Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 285
Filtrar
1.
J Am Chem Soc ; 146(14): 9583-9596, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38538061

RESUMO

Primases are crucial enzymes for DNA replication, as they synthesize a short primer required for initiating DNA replication. We herein present time-resolved nuclear magnetic resonance (NMR) spectroscopy in solution and in the solid state to study the initial dinucleotide formation reaction of archaeal pRN1 primase. Our findings show that the helix-bundle domain (HBD) of pRN1 primase prepares the two substrates and then hands them over to the catalytic domain to initiate the reaction. By using nucleotide triphosphate analogues, the reaction is substantially slowed down, allowing us to study the initial dinucleotide formation in real time. We show that the sedimented protein-DNA complex remains active in the solid-state NMR rotor and that time-resolved 31P-detected cross-polarization experiments allow monitoring the kinetics of dinucleotide formation. The kinetics in the sedimented protein sample are comparable to those determined by solution-state NMR. Protein conformational changes during primer synthesis are observed in time-resolved 1H-detected experiments at fast magic-angle spinning frequencies (100 kHz). A significant number of spectral changes cluster in the HBD pointing to the importance of the HBD for positioning the nucleotides and the dinucleotide.


Assuntos
Carcinoma Papilar , Carcinoma de Células Renais , DNA Primase , Replicação do DNA , Neoplasias da Glândula Tireoide , DNA Primase/química , Nucleotídeos , Espectroscopia de Ressonância Magnética
2.
J Mol Biol ; 436(9): 168542, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38492718

RESUMO

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.


Assuntos
DNA Primase , DNA Polimerase Dirigida por DNA , Enzimas Multifuncionais , Mutação de Sentido Incorreto , DNA Primase/metabolismo , DNA Primase/química , DNA Primase/genética , Humanos , Enzimas Multifuncionais/metabolismo , Enzimas Multifuncionais/genética , Enzimas Multifuncionais/química , DNA Polimerase Dirigida por DNA/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/química , Feminino , Replicação do DNA , Modelos Moleculares , Substituição de Aminoácidos , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/genética , Conformação Proteica , Cristalografia por Raios X , Domínio Catalítico
3.
Nat Struct Mol Biol ; 31(1): 68-81, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38177671

RESUMO

The Mpox pandemic, caused by the Mpox virus (or monkeypox virus, MPXV), has gained global attention. The D5 protein, a putative helicase-primase found in MPXV, plays a vital role in viral replication and genome uncoating. Here we determined multiple cryo-EM structures of full-length hexameric D5 in diverse states. These states were captured during ATP hydrolysis while moving along the single-stranded DNA (ssDNA) track. Through comprehensive structural analysis combined with the helicase activity system, we revealed that when the primase domain is truncated or the interaction between the primase and helicase domains is disrupted, the double-stranded DNA (dsDNA) unwinds into ssDNA, suggesting a critical regulatory role of the primase domain. Two transition states bound with ssDNA substrate during unwinding reveals that two ATP molecules were consumed to drive DNA moving forward two nucleotides. Collectively, our findings shed light on the molecular mechanism that links ATP hydrolysis to the DNA unwinding in poxviruses.


Assuntos
DNA Primase , Vírus da Varíola dos Macacos , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , Vírus da Varíola dos Macacos/genética , Vírus da Varíola dos Macacos/metabolismo , DNA Helicases/metabolismo , DNA/química , DNA de Cadeia Simples , Trifosfato de Adenosina/metabolismo
4.
Mol Inform ; 43(3): e202300284, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38123523

