Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 436(9): 168542, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38492718

ABSTRACT

PrimPol is a human DNA primase-polymerase which restarts DNA synthesis beyond DNA lesions and non-B DNA structures blocking replication. Disfunction of PrimPol in cells leads to slowing of DNA replication rates in mitochondria and nucleus, accumulation of chromosome aberrations, cell cycle delay, and elevated sensitivity to DNA-damaging agents. A defective PrimPol has been suggested to be associated with the development of ophthalmic diseases, elevated mitochondrial toxicity of antiviral drugs and increased cell resistance to chemotherapy. Here, we describe a rare missense PrimPol variant V102A with altered biochemical properties identified in patients suffering from ovarian and cervical cancer. The Val102 to Ala substitution dramatically reduced both the primase and DNA polymerase activities of PrimPol as well as specifically decreased its ability to incorporate ribonucleotides. Structural analysis indicates that the V102A substitution can destabilize the hydrophobic pocket adjacent to the active site, affecting dNTP binding and catalysis.


Subject(s)
DNA Primase , DNA-Directed DNA Polymerase , Multifunctional Enzymes , Mutation, Missense , Ovarian Neoplasms , Uterine Cervical Neoplasms , Female , Humans , Amino Acid Substitution , Catalytic Domain , Crystallography, X-Ray , DNA Primase/metabolism , DNA Primase/chemistry , DNA Primase/genetics , DNA Replication , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/chemistry , Models, Molecular , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/chemistry , Protein Conformation , Uterine Cervical Neoplasms/genetics , Ovarian Neoplasms/genetics
2.
Nucleic Acids Res ; 51(14): 7541-7551, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37326028

ABSTRACT

Human PrimPol possesses DNA primase and DNA polymerase activities and restarts stalled replication forks protecting cells against DNA damage in nuclei and mitochondria. The zinc-binding motif (ZnFn) of the C-terminal domain (CTD) of PrimPol is required for DNA primase activity but the mechanism is not clear. In this work, we biochemically demonstrate that PrimPol initiates de novo DNA synthesis in cis-orientation, when the N-terminal catalytic domain (NTD) and the CTD of the same molecule cooperate for substrates binding and catalysis. The modeling studies revealed that PrimPol uses a similar mode of initiating NTP coordination as the human primase. The ZnFn motif residue Arg417 is required for binding the 5'-triphosphate group that stabilizes the PrimPol complex with a DNA template-primer. We found that the NTD alone is able to initiate DNA synthesis, and the CTD stimulates the primase activity of NTD. The regulatory role of the RPA-binding motif in the modulation of PrimPol binding to DNA is also demonstrated.


Subject(s)
DNA Primase , DNA-Directed DNA Polymerase , Humans , DNA-Directed DNA Polymerase/metabolism , DNA Primase/metabolism , DNA Replication , DNA/genetics , DNA Primers , Catalysis , Multifunctional Enzymes/chemistry
3.
Biochemistry ; 61(7): 575-582, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35285625

ABSTRACT

Human phosphoribosylaminoimidazole carboxylase phosphoribosylaminoimdiazole succinocarboxamide synthetase (PAICS) is a dual activity enzyme catalyzing two consecutive reactions in de novo purine nucleotide synthesis. Crystallographic structures of recombinant human PAICS suggested the channeling of 4-carboxy-5-aminoimidazole-1-ribose-5'-phosphate (CAIR) between two active sites of PAICS, while a prior work of an avian PAICS suggested otherwise. Here, we present time-course mass spectrometric data supporting the channeling of CAIR between domains of recombinant human PAICS. Time-course mass spectral analysis showed that CAIR added to the bulk solution (CAIRbulk) is decarboxylated and re-carboxylated before the accumulation of succinyl-5-aminoimidazole-4-carboxamide-1-ribose-5'-phosphate (SAICAR). An experiment with 13C-bicarbonate showed that SAICAR production was proportional to re-carboxylated CAIR instead of total CAIR or CAIRbulk. This result indicates that the SAICAR synthase domain selectively uses enzyme-made CAIR over CAIRbulk, which is consistent with the channeling model. This channeling between PAICS domains may be a part of a larger channeling process in de novo purine nucleotide synthesis.


