Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 49(21): 12119-12135, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34747482

ABSTRACT

Telomere shortening can cause detrimental diseases and contribute to aging. It occurs due to the end replication problem in cells lacking telomerase. Furthermore, recent studies revealed that telomere shortening can be attributed to difficulties of the semi-conservative DNA replication machinery to replicate the bulk of telomeric DNA repeats. To investigate telomere replication in a comprehensive manner, we develop QTIP-iPOND - Quantitative Telomeric chromatin Isolation Protocol followed by isolation of Proteins On Nascent DNA - which enables purification of proteins that associate with telomeres specifically during replication. In addition to the core replisome, we identify a large number of proteins that specifically associate with telomere replication forks. Depletion of several of these proteins induces telomere fragility validating their importance for telomere replication. We also find that at telomere replication forks the single strand telomere binding protein POT1 is depleted, whereas histone H1 is enriched. Our work reveals the dynamic changes of the telomeric proteome during replication, providing a valuable resource of telomere replication proteins. To our knowledge, this is the first study that examines the replisome at a specific region of the genome.


Subject(s)
DNA Replication , Telomerase/metabolism , Telomere/metabolism , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Shelterin Complex/metabolism , Telomere Shortening , Telomere-Binding Proteins/metabolism
2.
Nature ; 587(7833): 303-308, 2020 11.
Article in English | MEDLINE | ID: mdl-33057192

ABSTRACT

Telomeres-repeated, noncoding nucleotide motifs and associated proteins that are found at the ends of eukaryotic chromosomes-mediate genome stability and determine cellular lifespan1. Telomeric-repeat-containing RNA (TERRA) is a class of long noncoding RNAs (lncRNAs) that are transcribed from chromosome ends2,3; these RNAs in turn regulate telomeric chromatin structure and telomere maintenance through the telomere-extending enzyme telomerase4-6 and homology-directed DNA repair7,8. The mechanisms by which TERRA is recruited to chromosome ends remain poorly defined. Here we develop a reporter system with which to dissect the underlying mechanisms, and show that the UUAGGG repeats of TERRA are both necessary and sufficient to target TERRA to chromosome ends. TERRA preferentially associates with short telomeres through the formation of telomeric DNA-RNA hybrid (R-loop) structures that can form in trans. Telomere association and R-loop formation trigger telomere fragility and are promoted by the recombinase RAD51 and its interacting partner BRCA2, but counteracted by the RNA-surveillance factors RNaseH1 and TRF1. RAD51 physically interacts with TERRA and catalyses R-loop formation with TERRA in vitro, suggesting a direct involvement of this DNA recombinase in the recruitment of TERRA by strand invasion. Together, our findings reveal a RAD51-dependent pathway that governs TERRA-mediated R-loop formation after transcription, providing a mechanism for the recruitment of lncRNAs to new loci in trans.


Subject(s)
R-Loop Structures , RNA, Long Noncoding/chemistry , Rad51 Recombinase/metabolism , Telomere/chemistry , Telomere/metabolism , Base Sequence , Biocatalysis , Genes, Reporter , HeLa Cells , Humans , RNA, Long Noncoding/genetics , Ribonuclease H/metabolism , Telomere/genetics , Telomeric Repeat Binding Protein 1/metabolism
3.
Nat Commun ; 10(1): 3884, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31467287