RESUMO

Tuberculosis (TB) is the second leading cause of mortality after COVID-19, with a global death toll of 1.6 million in 2021. The escalating situation of drug-resistant forms of TB has threatened the current TB management strategies. New therapeutics with novel mechanisms of action are urgently required to address the current global TB crisis. The essential mycobacterial primase DnaG with no structural homology to homo sapiens presents itself as a good candidate for drug targeting. In the present study, Mitoxantrone and Vapreotide, two FDA-approved drugs, were identified as potential anti-mycobacterial agents. Both Mitoxantrone and Vapreotide exhibit a strong Minimum Inhibitory Concentration (MIC) of ≤25µg/ml against both the virulent (M.tb-H37Rv) and avirulent (M.tb-H37Ra) strains of M.tb. Extending the validations further revealed the inhibitory potential drugs in ex vivo conditions. Leveraging the computational high-throughput multi-level docking procedures from the pool of ~2700 FDA-approved compounds, Mitoxantrone and Vapreotide were screened out as potential inhibitors of DnaG. Extensive 200 ns long all-atoms molecular dynamic simulation of DnaGDrugs complexes revealed that both drugs bind strongly and stabilize the DnaG during simulations. Reduced solvent exposure and confined motions of the active centre of DnaG upon complexation with drugs indicated that both drugs led to the closure of the active site of DnaG. From this study's findings, we propose Mitoxantrone and Vapreotide as potential anti-mycobacterial agents, with their novel mechanism of action against mycobacterial DnaG.


Assuntos
Mycobacterium tuberculosis , Somatostatina/análogos & derivados , Humanos , Antituberculosos/farmacologia , DNA Primase/química , DNA Primase/metabolismo , Mitoxantrona/farmacologia
5.
BMC Plant Biol ; 23(1): 467, 2023 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-37803262

RESUMO

BACKGROUND: The mechanisms and regulation for DNA replication in plant organelles are largely unknown, as few proteins involved in replisome assembly have been biochemically studied. A primase-helicase dubbed Twinkle (T7 gp4-like protein with intramitochondrial nucleoid localization) unwinds double-stranded DNA in metazoan mitochondria and plant organelles. Twinkle in plants is a bifunctional enzyme with an active primase module. This contrast with animal Twinkle in which the primase module is inactive. The organellar primase-helicase of Arabidopsis thaliana (AtTwinkle) harbors a primase module (AtPrimase) that consists of an RNA polymerase domain (RPD) and a Zn + + finger domain (ZFD). RESULTS: Herein, we investigate the mechanisms by which AtTwinkle recognizes its templating sequence and how primer synthesis and coupling to the organellar DNA polymerases occurs. Biochemical data show that the ZFD of the AtPrimase module is responsible for template recognition, and this recognition is achieved by residues N163, R166, and K168. The role of the ZFD in template recognition was also corroborated by swapping the RPDs of bacteriophage T7 primase and AtPrimase with their respective ZFDs. A chimeric primase harboring the ZFD of T7 primase and the RPD of AtPrimase synthesizes ribonucleotides from the T7 primase recognition sequence and conversely, a chimeric primase harboring the ZFD of AtPrimase and the RPD of T7 primase synthesizes ribonucleotides from the AtPrimase recognition sequence. A chimera harboring the RPDs of bacteriophage T7 and the ZBD of AtTwinkle efficiently synthesizes primers for the plant organellar DNA polymerase. CONCLUSIONS: We conclude that the ZFD is responsible for recognizing a single-stranded sequence and for primer hand-off into the organellar DNA polymerases active site. The primase activity of plant Twinkle is consistent with phylogeny-based reconstructions that concluded that Twinkle´s last eukaryotic common ancestor (LECA) was an enzyme with primase and helicase activities. In plants, the primase domain is active, whereas the primase activity was lost in metazoans. Our data supports the notion that AtTwinkle synthesizes primers at the lagging-strand of the organellar replication fork.