Subject(s)
Bicarbonates , Carboxy-Lyases , Peptide Synthases , Carboxy-Lyases/chemistry , Catalytic Domain , Humans , Multifunctional Enzymes/chemistry , Peptide Synthases/chemistry
4.
Biomolecules ; 12(2)2022 02 03.
Article in English | MEDLINE | ID: mdl-35204749

ABSTRACT

DNA replication can encounter blocking obstacles, leading to replication stress and genome instability. There are several mechanisms for evading this blockade. One mechanism consists of repriming ahead of the obstacles, creating a new starting point; in humans, PrimPol is responsible for carrying out this task. PrimPol is a primase that operates in both the nucleus and mitochondria. In contrast with conventional primases, PrimPol is a DNA primase able to initiate DNA synthesis de novo using deoxynucleotides, discriminating against ribonucleotides. In vitro, PrimPol can act as a DNA primase, elongating primers that PrimPol itself sythesizes, or as translesion synthesis (TLS) DNA polymerase, elongating pre-existing primers across lesions. However, the lack of evidence for PrimPol polymerase activity in vivo suggests that PrimPol only acts as a DNA primase. Here, we provide a comprehensive review of human PrimPol covering its biochemical properties and structure, in vivo function and regulation, and the processes that take place to fill the gap-containing lesion that PrimPol leaves behind. Finally, we explore the available data on human PrimPol expression in different tissues in physiological conditions and its role in cancer.


Subject(s)
Multifunctional Enzymes , Neoplasms , DNA Primase/chemistry , DNA Primase/genetics , DNA Primase/metabolism , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/chemistry , Humans , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Neoplasms/drug therapy , Neoplasms/genetics
5.
Nucleic Acids Res ; 49(20): 11643-11652, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34718701

ABSTRACT

The Repeat Expansion Diseases, a large group of human diseases that includes the fragile X-related disorders (FXDs) and Huntington's disease (HD), all result from expansion of a disease-specific microsatellite via a mechanism that is not fully understood. We have previously shown that mismatch repair (MMR) proteins are required for expansion in a mouse model of the FXDs, but that the FANCD2 and FANCI associated nuclease 1 (FAN1), a component of the Fanconi anemia (FA) DNA repair pathway, is protective. FAN1's nuclease activity has been reported to be dispensable for protection against expansion in an HD cell model. However, we show here that in a FXD mouse model a point mutation in the nuclease domain of FAN1 has the same effect on expansion as a null mutation. Furthermore, we show that FAN1 and another nuclease, EXO1, have an additive effect in protecting against MSH3-dependent expansions. Lastly, we show that the loss of FANCD2, a vital component of the Fanconi anemia DNA repair pathway, has no effect on expansions. Thus, FAN1 protects against MSH3-dependent expansions without diverting the expansion intermediates into the canonical FA pathway and this protection depends on FAN1 having an intact nuclease domain.


Subject(s)
Catalytic Domain , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/metabolism , Multifunctional Enzymes/metabolism , Trinucleotide Repeat Expansion , Animals , DNA Repair Enzymes/metabolism , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Mice , Mice, Inbred C57BL , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/genetics , MutS Homolog 3 Protein/metabolism , Point Mutation
6.
Biochemistry ; 60(42): 3162-3172, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34609847

