critical positions at the E6-p53 interface disrupts p53 degradation. The E6-binding site of p53 is distal from previously described DNA- and protein-binding surfaces of the core domain. This suggests that, in principle, E6 may avoid competition with cellular factors by targeting both free and bound p53 molecules. The E6/E6AP/p53 complex represents a prototype of viral hijacking of both the ubiquitin-mediated protein degradation pathway and the p53 tumour suppressor pathway. The present structure provides a framework for the design of inhibitory therapeutic strategies against oncogenesis mediated by human papilloma virus.


Subject(s)
Human papillomavirus 16/metabolism , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/metabolism , Proteolysis , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/chemistry , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Human papillomavirus 16/chemistry , Human papillomavirus 16/pathogenicity , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oncogene Proteins, Viral/genetics , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics
9.
PLoS One ; 10(7): e0132339, 2015.
Article in English | MEDLINE | ID: mdl-26151636

ABSTRACT

Oncogenic types of human papillomaviruses (HPVs) cause cervical cancer and other malignancies in humans. The HPV E6 oncoprotein is considered to be an attractive therapeutic target since its inhibition can lead to the apoptotic cell death of HPV-positive cancer cells. The HPV type 16 (HPV16) E6-binding peptide pep11, and variants thereof, induce cell death specifically in HPV16-positive cancer cells. Although they do not encompass the LxxLL binding motif found in cellular HPV16 E6 interaction partners, such as E6AP, the pep11 variants strongly bind to HPV16 E6 by contacting the recently identified E6AP binding pocket. Thus, these peptides can serve as prototype E6-inhibitory molecules which target the E6AP pocket. We here analyzed their intracellular interaction with HPV16 E6. By comprehensive intracellular binding studies and GST pull-down assays, we show that E6-binding competent pep11 variants induce the formation of a trimeric complex, consisting of pep11, HPV16 E6 and p53. These findings indicate that peptides, which do not contain the LxxLL motif, can reshape E6 to enable its interaction with p53. The formation of the trimeric HPV16 E6 / peptide / p53 complex was associated with an increase of endogenous HPV16 E6 protein amounts. Yet, total cellular p53 amounts were also increased, indicating that the E6 / E6AP-mediated degradation of p53 is blocked. These findings suggest that inhibition of oncogenic activities by targeting the E6AP pocket on HPV16 E6 could be a strategy for therapeutic intervention.


Subject(s)
Cytoplasm/metabolism , Oncogene Proteins, Viral/metabolism , Peptides/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Binding Sites/genetics , Cell Line, Tumor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Molecular Sequence Data , Mutation , Oncogene Proteins, Viral/genetics , Peptides/genetics , Protein Binding , Repressor Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/metabolism
10.
PLoS One ; 9(11): e112514, 2014.
Article in English | MEDLINE | ID: mdl-25383876

ABSTRACT

The HPV E6 oncoprotein maintains the malignant phenotype of HPV-positive cancer cells and represents an attractive therapeutic target. E6 forms a complex with the cellular E6AP ubiquitin ligase, ultimately leading to p53 degradation. The recently elucidated x-ray structure of a HPV16 E6/E6AP complex showed that HPV16 E6 forms a distinct binding pocket for E6AP. This discovery raises the question whether the E6AP binding pocket is druggable, i. e. whether it provides a docking site for functional E6 inhibitors. To address these issues, we performed a detailed analysis of the HPV16 E6 interactions with two small peptides: (i) E6APpep, corresponding to the E6 binding domain of E6AP, and (ii) pep11**, a peptide that binds to HPV16 E6 and, in contrast to E6APpep, induces apoptosis, specifically in HPV16-positive cancer cells. Surface plasmon resonance, NMR chemical shift perturbation, and mammalian two-hybrid analyses coupled to mutagenesis indicate that E6APpep contacts HPV16 E6 amino acid residues within the E6AP pocket, both in vitro and intracellularly. Many of these amino acids were also important for binding to pep11**, suggesting that the binding sites for the two peptides on HPV16 E6 overlap. Yet, few E6 amino acids were differentially involved which may contribute to the higher binding affinity of pep11**. Data from the HPV16 E6/pep11** interaction allowed the rational design of single amino acid exchanges in HPV18 and HPV31 E6 that enabled their binding to pep11**. Taken together, these results suggest that E6 molecular surfaces mediating E6APpep binding can also accommodate pro-apoptotic peptides that belong to different sequence families. As proof of concept, this study provides the first experimental evidence that the E6AP binding pocket is druggable, opening new possibilities for rational, structure-based drug design.