ABSTRACT

Genomic instability is a hallmark of cancer. Whether it also occurs in Cancer Associated Fibroblasts (CAFs) remains to be carefully investigated. Loss of CSL/RBP-Jκ, the effector of canonical NOTCH signaling with intrinsic transcription repressive function, causes conversion of dermal fibroblasts into CAFs. Here, we find that CSL down-modulation triggers DNA damage, telomere loss and chromosome end fusions that also occur in skin Squamous Cell Carcinoma (SCC)-associated CAFs, in which CSL is decreased. Separately from its role in transcription, we show that CSL is part of a multiprotein telomere protective complex, binding directly and with high affinity to telomeric DNA as well as to UPF1 and Ku70/Ku80 proteins and being required for their telomere association. Taken together, the findings point to a central role of CSL in telomere homeostasis with important implications for genomic instability of cancer stromal cells and beyond.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/metabolism , Fibroblasts/metabolism , Genomic Instability , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Telomere/metabolism , Carcinoma, Squamous Cell/genetics , DNA Damage , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Homeostasis , Humans , Ku Autoantigen/metabolism , Membrane Proteins , Molecular Docking Simulation , Mutagenesis , RNA Helicases/metabolism , Receptors, Notch/metabolism , Signal Transduction , Skin/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Trans-Activators/metabolism
4.
Nature ; 516(7531): 361-6, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25409151

ABSTRACT

Influenza virus polymerase uses a capped primer, derived by 'cap-snatching' from host pre-messenger RNA, to transcribe its RNA genome into mRNA and a stuttering mechanism to generate the poly(A) tail. By contrast, genome replication is unprimed and generates exact full-length copies of the template. Here we use crystal structures of bat influenza A and human influenza B polymerases (FluA and FluB), bound to the viral RNA promoter, to give mechanistic insight into these distinct processes. In the FluA structure, a loop analogous to the priming loop of flavivirus polymerases suggests that influenza could initiate unprimed template replication by a similar mechanism. Comparing the FluA and FluB structures suggests that cap-snatching involves in situ rotation of the PB2 cap-binding domain to direct the capped primer first towards the endonuclease and then into the polymerase active site. The polymerase probably undergoes considerable conformational changes to convert the observed pre-initiation state into the active initiation and elongation states.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Influenza A virus/enzymology , Influenza B virus/enzymology , Models, Molecular , RNA Caps , RNA, Viral/biosynthesis , RNA, Viral/chemistry , Catalytic Domain , Crystallization , DNA-Directed RNA Polymerases/chemistry , Gene Expression Regulation, Viral , Influenza A virus/chemistry , Influenza B virus/chemistry , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , RNA Caps/chemistry , RNA Caps/metabolism , Virus Replication
5.
PLoS One ; 9(1): e84973, 2014.
Article in English | MEDLINE | ID: mdl-24454773

ABSTRACT

Orthomyxovirus Influenza A virus (IAV) heterotrimeric polymerase performs transcription of viral mRNAs by cap-snatching, which involves generation of capped primers by host pre-mRNA binding via the PB2 subunit cap-binding site and cleavage 10-13 nucleotides from the 5' cap by the PA subunit endonuclease. Thogotoviruses, tick-borne orthomyxoviruses that includes Thogoto (THOV), Dhori (DHOV) and Jos (JOSV) viruses, are thought to perform cap-snatching by cleaving directly after the cap and thus have no heterogeneous, host-derived sequences at the 5' extremity of their mRNAs. Based on recent work identifying the cap-binding and endonuclease domains in IAV polymerase, we determined the crystal structures of two THOV PB2 domains, the putative cap-binding and the so-called '627-domain', and the structures of the putative endonuclease domains (PA-Nter) of THOV and DHOV. Despite low sequence similarity, corresponding domains have the same fold confirming the overall architectural similarity of orthomyxovirus polymerases. However the putative Thogotovirus cap-snatching domains in PA and PB2 have non-conservative substitutions of key active site residues. Biochemical analysis confirms that, unlike the IAV domains, the THOV and DHOV PA-Nter domains do not bind divalent cations and have no endonuclease activity and the THOV central PB2 domain does not bind cap analogues. On the other hand, sequence analysis suggests that other, non-influenza, orthomyxoviruses, such as salmon anemia virus (isavirus) and Quaranfil virus likely conserve active cap-snatching domains correlating with the reported occurrence of heterogeneous, host-derived sequences at the 5' end of the mRNAs of these viruses. These results highlight the unusual nature of transcription initiation by Thogotoviruses.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Orthomyxoviridae/genetics , RNA Caps , Amino Acid Sequence , DNA-Directed RNA Polymerases/chemistry , Molecular Sequence Data , Orthomyxoviridae/enzymology , Protein Conformation , Sequence Homology, Amino Acid
6.
J Med Chem ; 56(21): 8915-30, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24134208