ABSTRACT

Copalyl diphosphate (CPP) synthase from Penicillium verruculosum (PvCPS) is a bifunctional diterpene synthase with both prenyltransferase and class II cyclase activities. The prenyltransferase α domain catalyzes the condensation of C5 dimethylallyl diphosphate with three successively added C5 isopentenyl diphosphates (IPPs) to form C20 geranylgeranyl diphosphate (GGPP), which then undergoes a class II cyclization reaction at the ßγ domain interface to generate CPP. The prenyltransferase α domain mediates oligomerization to form a 648-kD (αßγ)6 hexamer. In the current study, we explore prenyltransferase structure-function relationships in this oligomeric assembly-line platform with the goal of generating alternative linear isoprenoid products. Specifically, we report steady-state enzyme kinetics, product analysis, and crystal structures of various site-specific variants of the prenyltransferase α domain. Crystal structures of the H786A, F760A, S723Y, S723F, and S723T variants have been determined at resolutions of 2.80, 3.10, 3.15, 2.65, and 2.00 Å, respectively. The substitution of S723 with bulky aromatic amino acids in the S723Y and S723F variants constricts the active site, thereby directing the formation of the shorter C15 isoprenoid, farnesyl diphosphate. While the S723T substitution only subtly alters enzyme kinetics and does not compromise GGPP biosynthesis, the crystal structure of this variant reveals a nonproductive binding mode for IPP that likely accounts for substrate inhibition at high concentrations. Finally, mutagenesis of the catalytic general acid in the class II cyclase domain, D313A, significantly compromises prenyltransferase activity. This result suggests molecular communication between the prenyltransferase and cyclase domains despite their distant connection by a flexible polypeptide linker.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Multifunctional Enzymes/chemistry , Plant Proteins/chemistry , Alkyl and Aryl Transferases/genetics , Catalytic Domain/genetics , Kinetics , Multifunctional Enzymes/genetics , Plant Proteins/genetics , Protein Domains/genetics , Protein Engineering , Talaromyces/enzymology
7.
Sci Rep ; 11(1): 17588, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34475447

ABSTRACT

Human PrimPol belongs to the archaeo-eukaryotic primase superfamily of primases and is involved in de novo DNA synthesis downstream of blocking DNA lesions and non-B DNA structures. PrimPol possesses both DNA/RNA primase and DNA polymerase activities, and also bypasses a number of DNA lesions in vitro. In this work, we have analyzed translesion synthesis activity of PrimPol in vitro on DNA with an 1,2-intrastrand cisplatin cross-link (1,2-GG CisPt CL) or a model DNA-protein cross-link (DpCL). PrimPol was capable of the 1,2-GG CisPt CL bypass in the presence of Mn2+ ions and preferentially incorporated two complementary dCMPs opposite the lesion. Nucleotide incorporation was stimulated by PolDIP2, and yeast Pol ζ efficiently extended from the nucleotides inserted opposite the 1,2-GG CisPt CL in vitro. DpCLs significantly blocked the DNA polymerase activity and strand displacement synthesis of PrimPol. However, PrimPol was able to reach the DpCL site in single strand template DNA in the presence of both Mg2+ and Mn2+ ions despite the presence of the bulky protein obstacle.


Subject(s)
Cisplatin/chemistry , DNA Damage , DNA Primase/chemistry , DNA Replication , DNA-Directed DNA Polymerase/chemistry , DNA/chemistry , Multifunctional Enzymes/chemistry , Cisplatin/pharmacology , Cross-Linking Reagents/chemistry , DNA Repair , Humans
8.
Mol Cell Biol ; 41(9): e0030321, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34228493

ABSTRACT

Germline mutations in the mismatch repair (MMR) genes MSH2, MSH6, MLH1, and PMS2 are linked to cancer of the colon and other organs, characterized by microsatellite instability and a large increase in mutation frequency. Unexpectedly, mutations in EXO1, encoding the only exonuclease genetically implicated in MMR, are not linked to familial cancer and cause a substantially weaker mutator phenotype. This difference could be explained if eukaryotic cells possessed additional exonucleases redundant with EXO1. Analysis of the MLH1 interactome identified FANCD2-associated nuclease 1 (FAN1), a novel enzyme with biochemical properties resembling EXO1. We now show that FAN1 efficiently substitutes for EXO1 in MMR assays and that this functional complementation is modulated by its interaction with MLH1. FAN1 also contributes to MMR in vivo; cells lacking both EXO1 and FAN1 have an MMR defect and display resistance to N-methyl-N-nitrosourea (MNU) and 6-thioguanine (TG). Moreover, FAN1 loss amplifies the mutational profile of EXO1-deficient cells, suggesting that the two nucleases act redundantly in the same antimutagenic pathway. However, the increased drug resistance and mutator phenotype of FAN1/EXO1-deficient cells are less prominent than those seen in cells lacking MSH6 or MLH1. Eukaryotic cells thus apparently possess additional mechanisms that compensate for the loss of EXO1.


