Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Nat Commun ; 13(1): 359, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35042867

RESUMO

Single-stranded DNA (ssDNA) commonly occurs as intermediates in DNA metabolic pathways. The ssDNA binding protein, RPA, not only protects the integrity of ssDNA, but also directs the downstream factor that signals or repairs the ssDNA intermediate. However, it remains unclear how these enzymes/factors outcompete RPA to access ssDNA. Using the budding yeast Saccharomyces cerevisiae as a model system, we find that Dna2 - a key nuclease in DNA replication and repair - employs a bimodal interface to act with RPA both in cis and in trans. The cis-activity makes RPA a processive unit for Dna2-catalyzed ssDNA digestion, where RPA delivers its bound ssDNA to Dna2. On the other hand, activity in trans is mediated by an acidic patch on Dna2, which enables it to function with a sub-optimal amount of RPA, or to overcome DNA secondary structures. The trans-activity mode is not required for cell viability, but is necessary for effective double strand break (DSB) repair.


Assuntos
DNA Helicases/metabolismo , DNA Fúngico/metabolismo , DNA de Cadeia Simples/metabolismo , Proteína de Replicação A/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Biocatálise , Sobrevivência Celular/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Modelos Biológicos , Mutação/genética , Peptídeos/metabolismo , Fleomicinas/farmacologia , Ligação Proteica , Domínios Proteicos , Proteínas de Saccharomyces cerevisiae/química , Tirosina/metabolismo
2.
Curr Genet ; 65(6): 1297-1300, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31076845

RESUMO

We use genetic assays to suggest that transcription-coupled repair or new origin formation in Escherichia coli involves removal of RNAP to create an RNA primer for DNA synthesis. Transcription factor DksA was shown to play a role in numerous reactions involving RNA polymerase. Some, but not all, of the activities of DksA at promoters or during transcription elongation require (p)ppGpp. In addition to its role during transcription, DksA is also involved in maintaining genome integrity. Cells lacking DksA are sensitive to multiple DNA damaging agents including UV light, ionizing radiation, mitomycin C, and nalidixic acid. Here, we focus on two recent studies addressing the importance of DksA in the repair of double-strand breaks (DSBs), one by Sivaramakrishnan et al. (Nature 550:214-218, 2017) and one originating in our laboratory, Myka et al. (Mol Microbiol 111:1382-1397. https://doi.org/10.1111/mmi.14227 , 2019). It appears that depending on the type and possibly location of DNA damage, DksA can play either a passive or an active role in DSB repair. The passive role relies on exclusion of anti-backtracking factors from the RNAP secondary channel. The exact mechanism of active DksA-mediated DNA repair is unknown. However, DksA was proposed to destabilize transcription complexes, thus clearing the way for recombination and DNA repair. Based on the requirement for DksA, both in repair of DSBs and the R-loop-dependent formation of new origins of DNA replication, we propose that DksA may allow for removal of RNAP without unwinding of the RNA:DNA hybrid, which can then be extended by a DNA polymerase. This mechanism obviates the need for RNAP backtracking to repair damaged DNA.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Proteínas de Escherichia coli/genética , Guanosina Pentafosfato/metabolismo , Ácido Nalidíxico/farmacologia , Fleomicinas/farmacologia , Regiões Promotoras Genéticas , Fatores de Transcrição/genética
3.
Folia Microbiol (Praha) ; 64(1): 33-39, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29938299

RESUMO

Three different transformation strategies were tested and compared in an attempt to facilitate and improve the genetic transformation of Acremonium chrysogenum, the exclusive producer of the pharmaceutically relevant ß-lactam antibiotic cephalosporin C. We investigated the use of high-voltage electric pulse to transform germinated conidia and young mycelium and compared these procedures with traditional PEG-mediated protoplast transformation, using phleomycin resistance as selection marker in all cases. The effect of the field strength and capacitance on transformation frequency and cell viability was evaluated. The electroporation of germinated conidia and young mycelium was found to be appropriate for transforming A. chrysogenum with higher transformation efficiencies than those obtained with the conventional protoplast-based transformation procedures. The developed electroporation strategy is fast, simple to perform, and highly reproducible and avoids the use of chemicals toxic to cells. Electroporation of young mycelium represents an alternative method for transformation of fungal strains with reduced or no sporulation, as often occurs in laboratory-developed strains in the search for high-yielding mutants for industrial bioprocesses.


