Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Biochem Pharmacol ; 222: 116051, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38354956

RESUMO

Holliday junction (HJ) is a four-way structured DNA intermediate in processes of homologous recombination and DNA double-stranded break (DSB) repair. In bacteria, HJs are processed via either the RuvABC or RecG-dependent pathways. In addition, RecG also plays a critical role in the reactivation of stalled replication forks, making it an attractive target for antibacterial drug development. Here, we conducted a high-throughput screening targeting the RecG helicase from a common opportunistic pathogen Pseudomonas aeruginosa (Pa). From a library containing 7920 compounds, we identified Ebselen and TPI-1 (2',5'-Dichloro-[1,1'-biphenyl]-2,5-dione) as two potent PaRecG inhibitors, with IC50 values of 0.31 ± 0.02 µM and 1.16 ± 0.06 µM, respectively. Further biochemical analyses suggested that both Ebselen and TPI-1 inhibited the ATPase activity of PaRecG, and hindered its binding to HJ DNA with high selectivity. These compounds, when combined with our previously reported RuvAB inhibitors, resulted in more severe DNA repair defects than the individual treatment, and potently enhanced the susceptibility of P. aeruginosa to the DNA damage agents. This work reports novel small molecule inhibitors of RecG, offering valuable chemical tools for advancing our understanding of RecG's function and mechanism. Additionally, these inhibitors might be further developed as promising antibacterial agents in the fight against P. aeruginosa infections.


Assuntos
Proteínas de Escherichia coli , Isoindóis , Compostos Organosselênicos , Pseudomonas aeruginosa , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Bactérias , DNA Helicases/metabolismo , Reparo do DNA , Dano ao DNA , DNA Cruciforme , Replicação do DNA
2.
EMBO J ; 43(2): 304-315, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38177499

RESUMO

Type III CRISPR systems are innate immune systems found in bacteria and archaea, which produce cyclic oligoadenylate (cOA) second messengers in response to viral infections. In these systems, Csm6 proteins serve as ancillary nucleases that degrade single-stranded RNA (ssRNA) upon activation by cOA. In addition, Csm6 proteins also possess cOA-degrading activity as an intrinsic off-switch to avoid degradation of host RNA and DNA that would eventually lead to cell dormancy or cell death. Here, we present the crystal structures of Thermus thermophilus (Tt) Csm6 alone, and in complex with cyclic tetra-adenylate (cA4) in both pre- and post-cleavage states. These structures establish the molecular basis of the long-range allosteric activation of TtCsm6 ribonuclease by cA4. cA4 binding induces significant conformational changes, including closure of the CARF domain, dimerization of the HTH domain, and reorganization of the R-X4-6-H motif within the HEPN domain. The cleavage of cA4 by the CARF domain restores each domain to a conformation similar to its apo state. Furthermore, we have identified hyperactive TtCsm6 variants that exhibit sustained cA4-activated RNase activity, showing great promise for their applications in genome editing and diagnostics.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Nucleotídeos Cíclicos , Ribonucleases , Ribonucleases/metabolismo , Regulação Alostérica , RNA/metabolismo
3.
Bioorg Med Chem ; 90: 117383, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37352577

RESUMO

The MUS81-EME1/2 structure-specific endonucleases play a crucial role in the processing of stalled replication forks and recombination intermediates, and have been recognized as an attractive drug target to potentiate the anti-cancer efficacy of DNA-damaging agents. Currently, no bioactive small-molecule inhibitors of MUS81 are available. Here, we performed a high-throughput small-molecule inhibitors screening, using the FRET-based DNA cleavage assay. From 7920 compounds, we identified dyngo-4a as a potent inhibitor of MUS81 complexes. Dyngo-4a effectively inhibits the endonuclease activities of both MUS81-EME1 and MUS81-EME2 complexes, with IC50 values of 0.57 µM and 2.90 µM, respectively. Surface plasmon resonance (SPR) and electrophoretic mobility shift assay (EMSA) assays reveal that dyngo-4a directly binds to MUS81 complexes (KD âˆ¼ 0.61 µM) and prevents them from binding to DNA substrates. In HeLa cells, dyngo-4a significantly suppresses bleomycin-triggered H2AX serine 139 phosphorylation (γH2AX). Together, our results demonstrate that dyngo-4a is a potent MUS81 inhibitor, which could be further developed as a potentially valuable chemical tool to explore more physiological roles of MUS81 in the cells.