Subject(s)
Avian Proteins/metabolism , DNA Mismatch Repair , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/metabolism , Multifunctional Enzymes/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chickens , Endodeoxyribonucleases/chemistry , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/deficiency , Exodeoxyribonucleases/genetics , Guanosine/analogs & derivatives , HEK293 Cells , Humans , Methylnitronitrosoguanidine , Multifunctional Enzymes/chemistry , Mutation/genetics , Thionucleosides
9.
ACS Chem Biol ; 16(10): 1994-2003, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34181849

ABSTRACT

Myogenic differentiation, the irreversible developmental process where precursor myoblast muscle stem cells become contractile myotubes, is heavily regulated by glycosylation and glycan-protein interactions at the cell surface and the extracellular matrix. The glycan-binding protein galectin-1 has been found to be a potent activator of myogenic differentiation. While it is being explored as a potential therapeutic for muscle repair, a precise understanding of its glycoprotein interactors is lacking. These gaps are due in part to the difficulties of capturing glycan-protein interactions in live cells. Here, we demonstrate the use of a proximity tagging strategy coupled with quantitative mass-spectrometry-based proteomics to capture, enrich, and identify the glycan-mediated glycoprotein interactors of galectin-1 in cultured live mouse myoblasts. Our interactome dataset can serve as a resource to aid the determination of mechanisms through which galectin-1 promotes myogenic differentiation. Moreover, it can also facilitate the determination of the physiological glycoprotein counter-receptors of galectin-1. Indeed, we identify several known and novel glycan-mediated ligands of galectin-1 as well as validate that galectin-1 binds the native CD44 glycoprotein in a glycan-mediated manner.


Subject(s)
Galectin 1/metabolism , Glycoproteins/metabolism , Animals , Biotin/analogs & derivatives , Biotinylation , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Endonucleases/chemistry , Endonucleases/metabolism , Galectin 1/chemistry , Glycomics , Glycoproteins/chemistry , Humans , Ligands , Mice , Molecular Probes/chemistry , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/metabolism , Myoblasts , Phenols/chemistry , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
10.
Biochemistry ; 60(26): 2071-2083, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34156819

ABSTRACT

Xylan and cellulose are the two major constituents of numerous types of lignocellulose. The bifunctional enzyme that exhibits xylanase/cellulase activity has attracted a great deal of attention in biofuel production. Previously, a thermostable GH10 family enzyme (XynA) from Bacillus sp. KW1 was found to degrade both xylan and cellulose. To improve bifunctional activity on the basis of structure, we first determined the crystal structure of XynA at 2.3 Å. Via molecular docking and activity assays, we revealed that Gln250 and His252 were indispensable to bifunctionality, because they could interact with two conserved catalytic residues, Glu182 and Glu280, while bringing the substrate close to the activity pocket. Then we used a structure-based engineering strategy to improve xylanase/cellulase activity. Although no mutants with increased bifunctional activity were obtained after much screening, we found the answer in the N-terminal 36-amino acid truncation of XynA. The activities of XynA_ΔN36 toward beechwood xylan, wheat arabinoxylan, filter paper, and barley ß-glucan were significantly increased by 0.47-, 0.53-, 2.46-, and 1.04-fold, respectively. Furthermore, upon application, this truncation released more reducing sugars than the wild type in the degradation of pretreated corn stover and sugar cane bagasse. These results showed the detailed molecular mechanism of the GH10 family bifunctional endoxylanase/cellulase. The basis of these catalytic performances and the screened XynA_ΔN36 provide clues for the further use of XynA in industrial applications.