Assuntos
Acremonium/genética , Eletroporação/métodos , Transformação Genética , Acremonium/efeitos dos fármacos , Acremonium/metabolismo , Cefalosporinas/biossíntese , Farmacorresistência Bacteriana , Viabilidade Microbiana , Micélio/efeitos dos fármacos , Micélio/genética , Micélio/metabolismo , Fleomicinas/farmacologia , Protoplastos/fisiologia , Esporos Fúngicos/efeitos dos fármacos , Esporos Fúngicos/genética , Esporos Fúngicos/metabolismo
4.
Free Radic Biol Med ; 129: 97-106, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30223018

RESUMO

A genetic analysis of synthetic lethal interactions in yeast revealed that the mutation of SOD1, encoding an antioxidant enzyme that scavenges superoxide anion radical, impaired the growth of a set of mutants defective in homologous recombination (HR) pathway. Hence, SOD1 inhibition has been proposed as a promising approach for the selective killing of HR-deficient cancer cells. However, we show that the deletion of RAD51 and SOD1 is not synthetic lethal but displays considerably slow growth and synergistic sensitivity to both reactive oxygen species (ROS)- and DNA double-strand break (DSB)-generating drugs in the budding yeast Saccharomyces cerevisiae. The function of Sod1 in regard to Rad51 is dependent on Ccs1, a copper chaperone for Sod1. Sod1 deficiency aggravates genomic instability in conjunction with the absence of Rad51 by inducing DSBs and an elevated mutation frequency. Inversely, lack of Rad51 causes a Sod1 deficiency-derived increase of intracellular ROS levels. Taken together, our results indicate that there is a significant and specific crosstalk between two major cellular damage response pathways, ROS signaling and DSB repair, for cell survival.


Assuntos
DNA Fúngico/genética , Regulação Fúngica da Expressão Gênica , Genoma Fúngico , Instabilidade Genômica/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Superóxido Dismutase-1/genética , 4-Nitroquinolina-1-Óxido/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , DNA Fúngico/metabolismo , Recombinação Homóloga , Peróxido de Hidrogênio/farmacologia , Hidroxiureia/farmacologia , Metanossulfonato de Metila/farmacologia , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutação , Paraquat/farmacologia , Fleomicinas/farmacologia , Quinolonas/farmacologia , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Reparo de DNA por Recombinação/efeitos dos fármacos , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Superóxido Dismutase-1/deficiência
5.
Nucleic Acids Res ; 46(6): 2990-3008, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29420790

RESUMO

Sae2 cooperates with the Mre11-Rad50-Xrs2 (MRX) complex to initiate resection of DNA double-strand breaks (DSBs) and to maintain the DSB ends in close proximity to allow their repair. How these diverse MRX-Sae2 functions contribute to DNA damage resistance is not known. Here, we describe mre11 alleles that suppress the hypersensitivity of sae2Δ cells to genotoxic agents. By assessing the impact of these mutations at the cellular and structural levels, we found that all the mre11 alleles that restore sae2Δ resistance to both camptothecin and phleomycin affect the Mre11 N-terminus and suppress the resection defect of sae2Δ cells by lowering MRX and Tel1 association to DSBs. As a consequence, the diminished Tel1 persistence potentiates Sgs1-Dna2 resection activity by decreasing Rad9 association to DSBs. By contrast, the mre11 mutations restoring sae2Δ resistance only to phleomycin are located in Mre11 C-terminus and bypass Sae2 function in end-tethering but not in DSB resection, possibly by destabilizing the Mre11-Rad50 open conformation. These findings unmask the existence of structurally distinct Mre11 domains that support resistance to genotoxic agents by mediating different processes.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Helicases/metabolismo , Reparo do DNA , Endodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Antineoplásicos/farmacologia , Camptotecina/farmacologia , DNA Helicases/química , DNA Helicases/genética , Farmacorresistência Fúngica/efeitos dos fármacos , Farmacorresistência Fúngica/genética , Endodesoxirribonucleases/química , Endodesoxirribonucleases/genética , Endonucleases/química , Endonucleases/genética , Endonucleases/metabolismo , Exodesoxirribonucleases/química , Exodesoxirribonucleases/genética , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Mutação , Fleomicinas/farmacologia , Domínios Proteicos , Multimerização Proteica/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
6.
Nucleic Acids Res ; 45(9): 5269-5284, 2017 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-28334931