Assuntos
Arabidopsis , DNA Primase , Animais , DNA Primase/genética , DNA Primase/química , DNA Primase/metabolismo , DNA Helicases/química , DNA Helicases/genética , DNA Helicases/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Arabidopsis/metabolismo , Mitocôndrias/metabolismo , Dedos de Zinco , Ribonucleotídeos , Replicação do DNA , Bacteriófago T7/genética
6.
Curr Opin Struct Biol ; 82: 102652, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37459807

RESUMO

Members of the primase-polymerase (Prim-Pol) superfamily are found in all domains of life and play diverse roles in genome stability, including primer synthesis during DNA replication, lesion repair and damage tolerance. This review focuses primarily on Prim-Pol members capable of de novo primer synthesis that have experimentally derived structural models available. We discuss the mechanism of DNA primer synthesis initiation by Prim-Pol catalytic domains, based on recent structural and functional studies. We also describe a general model for primer initiation that also includes the ancillary domains/subunits, which stimulate the initiation of primer synthesis.


Assuntos
DNA Primase , Replicação do DNA , DNA Primase/química , Domínio Catalítico
7.
Nat Struct Mol Biol ; 30(5): 579-583, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37069376

RESUMO

The synthesis of RNA-DNA primer by primosome requires coordination between primase and DNA polymerase α subunits, which is accompanied by unknown architectural rearrangements of multiple domains. Using cryogenic electron microscopy, we solved a 3.6 Å human primosome structure caught at an early stage of RNA primer elongation with deoxynucleotides. The structure confirms a long-standing role of primase large subunit and reveals new insights into how primosome is limited to synthesizing short RNA-DNA primers.


Assuntos
DNA Primase , DNA , Humanos , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , DNA/química , Replicação do DNA , Primers do DNA , RNA
8.
Viruses ; 14(10)2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36298761

RESUMO

Poxviruses are large DNA viruses with a linear double-stranded DNA genome circularized at the extremities. The helicase-primase D5, composed of six identical 90 kDa subunits, is required for DNA replication. D5 consists of a primase fragment flexibly attached to the hexameric C-terminal polypeptide (res. 323-785) with confirmed nucleotide hydrolase and DNA-binding activity but an elusive helicase activity. We determined its structure by single-particle cryo-electron microscopy. It displays an AAA+ helicase core flanked by N- and C-terminal domains. Model building was greatly helped by the predicted structure of D5 using AlphaFold2. The 3.9 Å structure of the N-terminal domain forms a well-defined tight ring while the resolution decreases towards the C-terminus, still allowing the fit of the predicted structure. The N-terminal domain is partially present in papillomavirus E1 and polyomavirus LTA helicases, as well as in a bacteriophage NrS-1 helicase domain, which is also closely related to the AAA+ helicase domain of D5. Using the Pfam domain database, a D5_N domain followed by DUF5906 and Pox_D5 domains could be assigned to the cryo-EM structure, providing the first 3D structures for D5_N and Pox_D5 domains. The same domain organization has been identified in a family of putative helicases from large DNA viruses, bacteriophages, and selfish DNA elements.


Assuntos
DNA Primase , Vírus Vaccinia , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , Microscopia Crioeletrônica , Vírus Vaccinia/genética , DNA Helicases/genética , DNA , Replicação do DNA , Nucleotídeos
9.
Nature ; 608(7924): 826-832, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35830881

RESUMO

The mammalian DNA polymerase-α-primase (Polα-primase) complex is essential for DNA metabolism, providing the de novo RNA-DNA primer for several DNA replication pathways1-4 such as lagging-strand synthesis and telomere C-strand fill-in. The physical mechanism underlying how Polα-primase, alone or in partnership with accessory proteins, performs its complicated multistep primer synthesis function is unknown. Here we show that CST, a single-stranded DNA-binding accessory protein complex for Polα-primase, physically organizes the enzyme for efficient primer synthesis. Cryogenic electron microscopy structures of the CST-Polα-primase preinitiation complex (PIC) bound to various types of telomere overhang reveal that template-bound CST partitions the DNA and RNA catalytic centres of Polα-primase into two separate domains and effectively arranges them in RNA-DNA synthesis order. The architecture of the PIC provides a single solution for the multiple structural requirements for the synthesis of RNA-DNA primers by Polα-primase. Several insights into the template-binding specificity of CST, template requirement for assembly of the CST-Polα-primase PIC and activation are also revealed in this study.