Subject(s)
Bacterial Proteins/chemistry , Cellulase/chemistry , Endo-1,4-beta Xylanases/chemistry , Multifunctional Enzymes/chemistry , Amino Acid Sequence , Bacillus/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Cellulase/genetics , Cellulase/metabolism , Endo-1,4-beta Xylanases/genetics , Endo-1,4-beta Xylanases/metabolism , Molecular Docking Simulation , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutagenesis, Site-Directed , Mutation , Oligosaccharides/metabolism , Protein Binding , Protein Engineering , Xylans/metabolism
11.
STAR Protoc ; 2(2): 100506, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997820

ABSTRACT

The ascorbate peroxidase APEX2 is commonly used to study the neighborhood of a protein of interest by proximity-dependent biotinylation. Here, we describe a protocol for sample processing compatible with immunoblotting and mass spectrometry, suitable to specifically map the content of autophagosomes and potentially other short-lived endomembrane transport vesicles without the need of subcellular fractionation. By combining live-cell biotinylation with proteinase K digestion of cell homogenates, proteins enriched in membrane-protected compartments can be readily enriched and identified. For complete details on the use and execution of this protocol, please refer to Zellner et al. (2021).


Subject(s)
Autophagosomes , DNA-(Apurinic or Apyrimidinic Site) Lyase , Endonucleases , Endopeptidase K/chemistry , Multifunctional Enzymes , Proteolysis , Proteomics , Autophagosomes/chemistry , Autophagosomes/metabolism , Biotinylation , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Endonucleases/chemistry , Endonucleases/metabolism , HeLa Cells , Humans , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/metabolism
12.
J Biol Inorg Chem ; 26(1): 93-108, 2021 02.
Article in English | MEDLINE | ID: mdl-33544225

ABSTRACT

The Schizosaccharomyces pombe Asp1 protein is a bifunctional kinase/pyrophosphatase that belongs to the highly conserved eukaryotic diphosphoinositol pentakisphosphate kinase PPIP5K/Vip1 family. The N-terminal Asp1 kinase domain generates specific high-energy inositol pyrophosphate (IPP) molecules, which are hydrolyzed by the C-terminal Asp1 pyrophosphatase domain (Asp1365-920). Thus, Asp1 activities regulate the intracellular level of a specific class of IPP molecules, which control a wide number of biological processes ranging from cell morphogenesis to chromosome transmission. Recently, it was shown that chemical reconstitution of Asp1371-920 leads to the formation of a [2Fe-2S] cluster; however, the biological relevance of the cofactor remained under debate. In this study, we provide evidence for the presence of the Fe-S cluster in Asp1365-920 inside the cell. However, we show that the Fe-S cluster does not influence Asp1 pyrophosphatase activity in vitro or in vivo. Characterization of the as-isolated protein by electronic absorption spectroscopy, mass spectrometry, and X-ray absorption spectroscopy is consistent with the presence of a [2Fe-2S]2+ cluster in the enzyme. Furthermore, we have identified the cysteine ligands of the cluster. Overall, our work reveals that Asp1 contains an Fe-S cluster in vivo that is not involved in its pyrophosphatase activity.


Subject(s)
Cytoskeletal Proteins/chemistry , Iron-Sulfur Proteins/chemistry , Pyrophosphatases/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/enzymology , Biocatalysis , Cysteine/chemistry , Cytoskeletal Proteins/genetics , Iron-Sulfur Proteins/genetics , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/genetics , Mutation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Pyrophosphatases/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/genetics
13.
Nucleic Acids Res ; 49(4): 2179-2191, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33533925

ABSTRACT

Replication forks often stall at damaged DNA. To overcome these obstructions and complete the DNA duplication in a timely fashion, replication can be restarted downstream of the DNA lesion. In mammalian cells, this repriming of replication can be achieved through the activities of primase and polymerase PrimPol. PrimPol is stimulated in DNA synthesis through interaction with PolDIP2, however the exact mechanism of this PolDIP2-dependent stimulation is still unclear. Here, we show that PrimPol uses a flexible loop to interact with the C-terminal ApaG-like domain of PolDIP2, and that this contact is essential for PrimPol's enhanced processivity. PolDIP2 increases primer-template and dNTP binding affinities of PrimPol, which concomitantly enhances its nucleotide incorporation efficiency. This stimulation is dependent on a unique arginine cluster in PolDIP2. Since the polymerase activity of PrimPol alone is very limited, this mechanism, where the affinity for dNTPs gets increased by PolDIP2 binding, might be critical for the in vivo function of PrimPol in tolerating DNA lesions at physiological nucleotide concentrations.