Subject(s)
Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/genetics , Peptides/chemistry , Repressor Proteins/chemistry , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , HeLa Cells , Humans , Models, Molecular , Molecular Docking Simulation , Oncogene Proteins, Viral/antagonists & inhibitors , Oncogene Proteins, Viral/metabolism , Peptides/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Structure-Activity Relationship
11.
Science ; 339(6120): 694-8, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23393263

ABSTRACT

E6 viral oncoproteins are key players in epithelial tumors induced by papillomaviruses in vertebrates, including cervical cancer in humans. E6 proteins target many host proteins by specifically interacting with acidic LxxLL motifs. We solved the crystal structures of bovine (BPV1) and human (HPV16) papillomavirus E6 proteins bound to LxxLL peptides from the focal adhesion protein paxillin and the ubiquitin ligase E6AP, respectively. In both E6 proteins, two zinc domains and a linker helix form a basic-hydrophobic pocket, which captures helical LxxLL motifs in a way compatible with other interaction modes. Mutational inactivation of the LxxLL binding pocket disrupts the oncogenic activities of both E6 proteins. This work reveals the structural basis of both the multifunctionality and the oncogenicity of E6 proteins.


Subject(s)
Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/metabolism , Paxillin/chemistry , Protein Interaction Domains and Motifs , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Amino Acid Motifs , Amino Acid Sequence , Bovine papillomavirus 1 , Crystallography, X-Ray , Human papillomavirus 16 , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Oncogene Proteins, Viral/genetics , Paxillin/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Point Mutation , Protein Structure, Secondary , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
12.
Structure ; 20(4): 604-17, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22483108

ABSTRACT

The viral oncoprotein E6 is an essential factor for cervical cancers induced by "high-risk" mucosal HPV. Among other oncogenic activities, E6 recruits the ubiquitin ligase E6AP to promote the ubiquitination and subsequent proteasomal degradation of p53. E6 is prone to self-association, which long precluded its structural analysis. Here we found that E6 specifically dimerizes through its N-terminal domain and that disruption of the dimer interface strongly increases E6 solubility. This allowed us to raise structural data covering the entire HPV16 E6 protein, including the high-resolution NMR structures of the two zinc-binding domains of E6 and a robust data-driven model structure of the N-terminal domain homodimer. Interestingly, homodimer interface mutations that disrupt E6 self-association also inactivate E6-mediated p53 degradation. These data suggest that E6 needs to self-associate via its N-terminal domain to promote the polyubiquitination of p53 by E6AP.


Subject(s)
Oncogene Proteins, Viral/chemistry , Repressor Proteins/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Binding Sites , Escherichia coli , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
13.
Protein Expr Purif ; 80(1): 8-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21777678

ABSTRACT

E6 is a small oncoprotein involved in tumorigenesis induced by papillomaviruses (PVs). E6 often recognizes its cellular targets by binding to short motifs presenting the consensus LXXLL. E6 proteins have long resisted structural analysis. We found that bovine papillomavirus type 1 (BPV1) E6 binds the N-terminal LXXLL motif of the cellular protein paxillin with significantly higher affinity as compared to other E6/peptide interactions. Although recombinant BPV1 E6 was poorly soluble in the free state, provision of the paxillin LXXLL peptide during BPV1 E6 biosynthesis greatly enhanced the protein's solubility. Expression of BPV1 E6/LXXLL peptide complexes was carried out in bacteria in the form of triple fusion constructs comprising, from N- to C-terminus, the soluble carrier protein maltose binding protein (MBP), the LXXLL motif and the E6 protein. A TEV protease cleavage site was placed either between MBP and LXXLL motif or between LXXLL motif and E6. These constructs allowed us to produce highly concentrated samples of BPV1 E6, either covalently fused to the C-terminus of the LXXLL motif (intra-molecular complex) or non-covalently bound to it (inter-molecular complex). Heteronuclear NMR measurements were performed and showed that the E6 protein was folded with similar conformations in both covalent and non-covalent complexes. These data open the way to novel structural and functional studies of the BPV1 E6 in complex with its preferential target motif.