Assuntos
DNA Primase , Complexo Shelterina , Telômero , Moldes Genéticos , DNA/metabolismo , DNA Primase/química , DNA Primase/metabolismo , Primers do DNA/biossíntese , Replicação do DNA , Humanos , Domínios Proteicos , RNA/biossíntese , RNA/metabolismo , Complexo Shelterina/química , Complexo Shelterina/metabolismo , Especificidade por Substrato , Telômero/química , Telômero/genética , Telômero/metabolismo
10.
Nature ; 608(7924): 813-818, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35831498

RESUMO

Telomeres are the physical ends of linear chromosomes. They are composed of short repeating sequences (such as TTGGGG in the G-strand for Tetrahymena thermophila) of double-stranded DNA with a single-strand 3' overhang of the G-strand and, in humans, the six shelterin proteins: TPP1, POT1, TRF1, TRF2, RAP1 and TIN21,2. TPP1 and POT1 associate with the 3' overhang, with POT1 binding the G-strand3 and TPP1 (in complex with TIN24) recruiting telomerase via interaction with telomerase reverse transcriptase5 (TERT). The telomere DNA ends are replicated and maintained by telomerase6, for the G-strand, and subsequently DNA polymerase α-primase7,8 (PolαPrim), for the C-strand9. PolαPrim activity is stimulated by the heterotrimeric complex CTC1-STN1-TEN110-12 (CST), but the structural basis of the recruitment of PolαPrim and CST to telomere ends remains unknown. Here we report cryo-electron microscopy (cryo-EM) structures of Tetrahymena CST in the context of the telomerase holoenzyme, in both the absence and the presence of PolαPrim, and of PolαPrim alone. Tetrahymena Ctc1 binds telomerase subunit p50, a TPP1 orthologue, on a flexible Ctc1 binding motif revealed by cryo-EM and NMR spectroscopy. The PolαPrim polymerase subunit POLA1 binds Ctc1 and Stn1, and its interface with Ctc1 forms an entry port for G-strand DNA to the POLA1 active site. We thus provide a snapshot of four key components that are required for telomeric DNA synthesis in a single active complex-telomerase-core ribonucleoprotein, p50, CST and PolαPrim-that provides insights into the recruitment of CST and PolαPrim and the handoff between G-strand and C-strand synthesis.


Assuntos
DNA Primase , Complexo Shelterina , Telomerase , Tetrahymena , Microscopia Crioeletrônica , DNA/genética , DNA/metabolismo , DNA Primase/química , DNA Primase/metabolismo , DNA Primase/ultraestrutura , Holoenzimas/química , Holoenzimas/metabolismo , Holoenzimas/ultraestrutura , Ligação Proteica , Complexo Shelterina/química , Complexo Shelterina/metabolismo , Complexo Shelterina/ultraestrutura , Telomerase/química , Telomerase/metabolismo , Telomerase/ultraestrutura , Telômero/genética , Telômero/metabolismo , Tetrahymena/química , Tetrahymena/enzimologia , Tetrahymena/metabolismo , Tetrahymena/ultraestrutura
11.
J Biol Chem ; 298(6): 101996, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35500649

RESUMO

The T7 primase-helicase plays a pivotal role in the replication of T7 DNA. Using affinity isolation of peptide-nucleic acid crosslinks and mass spectrometry, we identify protein regions in the primase-helicase and T7 DNA polymerase that form contacts with the RNA primer and DNA template. The contacts between nucleic acids and the primase domain of the primase-helicase are centered in the RNA polymerase subdomain of the primase domain, in a cleft between the N-terminal subdomain and the topoisomerase-primase fold. We demonstrate that residues along a beta sheet in the N-terminal subdomain that contacts the RNA primer are essential for phage growth and primase activity in vitro. Surprisingly, we found mutations in the primase domain that had a dramatic effect on the helicase. Substitution of a residue conserved in other DnaG-like enzymes, R84A, abrogates both primase and helicase enzymatic activities of the T7 primase-helicase. Alterations in this residue also decrease binding of the primase-helicase to ssDNA. However, mass photometry measurements show that these mutations do not interfere with the ability of the protein to form the active hexamer.