Subject(s)
Arginine/chemistry , DNA Primase/chemistry , DNA-Directed DNA Polymerase/chemistry , DNA/biosynthesis , Multifunctional Enzymes/chemistry , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Amino Acid Motifs , DNA Primase/metabolism , DNA-Directed DNA Polymerase/metabolism , Deoxyribonucleotides/chemistry , Deoxyribonucleotides/metabolism , Models, Molecular , Multifunctional Enzymes/metabolism , Protein Binding
14.
DNA Repair (Amst) ; 100: 103048, 2021 04.
Article in English | MEDLINE | ID: mdl-33571927

ABSTRACT

Human primase and DNA polymerase PrimPol re-starts stalled replication forks by repriming downstream DNA lesions and protects cells against DNA damage. Structure of the catalytic core of PrimPol with DNA primer, template and incoming dATP was solved but the mechanisms of DNA polymerase and primase activities of PrimPol are not fully understood. In this work, using site-directed mutagenesis we biochemically analyzed the role of active site residues Arg47 and Arg76 contacting DNA template in DNA polymerase and primase activities of PrimPol. The substitution R47A diminished the DNA polymerase and primase activities of PrimPol whereas the single amino acid substitution R76A caused almost complete loss of catalytic activities. Both amino acid substitutions affected the spectrum of dNMPs incorporation on undamaged DNA templates and opposite 8-oxoguanine. Finally, substitutions of the Arg47 and Arg76 residues attenuated the formation of the stable PrimPol:DNA complex in the presence of ATP/dNTPs. Together, these findings suggest a key role of the Arg47 and Arg76 in DNA synthesis by PrimPol.


Subject(s)
Catalytic Domain , DNA Damage , DNA Primase/metabolism , DNA Replication , DNA-Directed DNA Polymerase/metabolism , DNA/metabolism , Guanine/analogs & derivatives , Multifunctional Enzymes/metabolism , Amino Acid Sequence , DNA Primase/chemistry , DNA-Directed DNA Polymerase/chemistry , Guanine/metabolism , Humans , Multifunctional Enzymes/chemistry , Sequence Alignment
15.
EMBO J ; 40(9): e105853, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33555040

ABSTRACT

p97ATPase-mediated membrane fusion is required for the biogenesis of the Golgi complex. p97 and its cofactor p47 function in soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor (SNARE) priming, but the tethering complex for p97/p47-mediated membrane fusion remains unknown. In this study, we identified formiminotransferase cyclodeaminase (FTCD) as a novel p47-binding protein. FTCD mainly localizes to the Golgi complex and binds to either p47 or p97 via its association with their polyglutamate motifs. FTCD functions in p97/p47-mediated Golgi reassembly at mitosis in vivo and in vitro via its binding to p47 and to p97. We also showed that FTCD, p47, and p97 form a big FTCD-p97/p47-FTCD tethering complex. In vivo tethering assay revealed that FTCD that was designed to localize to mitochondria caused mitochondria aggregation at mitosis by forming a complex with endogenous p97 and p47, which support a role for FTCD in tethering biological membranes in cooperation with the p97/p47 complex. Therefore, FTCD is thought to act as a tethering factor by forming the FTCD-p97/p47-FTCD complex in p97/p47-mediated Golgi membrane fusion.