Assuntos
Proteínas de Ligação a DNA , Endodesoxirribonucleases , Humanos , Endodesoxirribonucleases/química , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Proteínas de Ligação a DNA/química , Células HeLa , Transferência Ressonante de Energia de Fluorescência , Ensaios de Triagem em Larga Escala , Endonucleases/metabolismo , Replicação do DNA , DNA/metabolismo
4.
J Clin Microbiol ; 61(6): e0188422, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37195177

RESUMO

The quantitative detection of drug-resistance mutations in Mycobacterium tuberculosis (MTB) is critical for determining the drug resistance status of a sample. We developed a drop-off droplet digital PCR (ddPCR) assay targeting all major isoniazid (INH)-resistant mutations. The ddPCR assay consisted of three reactions: reaction A detects mutations at katG S315; reaction B detects inhA promoter mutations; and reaction C detects ahpC promoter mutations. All reactions could quantify 1%-50% of mutants in the presence of the wild-type, ranging from 100 to 50,000 copies/reaction. Clinical evaluation with 338 clinical isolates yielded clinical sensitivity of 94.5% (95% confidence interval [CI] = 89.1%-97.3%) and clinical specificity of 97.6% (95% CI = 94.6%-99.0%) compared with the traditional drug susceptibility testing (DST). Further clinical evaluation using 194 nucleic acid-positive MTB sputum samples revealed clinical sensitivity of 87.8% (95% CI = 75.8%-94.3%) and clinical specificity of 96.5% (95% CI = 92.2%-98.5%) in comparison with DST. All the mutant and heteroresistant samples detected by the ddPCR assay but susceptible by DST were confirmed by combined molecular assays, including Sanger sequencing, mutant-enriched Sanger sequencing and a commercial melting curve analysis-based assay. Finally, the ddPCR assay was used to monitor longitudinally the INH-resistance status and the bacterial load in nine patients undergoing treatment. Overall, the developed ddPCR assay could be an indispensable tool for quantification of INH-resistant mutations in MTB and bacterial loads in patients.


Assuntos
Mycobacterium tuberculosis , Tuberculose Resistente a Múltiplos Medicamentos , Humanos , Isoniazida/farmacologia , Mycobacterium tuberculosis/genética , Antituberculosos/farmacologia , Testes de Sensibilidade Microbiana , Sensibilidade e Especificidade , Reação em Cadeia da Polimerase , Mutação , Tuberculose Resistente a Múltiplos Medicamentos/microbiologia , Proteínas de Bactérias/genética
5.
Front Plant Sci ; 14: 1139106, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37025142

RESUMO

Holliday junction (HJ) is a four-way structured DNA intermediate in homologous recombination. In bacteria, the HJ-specific binding protein RuvA and the motor protein RuvB together form the RuvAB complex to catalyze HJ branch migration. Pseudomonas aeruginosa (P. aeruginosa, Pa) is a ubiquitous opportunistic bacterial pathogen that can cause serious infection in a variety of host species, including vertebrate animals, insects and plants. Here, we describe the cryo-Electron Microscopy (cryo-EM) structure of the RuvAB-HJ intermediate complex from P. aeruginosa. The structure shows that two RuvA tetramers sandwich HJ at the junction center and disrupt base pairs at the branch points of RuvB-free HJ arms. Eight RuvB subunits are recruited by the RuvA octameric core and form two open-rings to encircle two opposite HJ arms. Each RuvB subunit individually binds a RuvA domain III. The four RuvB subunits within the ring display distinct subdomain conformations, and two of them engage the central DNA duplex at both strands with their C-terminal ß-hairpins. Together with the biochemical analyses, our structure implicates a potential mechanism of RuvB motor assembly onto HJ DNA.