Subject(s)
Bovine papillomavirus 1/genetics , Escherichia coli/genetics , Oncogene Proteins, Viral/genetics , Paxillin/genetics , Peptides/genetics , Recombinant Fusion Proteins/genetics , Amino Acid Sequence , Bovine papillomavirus 1/metabolism , Gene Expression , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/isolation & purification , Molecular Sequence Data , Oncogene Proteins, Viral/isolation & purification , Oncogene Proteins, Viral/metabolism , Paxillin/isolation & purification , Paxillin/metabolism , Peptides/isolation & purification , Peptides/metabolism , Protein Binding , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Solubility
14.
Nature ; 475(7356): 408-11, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21753750

ABSTRACT

Many cellular functions involve multi-domain proteins, which are composed of structurally independent modules connected by flexible linkers. Although it is often well understood how a given domain recognizes a cognate oligonucleotide or peptide motif, the dynamic interaction of multiple domains in the recognition of these ligands remains to be characterized. Here we have studied the molecular mechanisms of the recognition of the 3'-splice-site-associated polypyrimidine tract RNA by the large subunit of the human U2 snRNP auxiliary factor (U2AF65) as a key early step in pre-mRNA splicing. We show that the tandem RNA recognition motif domains of U2AF65 adopt two remarkably distinct domain arrangements in the absence or presence of a strong (that is, high affinity) polypyrimidine tract. Recognition of sequence variations in the polypyrimidine tract RNA involves a population shift between these closed and open conformations. The equilibrium between the two conformations functions as a molecular rheostat that quantitatively correlates the natural variations in polypyrimidine tract nucleotide composition, length and functional strength to the efficiency to recruit U2 snRNP to the intron during spliceosome assembly. Mutations that shift the conformational equilibrium without directly affecting RNA binding modulate splicing activity accordingly. Similar mechanisms of cooperative multi-domain conformational selection may operate more generally in the recognition of degenerate nucleotide or amino acid motifs by multi-domain proteins.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/physiology , RNA, Messenger/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Amino Acid Motifs , Base Sequence , Humans , Introns/genetics , Ligands , Models, Molecular , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Pyrimidines/metabolism , RNA Splice Sites/genetics , RNA, Messenger/genetics , Spliceosomes/chemistry , Spliceosomes/metabolism , Splicing Factor U2AF , Substrate Specificity
15.
J Mol Biol ; 396(1): 90-104, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-19917295

ABSTRACT

Papillomavirus E6 oncoproteins bind and often provoke the degradation of many cellular proteins important for the control of cell proliferation and/or cell death. Structural studies on E6 proteins have long been hindered by the difficulties of obtaining highly concentrated samples of recombinant E6. Here, we show that recombinant E6 proteins from eight human papillomavirus strains and one bovine papillomavirus strain exist as oligomeric and multimeric species. These species were characterized using a variety of biochemical and biophysical techniques, including analytical gel filtration, activity assays, surface plasmon resonance, electron microscopy and Fourier transform infrared spectroscopy. The characterization of E6 oligomers is facilitated by the fusion to the maltose binding protein, which slows the formation of higher-order multimeric species. The proportion of each oligomeric form varies depending on the viral strain considered. Oligomers appear to consist of folded units, which, in the case of high-risk mucosal human papillomavirus E6, retain binding to the ubiquitin ligase E6-associated protein and the capacity to degrade the proapoptotic protein p53. In addition to the small-size oligomers, E6 proteins spontaneously assemble into large organized multimeric structures, a process that is accompanied by a significant increase in the beta-sheet secondary structure content. Finally, co-localisation experiments using E6 equipped with different tags further demonstrate the occurrence of E6 self-association in eukaryotic cells. The ensemble of these data suggests that self-association is a general property of E6 proteins that occurs both in vitro and in vivo and might therefore be functionally relevant.