Assuntos
Bacteriófago T7 , DNA Helicases , DNA Primase , DNA , Proteínas Virais , Sequência de Aminoácidos , Bacteriófago T7/enzimologia , DNA/metabolismo , DNA Helicases/química , DNA Helicases/metabolismo , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , Mutação , Proteínas Virais/química , Proteínas Virais/metabolismo
12.
Biomolecules ; 12(2)2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35204749

RESUMO

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.


Assuntos
Enzimas Multifuncionais , Neoplasias , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , Reparo do DNA , Replicação do DNA , DNA Polimerase Dirigida por DNA/química , Humanos , Enzimas Multifuncionais/química , Enzimas Multifuncionais/genética , Enzimas Multifuncionais/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética
13.
J Mol Biol ; 434(2): 167401, 2022 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-34902429

RESUMO

The ParABS system is supposed to be responsible for plasmid partitioning and chromosome segregation in bacteria. ParABS ensures a high degree of fidelity in inheritance by dividing the genetic material equally between daughter cells during cell division. However, the molecular mechanisms underlying the assembly of the partition complex, representing the core of the ParABS system, are still far from being understood. Here we demonstrate that the partition complex is formed via liquid-liquid phase separation. Assembly of the partition complex is initiated by the formation of oligomeric ParB species, which in turn are regulated by CTP-binding. Phase diagrams and in vivo analysis show how the partition complex can further be spatially regulated by parS. By investigating the phylogenetic variation in phase separation and its regulation by CTP, we find a high degree of evolutionary conservation among distantly related prokaryotes. These results advance the understanding of partition complex formation and regulation in general, by confirming and extending recently proposed models.


Assuntos
Citidina Trifosfato/química , Citidina Trifosfato/metabolismo , DNA Primase/química , DNA Primase/metabolismo , Bactérias/metabolismo , Fenômenos Fisiológicos Bacterianos , Proteínas de Bactérias/metabolismo , Divisão Celular , Segregação de Cromossomos , Cromossomos Bacterianos , Corynebacterium glutamicum/metabolismo , DNA Primase/genética , DNA Primase/isolamento & purificação , DNA Bacteriano , Transição de Fase , Filogenia
14.
Phys Rev Lett ; 127(13): 138101, 2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34623846

RESUMO

The spatiotemporal organization of bacterial cells is crucial for the active segregation of replicating chromosomes. In several species, including Caulobacter crescentus, the ATPase ParA binds to DNA and forms a gradient along the long cell axis. The ParB partition complex on the newly replicated chromosome translocates up this ParA gradient, thereby contributing to chromosome segregation. A DNA-relay mechanism-deriving from the elasticity of the fluctuating chromosome-has been proposed as the driving force for this cargo translocation, but a mechanistic theoretical description remains elusive. Here, we propose a minimal model to describe force generation by the DNA-relay mechanism over a broad range of operational conditions. Conceptually, we identify four distinct force-generation regimes characterized by their dependence on chromosome fluctuations. These relay force regimes arise from an interplay of the imposed ParA gradient, chromosome fluctuations, and an emergent friction force due to chromosome-cargo interactions.