Subject(s)
Ammonia-Lyases/metabolism , Glutamate Formimidoyltransferase/metabolism , Golgi Apparatus/metabolism , Multifunctional Enzymes/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Valosin Containing Protein/metabolism , Ammonia-Lyases/chemistry , Binding Sites , Glutamate Formimidoyltransferase/chemistry , HeLa Cells , Hep G2 Cells , Humans , Membrane Fusion , Mitochondria , Mitosis , Multifunctional Enzymes/chemistry , Multiprotein Complexes/metabolism , Protein Binding
16.
Gene ; 771: 145360, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33333218

ABSTRACT

Variants in SETX have been implicated in recessively and dominantly inherited disorders, ataxia with oculomotor apraxia type 2 (AOA2 OMIM# 606002) and amyotrophic lateral sclerosis (ALS4, OMIM# 602433) respectively, in humans. We report two novel bi-allelic pathogenic variants in SETX in patients suffering from ataxia with oculomotor apraxia type 2, extending the allelic spectrum of the gene variants. We also discuss the pathogenicity of SETX variants in relation to the evolutionary conservation status of the affected amino acids. Our analyses suggest that variants of some amino acids which are not fully conserved in evolution, may cause a disorder in humans, provided the particular pathogenic variant is absent in other orthologues.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA Helicases/genetics , Multifunctional Enzymes/genetics , Mutation, Missense , RNA Helicases/genetics , Spinocerebellar Ataxias/congenital , Adolescent , DNA Helicases/chemistry , Evolution, Molecular , Female , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Multifunctional Enzymes/chemistry , Pedigree , Protein Domains , RNA Helicases/chemistry , Sequence Analysis, DNA , Spinocerebellar Ataxias/genetics , Young Adult
17.
J Cell Biol ; 220(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33306092

ABSTRACT

The nuclear lamina (NL) is a meshwork found beneath the inner nuclear membrane. The study of the NL is hindered by the insolubility of the meshwork and has driven the development of proximity ligation methods to identify the NL-associated/proximal proteins, RNA, and DNA. To simplify and improve temporal labeling, we fused APEX2 to the NL protein lamin-B1 to map proteins, RNA, and DNA. The identified NL-interacting/proximal RNAs show a long 3' UTR bias, a finding consistent with an observed bias toward longer 3' UTRs in genes deregulated in lamin-null cells. A C-rich motif was identified in these 3' UTR. Our APEX2-based proteomics identifies a C-rich motif binding regulatory protein that exhibits altered localization in lamin-null cells. Finally, we use APEX2 to map lamina-associated domains (LADs) during the cell cycle and uncover short, H3K27me3-rich variable LADs. Thus, the APEX2-based tools presented here permit identification of proteomes, transcriptomes, and genome elements associated with or proximal to the NL.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Endonucleases/metabolism , Multifunctional Enzymes/metabolism , Nuclear Lamina/metabolism , Protein Interaction Mapping , 3' Untranslated Regions/genetics , Base Sequence , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Endonucleases/chemistry , HCT116 Cells , HEK293 Cells , Humans , K562 Cells , Lamin Type B/metabolism , Multifunctional Enzymes/chemistry , Protein Domains , Proteome/metabolism , RNA/metabolism , RNA Splicing/genetics
18.
Crit Rev Biochem Mol Biol ; 56(1): 17-30, 2021 02.
Article in English | MEDLINE | ID: mdl-33179522

ABSTRACT

DNA replication forks are constantly challenged by DNA lesions induced by endogenous and exogenous sources. DNA damage tolerance mechanisms ensure that DNA replication continues with minimal effects on replication fork elongation either by using specialized DNA polymerases, which have the ability to replicate through the damaged template, or by skipping the damaged DNA, leaving it to be repaired after replication. These mechanisms are evolutionarily conserved in bacteria, yeast, and higher eukaryotes, and are paramount to ensure timely and faithful duplication of the genome. The Primase and DNA-directed Polymerase (PRIMPOL) is a recently discovered enzyme that possesses both primase and polymerase activities. PRIMPOL is emerging as a key player in DNA damage tolerance, particularly in vertebrate and human cells. Here, we review our current understanding of the function of PRIMPOL in DNA damage tolerance by focusing on the structural aspects that define its dual enzymatic activity, as well as on the mechanisms that control its chromatin recruitment and expression levels. We also focus on the latest findings on the mitochondrial and nuclear functions of PRIMPOL and on the impact of loss of these functions on genome stability and cell survival. Defining the function of PRIMPOL in DNA damage tolerance is becoming increasingly important in the context of human disease. In particular, we discuss recent evidence pointing at the PRIMPOL pathway as a novel molecular target to improve cancer cell response to DNA-damaging chemotherapy and as a predictive parameter to stratify patients in personalized cancer therapy.