6.
Nucleic Acids Res ; 51(5): 2485-2495, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36807980

RESUMO

The cyclic oligoadenylates (cOAs) act as second messengers of the type III CRISPR immunity system through activating the auxiliary nucleases for indiscriminate RNA degradation. The cOA-degrading nucleases (ring nucleases) provide an 'off-switch' regulation of the signaling, thereby preventing cell dormancy or cell death. Here, we describe the crystal structures of the founding member of CRISPR-associated ring nuclease 1 (Crn1) Sso2081 from Saccharolobus solfataricus, alone, bound to phosphate ions or cA4 in both pre-cleavage and cleavage intermediate states. These structures together with biochemical characterizations establish the molecular basis of cA4 recognition and catalysis by Sso2081. The conformational changes in the C-terminal helical insert upon the binding of phosphate ions or cA4 reveal a gate-locking mechanism for ligand binding. The critical residues and motifs identified in this study provide a new insight to distinguish between cOA-degrading and -nondegrading CARF domain-containing proteins.


Assuntos
Proteínas Associadas a CRISPR , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Sistemas do Segundo Mensageiro , Transdução de Sinais , Endonucleases/metabolismo , Íons/metabolismo , Sistemas CRISPR-Cas , Proteínas Associadas a CRISPR/metabolismo
7.
Bioorg Med Chem ; 73: 117022, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36155320

RESUMO

The Holliday junction (HJ) branch migrator RuvAB complex plays a fundamental role during homologous recombination and DNA damage repair, and therefore, is an attractive target for the treatment of bacterial pathogens. Pseudomonas aeruginosa (P. aeruginosa, Pa) is one of the most common clinical opportunistic bacterial pathogens, which can cause a series of life-threatening acute or chronic infections. Here, we performed a high throughput small-molecule screening targeting PaRuvAB using the FRET-based HJ branch migration assay. We identified that corilagin, bardoxolone methyl (BM) and 10-(6'-plastoquinonyl) decyltriphenylphosphonium (SKQ1) could efficiently inhibit the branch migration activity of PaRuvAB, with IC50 values of 0.40 ± 0.04 µM, 0.38 ± 0.05 µM and 4.64 ± 0.27 µM, respectively. Further biochemical and molecular docking analyses demonstrated that corilagin directly bound to PaRuvB at the ATPase domain, and thus prevented ATP hydrolysis. In contrast, BM and SKQ1 acted through blocking the interactions between PaRuvA and HJ DNA. Finally, these compounds were shown to increase the susceptibility of P. aeruginosa to UV-C irradiation. Our work, for the first time, reports the small-molecule inhibitors of RuvA and RuvB from any species, providing valuable chemical tools to dissect the functional role of each individual protein in vivo.


Assuntos
Proteínas de Escherichia coli , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , DNA Helicases , Reparo do DNA , DNA Bacteriano , DNA Cruciforme/metabolismo , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Glucosídeos , Taninos Hidrolisáveis , Simulação de Acoplamento Molecular , Ácido Oleanólico/análogos & derivados , Pseudomonas aeruginosa/metabolismo , Recombinação Genética
8.
Eur J Med Chem ; 243: 114749, 2022 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-36115207