Assuntos
DNA Bacteriano/metabolismo , Modelos Biológicos , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Transporte Biológico , Caulobacter crescentus/genética , Caulobacter crescentus/metabolismo , Segregação de Cromossomos , Cromossomos Bacterianos , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , DNA Bacteriano/química , DNA Bacteriano/genética
15.
Nucleic Acids Res ; 49(20): 11447-11458, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34718733

RESUMO

DNA-protein interactions play essential roles in all living cells. Understanding of how features embedded in the DNA sequence affect specific interactions with proteins is both challenging and important, since it may contribute to finding the means to regulate metabolic pathways involving DNA-protein interactions. Using a massive experimental benchmark dataset of binding scores for DNA sequences and a machine learning workflow, we describe the binding to DNA of T7 primase, as a model system for specific DNA-protein interactions. Effective binding of T7 primase to its specific DNA recognition sequences triggers the formation of RNA primers that serve as Okazaki fragment start sites during DNA replication.


Assuntos
DNA Primase/química , DNA/química , Motivos de Nucleotídeos , Sítios de Ligação , DNA/metabolismo , DNA Primase/metabolismo , Aprendizado de Máquina , Ligação Proteica
16.
Sci Rep ; 11(1): 17588, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34475447

RESUMO

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.


Assuntos
Cisplatino/química , Dano ao DNA , DNA Primase/química , Replicação do DNA , DNA Polimerase Dirigida por DNA/química , DNA/química , Enzimas Multifuncionais/química , Cisplatino/farmacologia , Reagentes de Ligações Cruzadas/química , Reparo do DNA , Humanos
17.
Chem Biol Drug Des ; 98(5): 722-732, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34265158

RESUMO

Mycobacterium tuberculosis (Mtb) is a pathogenic bacterium that caused 1.5 million fatalities globally in 2018. New strains of Mtb resistant to all known classes of antibiotics pose a global healthcare problem. In this work, we have conjugated novel indole-3-acetic acid-based DNA primase/gyrase inhibitor with cell-penetrating peptide via cleavable and non-cleavable bonds. For non-cleavable linkage, inhibitor was conjugated with peptide via an amide bond to the N-terminus, whereas a cleavable linkage was obtained by conjugating the inhibitor through a disulfide bond. We performed the conjugation of the inhibitor either directly on a solid surface or by using solution-phase chemistry. M. smegmatis (non-pathogenic model of Mtb) was used to determine the minimal inhibitory concentration (MIC) of the synthetic conjugates. Conjugates were found more active as compared to free inhibitor molecules. Strikingly, the conjugate also impairs the development of biofilm, showing a therapeutic potential against infections caused by both planktonic and sessile forms of mycobacterium species.


Assuntos
Antituberculosos/química , Peptídeos Penetradores de Células/química , DNA Primase/química , Ácidos Indolacéticos/química , Inibidores da Topoisomerase II/química , Antituberculosos/farmacologia , Biofilmes , DNA Primase/metabolismo , Testes de Sensibilidade Microbiana , Mycobacterium smegmatis/efeitos dos fármacos , Plâncton , Inibidores da Topoisomerase II/metabolismo
18.
Open Biol ; 11(6): 210011, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34102080

RESUMO

Herpes simplex virus type 1 (HSV-1) is one of the nine herpesviruses that infect humans. HSV-1 encodes seven proteins to replicate its genome in the hijacked human cell. Among these are the herpes virus DNA helicase and primase that are essential components of its replication machinery. In the HSV-1 replisome, the helicase-primase complex is composed of three components including UL5 (helicase), UL52 (primase) and UL8 (non-catalytic subunit). UL5 and UL52 subunits are functionally interdependent, and the UL8 component is required for the coordination of UL5 and UL52 activities proceeding in opposite directions with respect to the viral replication fork. Anti-viral compounds currently under development target the functions of UL5 and UL52. Here, we review the structural and functional properties of the UL5/UL8/UL52 complex and highlight the gaps in knowledge to be filled to facilitate molecular characterization of the structure and function of the helicase-primase complex for development of alternative anti-viral treatments.