ABSTRACT

The heterotrimeric influenza virus polymerase performs replication and transcription of viral RNA in the nucleus of infected cells. Transcription by "cap-snatching" requires that host-cell pre-mRNAs are bound via their 5' cap to the PB2 subunit. Thus, the PB2 cap-binding site is potentially a good target for new antiviral drugs that will directly inhibit viral replication. Docking studies using the structure of the PB2 cap-binding domain suggested that 7-alkylguanine derivatives substituted at position N-9 and N-2 could be good candidates. Four series of 7,9-di- and 2,7,9-trialkyl guanine derivatives were synthesized and evaluated by an AlphaScreen assay in competition with a biotinylated cap analogue. Three synthesized compounds display potent in vitro activity with IC50 values lower than 10 µM. High-resolution X-ray structures of three inhibitors in complex with the H5N1 PB2 cap-binding domain confirmed the binding mode and provide detailed information for further compound optimization.


Subject(s)
Guanine/analogs & derivatives , Influenza A virus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Binding Sites/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Guanine/chemical synthesis , Guanine/chemistry , Guanine/pharmacology , Models, Molecular , Molecular Structure , RNA-Dependent RNA Polymerase/metabolism , Structure-Activity Relationship , Viral Proteins/metabolism
7.
Cell ; 147(2): 423-35, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-22000019

ABSTRACT

RIG-I is a key innate immune pattern-recognition receptor that triggers interferon expression upon detection of intracellular 5'triphosphate double-stranded RNA (5'ppp-dsRNA) of viral origin. RIG-I comprises N-terminal caspase activation and recruitment domains (CARDs), a DECH helicase, and a C-terminal domain (CTD). We present crystal structures of the ligand-free, autorepressed, and RNA-bound, activated states of RIG-I. Inactive RIG-I has an open conformation with the CARDs sequestered by a helical domain inserted between the two helicase moieties. ATP and dsRNA binding induce a major rearrangement to a closed conformation in which the helicase and CTD bind the blunt end 5'ppp-dsRNA with perfect complementarity but incompatibly with continued CARD binding. We propose that after initial binding of 5'ppp-dsRNA to the flexibly linked CTD, co-operative tight binding of ATP and RNA to the helicase domain liberates the CARDs for downstream signaling. These findings significantly advance our molecular understanding of the activation of innate immune signaling helicases.


Subject(s)
Ducks/metabolism , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Receptors, Pattern Recognition/chemistry , Receptors, Retinoic Acid/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Chickens/immunology , Ducks/immunology , Humans , Models, Molecular , Protein Structure, Tertiary , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Receptors, Pattern Recognition/metabolism , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/immunology
8.
J Immunol ; 185(2): 808-12, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20548024

ABSTRACT

C1q, the recognition subunit of the C1 complex of complement, is an archetypal pattern recognition molecule with the striking ability to sense a wide variety of targets, including a number of altered self-motifs. The recognition properties of its globular domain were further deciphered by means of x-ray crystallography using deoxy-D-ribose and heparan sulfate as ligands. Highly specific recognition of deoxy-D-ribose, involving interactions with Arg C98, Arg C111, and Asn C113, was observed at 1.2 A resolution. Heparin-derived tetrasaccharide interacted more loosely through Lys C129, Tyr C155, and Trp C190. These data together with previous findings define a unique binding area exhibiting both polyanion and deoxy-D-ribose recognition properties, located on the inner face of C1q. DNA and heparin compete for C1q binding but are poor C1 activators compared with immune complexes. How the location of this binding area in C1q may regulate the level of C1 activation is discussed.