RESUMO

Herein, we fabricate a multifunctional molecular prodrug BAC where the chemotherapeutical agent camptothecin (CPT) is linked with a boron dipyrromethene (BODIPY)-based photosensitizer by an azobenzene chain which is sensitive to over-expressed azoreductase in hypoxic tumor cells. This prodrug was further loaded into biodegradable monomethoxy poly(ethylene glycol)-b-poly(caprolactone) (mPEG-b-PCL) to improve its solubility and tumor accumulation. The formed BAC nanoparticles (BAC NPs) can destroy aerobic tumor cells with relatively short distance from blood vessels by photodynamic therapy (PDT) under illumination. The PDT action inevitably leads to consumption of O2, and subsequently acute hypoxia which can induce cleavage of azobenzene linkage to boost release of CPT killing the other hypoxic interior tumor cells survived from PDT. Both in vitro and in vivo studies have verified that BAC NPs possess remarkable antitumor activity by a synergistic action of PDT and chemotherapy.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Pró-Fármacos , Humanos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Neoplasias/tratamento farmacológico , Camptotecina/uso terapêutico , Hipóxia/tratamento farmacológico , Linhagem Celular Tumoral
9.
ACS Appl Mater Interfaces ; 14(36): 40546-40558, 2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36059107

RESUMO

Hypoxia-activated prodrugs (HAPs) have drawn increasing attention for improving the antitumor effects while minimizing side effects. However, the heterogeneous distribution of the hypoxic region in tumors severely impedes the curative effect of HAPs. Additionally, most HAPs are not amenable to optical imaging, and it is difficult to precisely trace them in tissues. Herein, we carefully designed and synthesized a multifunctional therapeutic BAC prodrug by connecting the chemotherapeutic drug camptothecin (CPT) and the fluorescent photothermal agent boron dipyrromethene (BODIPY) via hypoxia-responsive azobenzene linkers. To enhance the solubility and tumor accumulation, the prepared BAC was further encapsulated into a human serum albumin (HSA)-based drug delivery system to form HSA@BAC nanoparticles. Since the CPT was caged by a BODIPY-based molecule at the active site, the BAC exhibited excellent biosafety. Importantly, the activated CPT could be quickly released from BAC and could perform chemotherapy in hypoxic cancer cells, which was ascribed to the cleavage of the azobenzene linker by overexpressed azoreductase. After irradiation with a 730 nm laser, HSA@BAC can efficiently generate hyperthermia to achieve irreversible cancer cell death by oxygen-independent photothermal therapy. Under fluorescence imaging-guided local irradiation, both in vitro and in vivo studies demonstrated that HSA@BAC exhibited superior antitumor effects with minimal side effects.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Pró-Fármacos , Compostos Azo , Boro , Compostos de Boro , Camptotecina/química , Linhagem Celular Tumoral , Humanos , Hipóxia , Nanopartículas/química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Fototerapia , Terapia Fototérmica , Porfobilinogênio/análogos & derivados , Pró-Fármacos/química
10.
Antiviral Res ; 206: 105389, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35985407

RESUMO

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) helicase NSP13 plays a conserved role in the replication of coronaviruses and has been identified as an ideal target for the development of antiviral drugs against SARS-CoV-2. Here, we identify a novel NSP13 helicase inhibitor punicalagin (PUG) through high-throughput screening. Surface plasmon resonance (SPR)-based analysis and molecular docking calculation reveal that PUG directly binds NSP13 on the interface of domains 1A and 2A, with a KD value of 21.6 nM. Further biochemical and structural analyses suggest that PUG inhibits NSP13 on ATP hydrolysis and prevents it binding to DNA substrates. Finally, the antiviral studies show that PUG effectively suppresses the SARS-CoV-2 replication in A549-ACE2 and Vero cells, with EC50 values of 347 nM and 196 nM, respectively. Our work demonstrates the potential application of PUG in the treatment of coronavirus disease 2019 (COVID-19) and identifies an allosteric inhibition mechanism for future drug design targeting the viral helicases.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Animais , Antivirais/química , Antivirais/farmacologia , Chlorocebus aethiops , DNA Helicases/metabolismo , Humanos , Taninos Hidrolisáveis , Simulação de Acoplamento Molecular , RNA Helicases/química , Células Vero
11.
STAR Protoc ; 3(3): 101528, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-35819885