Assuntos
DNA Helicases/química , DNA Helicases/metabolismo , DNA Primase/química , DNA Primase/metabolismo , Herpesvirus Humano 1/enzimologia , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Animais , Antivirais/farmacologia , DNA Helicases/genética , DNA Primase/genética , Desenvolvimento de Medicamentos , Herpes Simples/tratamento farmacológico , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Humanos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Relação Estrutura-Atividade , Replicação Viral/efeitos dos fármacos
19.
Nucleic Acids Res ; 49(8): 4599-4612, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33849056

RESUMO

The eukaryotic replisome is comprised of three family-B DNA polymerases (Polα, δ and ϵ). Polα forms a stable complex with primase to synthesize short RNA-DNA primers, which are subsequently elongated by Polδ and Polϵ in concert with proliferating cell nuclear antigen (PCNA). In some species of archaea, family-D DNA polymerase (PolD) is the only DNA polymerase essential for cell viability, raising the question of how it alone conducts the bulk of DNA synthesis. We used a hyperthermophilic archaeon, Thermococcus kodakarensis, to demonstrate that PolD connects primase to the archaeal replisome before interacting with PCNA. Whereas PolD stably connects primase to GINS, a component of CMG helicase, cryo-EM analysis indicated a highly flexible PolD-primase complex. A conserved hydrophobic motif at the C-terminus of the DP2 subunit of PolD, a PIP (PCNA-Interacting Peptide) motif, was critical for the interaction with primase. The dissociation of primase was induced by DNA-dependent binding of PCNA to PolD. Point mutations in the alternative PIP-motif of DP2 abrogated the molecular switching that converts the archaeal replicase from de novo to processive synthesis mode.


Assuntos
Proteínas Arqueais/metabolismo , DNA Helicases/metabolismo , DNA Polimerase III/metabolismo , DNA Primase/química , Antígeno Nuclear de Célula em Proliferação/metabolismo , Thermococcus/metabolismo , Motivos de Aminoácidos , Proteínas Arqueais/química , Cromatografia em Gel , DNA Helicases/genética , DNA Polimerase III/química , DNA Primase/genética , DNA Primase/metabolismo , Escherichia coli/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Mutagênese Sítio-Dirigida , Eletroforese em Gel de Poliacrilamida Nativa , Antígeno Nuclear de Célula em Proliferação/genética , Ligação Proteica , Proteínas Recombinantes , Ressonância de Plasmônio de Superfície , Thermococcus/genética
20.
Nat Commun ; 12(1): 2420, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33893297

RESUMO

Bacteriophages have long been known to use modified bases in their DNA to prevent cleavage by the host's restriction endonucleases. Among them, cyanophage S-2L is unique because its genome has all its adenines (A) systematically replaced by 2-aminoadenines (Z). Here, we identify a member of the PrimPol family as the sole possible polymerase of S-2L and we find it can incorporate both A and Z in front of a T. Its crystal structure at 1.5 Å resolution confirms that there is no structural element in the active site that could lead to the rejection of A in front of T. To resolve this contradiction, we show that a nearby gene is a triphosphohydolase specific of dATP (DatZ), that leaves intact all other dNTPs, including dZTP. This explains the absence of A in S-2L genome. Crystal structures of DatZ with various ligands, including one at sub-angstrom resolution, allow to describe its mechanism as a typical two-metal-ion mechanism and to set the stage for its engineering.


Assuntos
2-Aminopurina/análogos & derivados , Adenina/química , Bacteriófagos/genética , Cianobactérias/virologia , DNA Viral/química , Synechococcus/virologia , 2-Aminopurina/química , 2-Aminopurina/metabolismo , Adenina/metabolismo , Bacteriófagos/metabolismo , Sítios de Ligação/genética , Biocatálise , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Ligação de Hidrogênio , Modelos Moleculares , Estrutura Molecular , Domínios Proteicos , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...