Subject(s)
Complement C1q/chemistry , Deoxyribose/chemistry , Heparitin Sulfate/chemistry , Protein Structure, Tertiary , Binding Sites , Complement C1q/metabolism , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , Deoxyribose/metabolism , Heparitin Sulfate/metabolism , Humans , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Protein Binding , Tryptophan/chemistry , Tryptophan/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
9.
J Biol Chem ; 285(9): 6612-22, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20032467

ABSTRACT

Ficolins are oligomeric innate immune recognition proteins consisting of a collagen-like region and a fibrinogen-like recognition domain that bind to pathogen- and apoptotic cell-associated molecular patterns. To investigate their carbohydrate binding specificities, serum-derived L-ficolin and recombinant H- and M-ficolins were fluorescently labeled, and their carbohydrate binding ability was analyzed by glycan array screening. L-ficolin preferentially recognized disulfated N-acetyllactosamine and tri- and tetrasaccharides containing terminal galactose or N-acetylglucosamine. Binding was sensitive to the position and orientation of the bond between N-acetyllactosamine and the adjacent carbohydrate. No significant binding of H-ficolin to any of the 377 glycans probed could be detected, providing further evidence for its poor lectin activity. M-ficolin bound preferentially to 9-O-acetylated 2-6-linked sialic acid derivatives and to various glycans containing sialic acid engaged in a 2-3 linkage. To further investigate the structural basis of sialic acid recognition by M-ficolin, point mutants were produced in which three residues of the fibrinogen domain were replaced by their counterparts in L-ficolin. Mutations G221F and A256V inhibited binding to the 9-O-acetylated sialic acid derivatives, whereas Y271F abolished interaction with all sialic acid-containing glycans. The crystal structure of the Y271F mutant fibrinogen domain was solved, showing that the mutation does not alter the structure of the ligand binding pocket. These analyses reveal novel ficolin ligands such as sulfated N-acetyllactosamine (L-ficolin) and gangliosides (M-ficolin) and provide precise insights into the sialic acid binding specificity of M-ficolin, emphasizing the essential role of Tyr(271) in this respect.


Subject(s)
Carbohydrates/chemistry , Lectins/metabolism , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Crystallography, X-Ray , Fibrinogen/chemistry , Fibrinogen/genetics , Humans , Lectins/chemistry , Mutagenesis, Site-Directed , Mutation, Missense , N-Acetylneuraminic Acid/chemistry , Polysaccharides/chemistry , Protein Array Analysis , Protein Binding , Tyrosine , Ficolins
10.
Mol Cell Proteomics ; 9(4): 593-610, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20008834

ABSTRACT

C1q is a subunit of the C1 complex, a key player in innate immunity that triggers activation of the classical complement pathway. Featuring a unique structural organization and comprising a collagen-like domain with a high level of post-translational modifications, C1q represents a challenging protein assembly for structural biology. We report for the first time a comprehensive proteomics study of C1q combining bottom-up and top-down analyses. C1q was submitted to proteolytic digestion by a combination of collagenase and trypsin for bottom-up analyses. In addition to classical LC-MS/MS analyses, which provided reliable identification of hydroxylated proline and lysine residues, sugar loss-triggered MS(3) scans were acquired on an LTQ-Orbitrap (Linear Quadrupole Ion Trap-Orbitrap) instrument to strengthen the localization of glucosyl-galactosyl disaccharide moieties on hydroxylysine residues. Top-down analyses performed on the same instrument allowed high accuracy and high resolution mass measurements of the intact full-length C1q polypeptide chains and the iterative fragmentation of the proteins in the MS(n) mode. This study illustrates the usefulness of combining the two complementary analytical approaches to obtain a detailed characterization of the post-translational modification pattern of the collagen-like domain of C1q and highlights the structural heterogeneity of individual molecules. Most importantly, three lysine residues of the collagen-like domain, namely Lys(59) (A chain), Lys(61) (B chain), and Lys(58) (C chain), were unambiguously shown to be completely unmodified. These lysine residues are located about halfway along the collagen-like fibers. They are thus fully available and in an appropriate position to interact with the C1r and C1s protease partners of C1q and are therefore likely to play an essential role in C1 assembly.