RESUMO

Rho GTPases are conserved molecules that control cytoskeletal dynamics. These functions are expedited by Rho GEFs that stimulate the release of GDP to enable GTP binding, thereby allowing Rho proteins to initiate intracellular signaling. How Rho GEFs and Rho GTPases protect cells from DNA damage is unknown. Here, we explore the extreme sensitivity of a deletion mutation in the Rho1p exchange factor Rgf1p to the DNA break/inducing antibiotic phleomycin (Phl). The Rgf1p mutant cells are defective in reentry into the cell cycle following the induction of severe DNA damage. This phenotype correlates with the inability of rgf1Δ cells to efficiently repair fragmented chromosomes after Phl treatment. Consistent with this observation Rad11p (ssDNA binding protein, RPA), Rad52p, Rad54p and Rad51p, which facilitate strand invasion in the process of homology-directed repair (HDR), are permanently stacked in Phl-induced foci in rgf1Δ cells. These phenotypes are phenocopied by genetic inhibition of Rho1p. Our data provide evidence that Rgf1p/Rho1p activity positively controls a repair function that confers resistance against the anti-cancer drug Phl.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Cromossomos Fúngicos/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Recombinação Homóloga/efeitos dos fármacos , Viabilidade Microbiana/efeitos dos fármacos , Mutação/genética , Fleomicinas/farmacologia , Schizosaccharomyces/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
7.
Curr Genet ; 63(2): 359-371, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27400920

RESUMO

Rhodosporidium toruloides DMKU3-TK16 (TK16), a basidiomycetous yeast isolated in Thailand, can produce a large amount of oil corresponding to approximately 70 % of its dry cell weight. However, lack of a sufficient and efficient transformation method makes further genetic manipulation of this organism difficult. We here developed a new transformation system for R. toruloides using a lithium acetate method with the Sh ble gene as a selective marker under the control of the R. toruloides ATCC 10657 GPD1 promoter. A linear DNA fragment containing the Sh ble gene expression cassette was integrated into the genome, and its integration was confirmed by colony PCR and Southern blot. Then, we further optimized the parameters affecting the transformation efficiency, such as the amount of linear DNA, the growth phase, the incubation time in the transformation mixture, the heat shock treatment temperature, the addition of DMSO and carrier DNA, and the recovery incubation time. With the developed method, the transformation efficiency of approximately 25 transformants/µg DNA was achieved. Compared with the initial trial, transformation efficiency was enhanced 417-fold. We further demonstrated the heterologous production of EGFP in TK16 by microscopic observation and immunoblot analysis, and use the technique to disrupt the endogenous URA3 gene. The newly developed method is thus simple and time saving, making it useful for efficient introduction of an exogenous gene into R. toruloides strains. Accordingly, this new practical approach should facilitate the molecular manipulation, such as target gene introduction and deletion, of TK16 and other R. toruloides strains as a major source of biodiesel.


Assuntos
Basidiomycota/genética , Basidiomycota/metabolismo , Engenharia Genética/métodos , Genoma Fúngico/genética , Óleos/metabolismo , Transformação Genética , Proteínas de Bactérias/genética , Basidiomycota/crescimento & desenvolvimento , Southern Blotting , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Microscopia Confocal , Modelos Genéticos , Mutação , Fleomicinas/farmacologia , Reação em Cadeia da Polimerase , Reprodutibilidade dos Testes , Temperatura , Transgenes/genética
8.
Cancer Lett ; 380(2): 467-475, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27431310

RESUMO

Cancer cells often exhibit altered epigenetic signatures that can misregulate genes involved in processes such as transcription, proliferation, apoptosis and DNA repair. As regulation of chromatin structure is crucial for DNA repair processes, and both DNA repair and epigenetic controls are deregulated in many cancers, we speculated that simultaneously targeting both might provide new opportunities for cancer therapy. Here, we describe a focused screen that profiled small-molecule inhibitors targeting epigenetic regulators in combination with DNA double-strand break (DSB) inducing agents. We identify UNC0638, a catalytic inhibitor of histone lysine N-methyl-transferase G9a, as hypersensitising tumour cells to low doses of DSB-inducing agents without affecting the growth of the non-tumorigenic cells tested. Similar effects are also observed with another, structurally distinct, G9a inhibitor A-366. We also show that small-molecule inhibition of G9a or siRNA-mediated G9a depletion induces tumour cell death under low DNA damage conditions by impairing DSB repair in a p53 independent manner. Furthermore, we establish that G9a promotes DNA non-homologous end-joining in response to DSB-inducing genotoxic stress. This study thus highlights the potential for using G9a inhibitors as anti-cancer therapeutic agents in combination with DSB-inducing chemotherapeutic drugs such as etoposide.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Quebras de DNA de Cadeia Dupla , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Fleomicinas/farmacologia , Quinazolinas/farmacologia , Inibidores da Topoisomerase II/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HCT116 , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Proteína Supressora de Tumor p53/genética
9.
J Biol Chem ; 291(33): 17228-46, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27325700