RESUMO

The human MUS81-EME1&2 complexes are structure-selective endonucleases that play important roles in DNA damage repair. Here, we describe a protocol to determine the endonuclease activities of MUS81-EME1&2 complexes toward various DNA structures. We co-express MUS81 with EME1 or EME2 and purify the complexes with high purity, and determine their activities on the cleavages of 3' flaps, 5' flaps, nicked double-stranded DNAs, and Holliday junctions. This protocol can also be used for the determination of substrate preferences of other structure-selective endonucleases. For complete details on the use and execution of this protocol, please refer to Hua et al. (2022).


Assuntos
DNA Cruciforme , Endonucleases , DNA/química , Proteínas de Ligação a DNA/genética , Endodesoxirribonucleases/química , Endonucleases/química , Humanos , Especificidade por Substrato
12.
Structure ; 30(5): 743-752.e3, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35290797

RESUMO

MUS81 is an important structure-specific endonuclease responsible for the processing of stalled replication forks and recombination intermediates. In human, MUS81 functions by forming complexes with its regulatory subunits EME1 and EME2, playing distinct roles in G2/M and S phases. Although the structures of MUS81-EME1 have been intensively studied, there is no structure information available about MUS81-EME2. Here, we report the crystal structure of MUS81-EME2, which reveals an overall protein fold similar to that of MUS81-EME1 complex. Further biochemical and structural characterization shows that the MUS81-EME1 and MUS81-EME2 complexes are identical in substrate recognition and endonuclease activities in vitro, implying that the distinct cellular roles of the two complexes could arise from temporal controls in cells. Finally, an extensive structure-guided mutagenesis analysis provides implications for the molecular basis of how the MUS81-EME endonucleases recognize various DNA substrates in a structure-selective manner.


Assuntos
Proteínas de Ligação a DNA , Endodesoxirribonucleases , Replicação do DNA , Proteínas de Ligação a DNA/química , Endodesoxirribonucleases/química , Endonucleases/química , Humanos , Especificidade por Substrato
13.
J Enzyme Inhib Med Chem ; 37(1): 109-117, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34894976

RESUMO

Invasive fungal infections including Candidiasis and Aspergillosis are associated with considerable morbidity and mortality in immunocompromised individuals, such as cancer patients. Aurora B is a key mitotic kinase required for the cell division of eukaryotes from fungus to man. Here, we identified a novel Aurora B inhibitor GSK650394 that can inhibit the recombinant Aurora B from human and Aspergillus fumigatus, with IC50 values of 5.68 and 1.29 µM, respectively. In HeLa and HepG2 cells, GSK650394 diminishes the endogenous Aurora B activity and causes cell cycle arrest in G2/M phase. Further cell-based assays demonstrate that GSK650394 efficiently suppresses the proliferation of both cancer cells and Aspergillus fumigatus. Finally, the molecular docking calculation and site-directed mutagenesis analyses reveal the molecular mechanism of Aurora B inhibition by GSK650394. Our work is expected to provide new insight into the combinational therapy of cancer and Aspergillus fumigatus infection.


Assuntos
Antifúngicos/farmacologia , Antineoplásicos/farmacologia , Aspergillus fumigatus/efeitos dos fármacos , Aurora Quinase B/antagonistas & inibidores , Benzoatos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Descoberta de Drogas , Antifúngicos/química , Antineoplásicos/química , Aurora Quinase B/metabolismo , Benzoatos/química , Compostos Bicíclicos Heterocíclicos com Pontes/química , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade , Células Tumorais Cultivadas
14.
ACS Appl Mater Interfaces ; 13(29): 33905-33914, 2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34278780