Subject(s)
Complement C1q/analysis , Complement C1r/metabolism , Complement C1s/metabolism , Mass Spectrometry/methods , Protein Interaction Mapping/methods , Amino Acid Sequence , Binding Sites , Complement C1q/chemistry , Complement C1q/metabolism , Complement C1r/chemistry , Complement C1s/chemistry , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Processing, Post-Translational/physiology , Protein Structure, Quaternary , Sequence Analysis, Protein , Structure-Activity Relationship
11.
J Biol Chem ; 284(29): 19340-8, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19473974

ABSTRACT

The C1 complex of complement is assembled from a recognition protein C1q and C1s-C1r-C1r-C1s, a Ca(2+)-dependent tetramer of two modular proteases C1r and C1s. Resolution of the x-ray structure of the N-terminal CUB(1)-epidermal growth factor (EGF) C1s segment has led to a model of the C1q/C1s-C1r-C1r-C1s interaction where the C1q collagen stem binds at the C1r/C1s interface through ionic bonds involving acidic residues contributed by the C1r EGF module (Gregory, L. A., Thielens, N. M., Arlaud, G. J., Fontecilla-Camps, J. C., and Gaboriaud, C. (2003) J. Biol. Chem. 278, 32157-32164). To identify the C1q-binding sites of C1s-C1r-C1r-C1s, a series of C1r and C1s mutants was expressed, and the C1q binding ability of the resulting tetramer variants was assessed by surface plasmon resonance. Mutations targeting the Glu(137)-Glu-Asp(139) stretch in the C1r EGF module had no effect on C1 assembly, ruling out our previous interaction model. Additional mutations targeting residues expected to participate in the Ca(2+)-binding sites of the C1r and C1s CUB modules provided evidence for high affinity C1q-binding sites contributed by the C1r CUB(1) and CUB(2) modules and lower affinity sites contributed by C1s CUB(1). All of the sites implicate acidic residues also contributing Ca(2+) ligands. C1s-C1r-C1r-C1s thus contributes six C1q-binding sites, one per C1q stem. Based on the location of these sites and available structural information, we propose a refined model of C1 assembly where the CUB(1)-EGF-CUB(2) interaction domains of C1r and C1s are entirely clustered inside C1q and interact through six binding sites with reactive lysines of the C1q stems. This mechanism is similar to that demonstrated for mannan-binding lectin (MBL)-MBL-associated serine protease and ficolin-MBL-associated serine protease complexes.


Subject(s)
Complement C1 Inactivator Proteins/metabolism , Complement C1q/metabolism , Complement C1r/metabolism , Amino Acid Sequence , Binding Sites/genetics , Calcium/metabolism , Complement C1 Inactivator Proteins/chemistry , Complement C1 Inactivator Proteins/genetics , Complement C1 Inhibitor Protein , Complement C1q/chemistry , Complement C1q/genetics , Complement C1r/chemistry , Complement C1r/genetics , Electrophoresis, Polyacrylamide Gel , Epidermal Growth Factor/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutation , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance
12.
FEBS Lett ; 582(20): 3111-6, 2008 Sep 03.
Article in English | MEDLINE | ID: mdl-18703056

ABSTRACT

C1q, the binding subunit of the C1 complex of complement, is an archetypal pattern recognition molecule known for its striking ability to recognize a wide variety of targets, ranging from pathogenic non self to altered self. DNA is one of the C1q ligands, but the precise region of C1q and the DNA motifs that support interaction have not been characterized yet. Here, we report for the first time that the peripheral globular region of the C1q molecule displays a lectin-like activity, which contributes to DNA binding through interaction with its deoxy-d-ribose moiety and may participate in apoptotic cell recognition.


Subject(s)
Apoptosis , Complement C1q/metabolism , DNA/metabolism , Lectins/metabolism , Binding Sites , Complement C1q/genetics , DNA/chemistry , Electrophoretic Mobility Shift Assay , Hexoses/chemistry , Hexoses/metabolism , Humans , Pentoses/chemistry , Pentoses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...