Subject(s)
Viral Proteins/metabolism , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Chromatography, Gel , Humans , Maltose-Binding Proteins , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Stability , Protein Structure, Quaternary , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Solubility , Spectroscopy, Fourier Transform Infrared , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Zinc/chemistry
16.
J Virol ; 83(17): 8759-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19553340

ABSTRACT

Papillomaviruses (PVs) are a large family of small DNA viruses infecting mammals, reptiles, and birds. PV infection induces cell proliferation that may lead to the formation of orogenital or skin tumors. PV-induced cell proliferation has been related mainly to the expression of two small oncoproteins, E6 and E7. In mammalian PVs, E6 contains two 70-residue zinc-binding repeats, whereas E7 consists of a natively unfolded N-terminal region followed by a zinc-binding domain which folds as an obligate homodimer. Here, we show that both the novel francolin bird PV Francolinus leucoscepus PV type 1 (FlPV-1) and the chaffinch bird PV Fringilla coelebs PV contain unusual E6 and E7 proteins. The avian E7 proteins contain an extended unfolded N terminus and a zinc-binding domain of reduced size, whereas the avian E6 proteins consist of a single zinc-binding domain. A comparable single-domain E6 protein may have existed in a common ancestor of mammalian and avian PVs. Mammalian E6 C-terminal domains are phylogenetically related to those of single-domain avian E6, whereas mammalian E6 N-terminal domains seem to have emerged by duplication and subsequently diverged from the original ancestral domain. In avian and mammalian cells, both FlPV-1 E6 and FlPV-1 E7 were evenly expressed in the cytoplasm and the nucleus. Finally, samples of full-length FlPV-1 E6 and the FlPV-1 E7 C-terminal zinc-binding domain were prepared for biophysical analysis. Both constructs were highly soluble and well folded, according to nuclear magnetic resonance spectroscopy measurements.


Subject(s)
Bird Diseases/virology , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus Infections/veterinary , Amino Acid Sequence , Animals , Birds , Cell Nucleus/chemistry , Cluster Analysis , Cytoplasm/chemistry , DNA, Viral/chemistry , DNA, Viral/genetics , Evolution, Molecular , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Oncogene Proteins, Viral/analysis , Papillomaviridae/isolation & purification , Phylogeny , Protein Folding , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology
17.
Nucleic Acids Res ; 36(22): 7068-77, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18987004

ABSTRACT

All RNA sequences that fold into hairpins possess the intrinsic potential to form intermolecular duplexes because of their high self-complementarity. The thermodynamically more stable duplex conformation is favored under high salt conditions and at high RNA concentrations, posing a challenging problem for structural studies of small RNA hairpin conformations. We developed and applied a novel approach to unambiguously distinguish RNA hairpin and duplex conformations for the structural analysis of a Xist RNA A-repeat. Using a combination of a quantitative HNN-COSY experiment and an optimized double isotope-filtered NOESY experiment we could define the conformation of the 26-mer A-repeat RNA. In contrast to a previous secondary structure prediction of a double hairpin structure, the NMR data show that only the first predicted hairpin is formed, while the second predicted hairpin mediates dimerization of the A-repeat by duplex formation with a second A-repeat. The strategy employed here will be generally applicable to identify and quantify populations of hairpin and duplex conformations and to define RNA folding topology from inter- and intra-molecular base-pairing patterns.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , RNA, Double-Stranded/chemistry , RNA, Untranslated/chemistry , Base Sequence , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Long Noncoding , Repetitive Sequences, Nucleic Acid
18.
Protein Expr Purif ; 51(1): 59-70, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17055740

ABSTRACT

Protein aggregation is a main barrier hindering structural and functional studies of a number of interesting biological targets. The E6 oncoprotein of Human Papillomavirus strain 16 (E6(16)) is difficult to express under a native soluble form in bacteria. Produced as an unfused sequence, it forms inclusion bodies. Fused to the C-terminus of MBP, it is mainly produced in the form of soluble high molecular weight aggregates. Here, we produced as MBP-fusions seven E6 proteins from other HPV strains (5, 11, 18, 33, 45, 52, and 58) belonging to four different species, and we compared their aggregation state to that of MBP-E6(16). Using a fast mutagenesis method, we changed most non-conserved cysteines to the isosteric residue serine to minimize disulfide bridge-mediated aggregation during purification. Static and dynamic light scattering measurements, ultracentrifugation and electron microscopy demonstrated the presence in all MBP-E6 preparations of soluble high-molecular weight aggregates with a well-defined spherical shape. These aggregated particles are relatively monodisperse but their amount and their size vary depending on the conditions of expression and the strain considered. For all strains, minimal aggregate formation occurs when the expression is performed at 15 degrees C. Such observations suggest that the assembly of MBP-E6 aggregates takes place in vivo during protein biosynthesis, rather than occurring during purification. Finally, we show that all MBP-E6 preparations contain two zinc ions per protein monomer, suggesting that E6 domains within the high molecular weight aggregates possess a native-like fold, which enables correct coordination to the metal center.