RESUMO

Coencapsulation of chemotherapeutic agents and photosensitizers into nanocarriers can help to achieve a combination of chemotherapy and photodynamic therapy for superior antitumor effects. However, precise on-demand drug release remains a major challenge. In addition, the loaded photosensitizers usually tend to aggregate, which can significantly weaken their fluorescent signals and photodynamic activities. To address these issues, herein, a smart nanocarrier termed as singlet oxygen-responsive nanoparticle (SOR-NP) was constructed by introducing singlet oxygen (1O2)-sensitive aminoacrylate linkers into amphiphilic mPEG-b-PCL copolymers. Boron dipyrromethene (BDP) and paclitaxel (PTX) as model therapeutic agents were coloaded into an 1O2-responsive nanocarrier for realizing light-controlled drug release and combination cancer treatment. This polymeric nanocarrier could substantially relieve the aggregation of encapsulated BDP due to the presence of a long hydrophobic chain. Therefore, the formed SOR-NPBDP/PTX nanodrug could generate bright fluorescent signals and high levels of 1O2, which could mediate cell death via PDT and rupture aminoacrylate linker simultaneously, leading to collapse of SOR-NPBDP/PTX and subsequent PTX release. The light-triggered drug release and combined anticancer effects of SOR-NPBDP/PTX were validated in HepG2 and MCF-7 cancer cells and H22 tumor-bearing mice. This study provides a promising strategy for tumor-specific drug release and selective photodynamic-chemo combination treatment.


Assuntos
Antineoplásicos/uso terapêutico , Portadores de Fármacos/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Acrilatos/síntese química , Acrilatos/química , Animais , Antineoplásicos/química , Compostos de Boro/química , Compostos de Boro/uso terapêutico , Linhagem Celular Tumoral , Portadores de Fármacos/síntese química , Liberação Controlada de Fármacos , Feminino , Humanos , Camundongos , Paclitaxel/química , Paclitaxel/uso terapêutico , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Poliésteres/síntese química , Poliésteres/química , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Pirróis/química , Pirróis/uso terapêutico , Oxigênio Singlete/metabolismo
15.
Ying Yong Sheng Tai Xue Bao ; 32(7): 2415-2425, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34313059

RESUMO

An in-depth understanding of variations in grassland productivity and forage-livestock balance is the basis of ecological barrier construction and ecosystem conservation in the Qinghai-Tibetan Plateau. Using an ecohydrological process-based model VIP with remotely sensed vegetation index and leaf area index, we simulated the spatial and temporal variations of grassland productivity in the Tibetan Plateau in 2000-2018. The variations in the status of forage-livestock balance at the county level were analyzed, combining with agriculture and animal husbandry statistics in the same period. The results showed that the mean annual net primary productivity (NPP) of grassland in the Tibetan Plateau was 158.4 g C·m-2·a-1, which had increased significantly in the past 20 years, with a significant increase in 44.7% of the total area. Climate warming, increased precipitation, prolonged growing season, and elevated carbon dioxide concentration were main driving forces for grassland productivity. The mean theoretical livestock carrying capacity estimated based on pasture yield was 1.17 SU·hm-2, with a growth rate of 0.011 SU·hm-2. The situation of overgrazing in the Tibetan Plateau had generally improved since 2000. The proportion of counties with severe overgrazing had dropped to less than 20%. In areas with more severe overgrazing, animal husbandry's maintenance and development mainly relied on supplementation of crop straw. The results could provide a scientific basis for regional agricultural and animal husbandry structural adjustment and environmental protection.


Assuntos
Ecossistema , Pradaria , Animais , China , Mudança Climática , Gado , Tibet
16.
Biochem Biophys Res Commun ; 525(2): 265-271, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32085896