Subject(s)
DNA Damage/genetics , DNA Primase/genetics , DNA Primase/metabolism , DNA Replication/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Cell Nucleus/metabolism , Cell Survival/genetics , Chromatin/metabolism , DNA/genetics , DNA/metabolism , DNA Primase/chemistry , DNA-Directed DNA Polymerase/chemistry , Gene Knockdown Techniques , Genomic Instability , Humans , Mitochondria/metabolism , Multifunctional Enzymes/chemistry
19.
Sci Rep ; 10(1): 9343, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32518272

ABSTRACT

A key component of antiretroviral therapy (ART) for HIV patients is the nucleoside reverse transcriptase inhibitor (NRTI) is tenofovir. Recent reports of tenofovir toxicity in patients taking ART for HIV cannot be explained solely on the basis of off-target inhibition of mitochondrial DNA polymerase gamma (Polγ). PrimPol was discovered as a primase-polymerase localized to the mitochondria with repriming and translesion synthesis capabilities and, therefore, a potential contributor to mitochondrial toxicity. We established a possible role of PrimPol in tenofovir-induced toxicity in vitro and show that tenofovir-diphosphate incorporation by PrimPol is dependent on the n-1 nucleotide. We identified and characterized a PrimPol mutation, D114N, in an HIV+ patient on tenofovir-based ART with mitochondrial toxicity. This mutant form of PrimPol, targeting a catalytic metal ligand, was unable to synthesize primers, likely due to protein instability and weakened DNA binding. We performed cellular respiration and toxicity assays using PrimPol overexpression and shRNA knockdown strains in renal proximal tubular epithelial cells. The PrimPol-knockdown strain was hypersensitive to tenofovir treatment, indicating that PrimPol protects against tenofovir-induced mitochondrial toxicity. We show that a major cellular role of PrimPol is protecting against toxicity caused by ART and individuals with inactivating mutations may be predisposed to these effects.


Subject(s)
DNA Primase/genetics , DNA Primase/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , HIV Infections/enzymology , HIV Infections/genetics , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutation , Tenofovir/toxicity , Animals , Biocatalysis , DNA Primase/chemistry , DNA Primase/deficiency , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/deficiency , Enzyme Stability , Gene Knockdown Techniques , Humans , Kidney/drug effects , Kinetics , Mitochondria/drug effects , Mitochondria/metabolism , Models, Molecular , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/deficiency , Protein Multimerization , Protein Structure, Quaternary
20.
Methods Mol Biol ; 2169: 1-10, 2020.
Article in English | MEDLINE | ID: mdl-32548814

ABSTRACT

Caveolae are small flask- or cup-shaped invaginations of the plasma membrane found in almost all vertebrate cells. Due to their small size (50-100 nm), transmission electron microscopy (TEM) has been the method of choice to study caveolae formation and ultrastructure and, more recently, to resolve the sub-caveolar localization of its protein components using novel protein labeling methods for TEM. This chapter describes a protocol for the selective visualization of caveolae and caveolar proteins by TEM, 3D tomography, and correlative light and electron microscopy (CLEM) using the peroxidase APEX2.


Subject(s)
Caveolae/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Endonucleases/chemistry , Microscopy, Electron, Transmission/methods , Multifunctional Enzymes/chemistry , Animals , Caveolae/ultrastructure , Caveolin 1/metabolism , Cell Line , Humans , RNA-Binding Proteins/metabolism , Tomography/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...