Subject(s)
Alphapapillomavirus/chemistry , Carrier Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Recombinant Fusion Proteins/chemistry , Repressor Proteins/metabolism , Amino Acid Sequence , DNA-Binding Proteins/metabolism , Disulfides/analysis , Humans , Light , Maltose-Binding Proteins , Microscopy, Electron , Molecular Sequence Data , Mutagenesis , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/ultrastructure , Protein Engineering/methods , Protein Structure, Quaternary , Recombinant Fusion Proteins/ultrastructure , Repressor Proteins/chemistry , Repressor Proteins/ultrastructure , Scattering, Radiation , Sequence Alignment , Temperature , Ultracentrifugation , Zinc/analysis
19.
Protein Expr Purif ; 50(1): 89-101, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16884919

ABSTRACT

The popular pulldown chromatographic assay detects complexes mediated by fusion proteins retained on affinity resin. The main limitation of this method is that it does not analyze complexes at equilibrium but after several washing steps. Consequently, fast-dissociating complexes may remain undetected. Here, we present the holdup assay, based on the principle of comparative chromatographic retention which eliminates the use of washing steps. The assay evaluates fractions of free and bound species at equilibrium. We used human papillomavirus oncoprotein E6, an E6-binding peptide and an E6-binding PDZ domain, to test several protocols utilizing pure proteins or expression extracts. The holdup assay is faster and more informative than the pulldown assay. It detects fast-dissociating complexes and it is also suited for evaluating equilibrium constants. It is potentially adaptable for automated determination of affinity constants and high-throughput analysis of interactions between proteins and other proteins, peptides, nucleic acids, or small regulatory molecules.


Subject(s)
Chromatography, Affinity/methods , Glutathione Transferase/chemistry , Oncogene Proteins, Viral/chemistry , Repressor Proteins/chemistry , Cell Line, Tumor , Humans , Ligands , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/isolation & purification , Protein Binding , Repressor Proteins/biosynthesis , Repressor Proteins/isolation & purification , Resins, Synthetic/chemistry , Sensitivity and Specificity
20.
Science ; 312(5782): 1961-5, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16809543

ABSTRACT

Discrimination between splice sites and similar, nonsplice sequences is essential for correct intron removal and messenger RNA formation in eukaryotes. The 65- and 35-kD subunits of the splicing factor U2AF, U2AF65 and U2AF35, recognize, respectively, the pyrimidine-rich tract and the conserved terminal AG present at metazoan 3' splice sites. We report that DEK, a chromatin- and RNA-associated protein mutated or overexpressed in certain cancers, enforces 3' splice site discrimination by U2AF. DEK phosphorylated at serines 19 and 32 associates with U2AF35, facilitates the U2AF35-AG interaction and prevents binding of U2AF65 to pyrimidine tracts not followed by AG. DEK and its phosphorylation are required for intron removal, but not for splicing complex assembly, which indicates that proofreading of early 3' splice site recognition influences catalytic activation of the spliceosome.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Introns , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , RNA Precursors/metabolism , RNA Splicing , Ribonucleoproteins/metabolism , Adenosine Triphosphate/metabolism , Chromosomal Proteins, Non-Histone/genetics , Dimerization , Dinucleoside Phosphates/metabolism , HeLa Cells , Humans , Mutation , Oncogene Proteins/genetics , Phosphorylation , Poly-ADP-Ribose Binding Proteins , Pyrimidines/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear , Spliceosomes/metabolism , Splicing Factor U2AF
SELECTION OF CITATIONS
SEARCH DETAIL
...