RESUMO

The Holliday junction, a four-way DNA structure, is an important intermediate of homologous recombination. Proper Holliday junction resolution is critical to complete the recombination process. In most bacterial cells, the Holliday junction cleavage is mainly performed by a specific endonuclease RuvC. Here, we describe the biochemical properties and the crystal structure of RuvC from an opportunistic pathogen, Pseudomonas aeruginosa (PaRuvC). PaRuvC specifically binds to the Holliday junction DNA and preferentially cleaves it at the consensus 5'-TTC-3'. PaRuvC uses Mg2+ as the preferred divalent metal cofactor for Holliday junction cleavage and its optimum pH is 8.0-9.0. Elevated temperatures (37-60 °C) boost the catalytic activity, but temperatures higher than 53 °C reduce the protein stability. The crystal structure of PaRuvC determined at 2.4 Å and mutagenesis analysis reveal key residues involved in the dimer formation, substrate binding and catalysis. Our results are expected to provide useful information to combat antibiotic resistance of Pseudomonas aeruginosa by targeting its homologous recombination system.


Assuntos
Cristalografia por Raios X/métodos , DNA Cruciforme/metabolismo , Resolvases de Junção Holliday/química , Pseudomonas aeruginosa/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/química , Biocatálise , Resolvases de Junção Holliday/genética , Resolvases de Junção Holliday/metabolismo , Recombinação Homóloga , Mutagênese , Multimerização Proteica , Estabilidade Proteica
17.
Nat Chem Biol ; 15(12): 1241-1248, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31611704

RESUMO

The Holliday junction (HJ) is a key intermediate during homologous recombination and DNA double-strand break repair. Timely HJ resolution by resolvases is critical for maintaining genome stability. The mechanisms underlying sequence-specific substrate recognition and cleavage by resolvases remain elusive. The monokaryotic chloroplast 1 protein (MOC1) specifically cleaves four-way DNA junctions in a sequence-specific manner. Here, we report the crystal structures of MOC1 from Zea mays, alone or bound to HJ DNA. MOC1 uses a unique ß-hairpin to embrace the DNA junction. A base-recognition motif specifically interacts with the junction center, inducing base flipping and pseudobase-pair formation at the strand-exchanging points. Structures of MOC1 bound to HJ and different metal ions support a two-metal ion catalysis mechanism. Further molecular dynamics simulations and biochemical analyses reveal a communication between specific substrate recognition and metal ion-dependent catalysis. Our study thus provides a mechanism for how a resolvase turns substrate specificity into catalytic efficiency.


Assuntos
Cloroplastos/metabolismo , Resolvases de Junção Holliday/metabolismo , Proteínas de Plantas/metabolismo , Resolvases de Junção Holliday/química , Simulação de Dinâmica Molecular , Conformação Proteica , Especificidade por Substrato
18.
Nat Commun ; 9(1): 3848, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30242288

RESUMO

Chromatin remodelers regulate the nucleosome barrier during transcription, DNA replication, and DNA repair. The chromatin remodeler RSF1 is enriched at mitotic centromeres, but the functional consequences of this enrichment are not completely understood. Shugoshin (Sgo1) protects centromeric cohesion during mitosis and requires BuB1-dependent histone H2A phosphorylation (H2A-pT120) for localization. Loss of Sgo1 at centromeres causes chromosome missegregation. Here, we show that RSF1 regulates Sgo1 localization to centromeres through coordinating a crosstalk between histone acetylation and phosphorylation. RSF1 interacts with and recruits HDAC1 to centromeres, where it counteracts TIP60-mediated acetylation of H2A at K118. This deacetylation is required for the accumulation of H2A-pT120 and Sgo1 deposition, as H2A-K118 acetylation suppresses H2A-T120 phosphorylation by Bub1. Centromeric tethering of HDAC1 prevents premature chromatid separation in RSF1 knockout cells. Our results indicate that RSF1 regulates the dynamics of H2A histone modifications at mitotic centromeres and contributes to the maintenance of chromosome stability.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Segregação de Cromossomos , Histona Desacetilase 1/metabolismo , Proteínas Nucleares/fisiologia , Transativadores/fisiologia , Acetilação , Instabilidade Cromossômica , Células HeLa , Código das Histonas , Humanos , Lisina Acetiltransferase 5/metabolismo , Fosforilação
19.
J Cell Mol Med ; 20(10): 1851-60, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27197780

RESUMO

Fibrinolysis is a process responsible for the dissolution of formed thrombi to re-establish blood flow after thrombus formation. Plasminogen activator inhibitor-1 (PAI-1) inhibits urokinase-type and tissue-type plasminogen activator (uPA and tPA) and is the major negative regulator of fibrinolysis. Inhibition of PAI-1 activity prevents thrombosis and accelerates fibrinolysis. However, a specific antagonist of PAI-1 is currently unavailable for therapeutic use. We screened a panel of uPA variants with mutations at and near the active site to maximize their binding to PAI-1 and identified a potent PAI-1 antagonist, PAItrap. PAItrap is the serine protease domain of urokinase containing active-site mutation (S195A) and four additional mutations (G37bR-R217L-C122A-N145Q). PAItrap inhibits human recombinant PAI-1 with high potency (Kd = 0.15 nM) and high specificity. In vitro using human plasma, PAItrap showed significant thrombolytic activity by inhibiting endogenous PAI-1. In addition, PAItrap inhibits both human and murine PAI-1, allowing the evaluation in murine models. In vivo, using a laser-induced thrombosis mouse model in which thrombus formation and fibrinolysis are monitored by intravital microscopy, PAItrap reduced fibrin generation and inhibited platelet accumulation following vascular injury. Therefore, this work demonstrates the feasibility to generate PAI-1 inhibitors using inactivated urokinase.


Assuntos
Fragmentos de Peptídeos/farmacologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Animais , Domínio Catalítico , Cristalografia por Raios X , Modelos Animais de Doenças , Fibrinólise , Humanos , Concentração Inibidora 50 , Cinética , Camundongos Endogâmicos C57BL , Modelos Moleculares , Proteínas Mutantes/química , Fragmentos de Peptídeos/química , Ligação Proteica , Trombose/patologia , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/farmacologia
20.
Nature ; 532(7597): 131-4, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27027290

RESUMO

Accurate chromosome segregation requires timely dissolution of chromosome cohesion after chromosomes are properly attached to the mitotic spindle. Separase is absolutely essential for cohesion dissolution in organisms from yeast to man. It cleaves the kleisin subunit of cohesin and opens the cohesin ring to allow chromosome segregation. Cohesin cleavage is spatiotemporally controlled by separase-associated regulatory proteins, including the inhibitory chaperone securin, and by phosphorylation of both the enzyme and substrates. Dysregulation of this process causes chromosome missegregation and aneuploidy, contributing to cancer and birth defects. Despite its essential functions, atomic structures of separase have not been determined. Here we report crystal structures of the separase protease domain from the thermophilic fungus Chaetomium thermophilum, alone or covalently bound to unphosphorylated and phosphorylated inhibitory peptides derived from a cohesin cleavage site. These structures reveal how separase recognizes cohesin and how cohesin phosphorylation by polo-like kinase 1 (Plk1) enhances cleavage. Consistent with a previous cellular study, mutating two securin residues in a conserved motif that partly matches the separase cleavage consensus converts securin from a separase inhibitor to a substrate. Our study establishes atomic mechanisms of substrate cleavage by separase and suggests competitive inhibition by securin.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Chaetomium/enzimologia , Proteínas Cromossômicas não Histona/metabolismo , Separase/química , Separase/metabolismo , Sequência de Aminoácidos , Ligação Competitiva/efeitos dos fármacos , Proteínas de Ciclo Celular/química , Proteínas Cromossômicas não Histona/química , Segregação de Cromossomos , Cristalografia por Raios X , Modelos Moleculares , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteólise , Proteínas Proto-Oncogênicas/metabolismo , Securina/química , Securina/genética , Securina/metabolismo , Securina/farmacologia , Separase/antagonistas & inibidores , Relação Estrutura-Atividade , Especificidade por Substrato/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...