RESUMO

At the onset of anaphase, a protease called separase breaks the link between sister chromatids by cleaving the cohesin subunit Scc1. This irreversible step in the cell cycle is promoted by degradation of the separase inhibitor, securin, and polo-like kinase (Plk) 1-dependent phosphorylation of the Scc1 subunit. Plk could recognize substrates through interaction between its phosphopeptide interaction domain, the polo-box domain, and a phosphorylated priming site in the substrate, which has been generated by a priming kinase beforehand. However, the physiological relevance of this targeting mechanism remains to be addressed for many of the Plk1 substrates. Here, we show that budding yeast Plk1, Cdc5, is pre-deposited onto cohesin engaged in cohesion on chromosome arms in G2/M phase cells. The Cdc5-cohesin association is mediated by direct interaction between the polo-box domain of Cdc5 and Scc1 phosphorylated at multiple sites in its middle region. Alanine substitutions of the possible priming phosphorylation sites (scc1-15A) impair Cdc5 association with chromosomal cohesin, but they make only a moderate impact on mitotic cell growth even in securin-deleted cells (pds1Δ), where Scc1 phosphorylation by Cdc5 is indispensable. The same scc1-15A pds1Δ double mutant, however, exhibits marked sensitivity to the DNA-damaging agent phleomycin, suggesting that the priming phosphorylation of Scc1 poses an additional layer of regulation that enables yeast cells to adapt to genotoxic environments.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Substituição de Aminoácidos , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Mutação de Sentido Incorreto , Fleomicinas/farmacologia , Fosforilação/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Coesinas , Quinase 1 Polo-Like
10.
Nucleic Acids Res ; 44(8): 3728-38, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-26883631

RESUMO

Damaged DNA can be repaired by removal and re-synthesis of up to 30 nucleotides during base or nucleotide excision repair. An important question is what happens when many more nucleotides are removed, resulting in long single-stranded DNA (ssDNA) lesions. Such lesions appear on chromosomes during telomere damage, double strand break repair or after the UV damage of stationary phase cells. Here, we show that long single-stranded lesions, formed at dysfunctional telomeres in budding yeast, are re-synthesized when cells are removed from the telomere-damaging environment. This process requires Pol32, an accessory factor of Polymerase δ. However, re-synthesis takes place even when the telomere-damaging conditions persist, in which case the accessory factors of both polymerases δ and ε are required, and surprisingly, salt. Salt added to the medium facilitates the DNA synthesis, independently of the osmotic stress responses. These results provide unexpected insights into the DNA metabolism and challenge the current view on cellular responses to telomere dysfunction.


Assuntos
DNA Polimerase III/metabolismo , DNA Polimerase II/metabolismo , Reparo do DNA , Cloreto de Sódio/farmacologia , Telômero/enzimologia , Proliferação de Células/efeitos dos fármacos , Cromossomos Fúngicos/efeitos dos fármacos , Cromossomos Fúngicos/enzimologia , Cromossomos Fúngicos/metabolismo , DNA Polimerase I/fisiologia , DNA Fúngico/biossíntese , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , DNA Polimerase Dirigida por DNA/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fleomicinas/farmacologia , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Telômero/efeitos dos fármacos , Telômero/metabolismo , Homeostase do Telômero , Fatores de Transcrição/metabolismo
11.
Oncotarget ; 6(29): 28269-81, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26293673

RESUMO

To investigate how mineral dust-induced gene (mdig, also named as mina53, MINA, or NO52) promotes carcinogenesis through inducing active chromatin, we performed proteomics analyses for the interacting proteins that were co-immunoprecipitated by anti-mdig antibody from either the lung cancer cell line A549 cells or the human bronchial epithelial cell line BEAS-2B cells. On SDS-PAGE gels, three to five unique protein bands were consistently observed in the complexes pulled-down by mdig antibody, but not the control IgG. In addition to the mdig protein, several DNA repair or chromatin binding proteins, including XRCC5, XRCC6, RBBP4, CBX8, PRMT5, and TDRD, were identified in the complexes by the proteomics analyses using both Orbitrap Fusion and Orbitrap XL nanoESI-MS/MS in four independent experiments. The interaction of mdig with some of these proteins was further validated by co-immunoprecipitation using antibodies against mdig and its partner proteins, respectively. These data, thus, provide evidence suggesting that mdig accomplishes its functions on chromatin, DNA repair and cell growth through interacting with the partner proteins.


Assuntos
Reparo do DNA , Proteínas Nucleares/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Antibióticos Antineoplásicos/farmacologia , Antígenos Nucleares/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Eletroforese em Gel de Poliacrilamida , Histona Desmetilases , Humanos , Autoantígeno Ku , Dados de Sequência Molecular , Fleomicinas/farmacologia , Proteína 4 de Ligação ao Retinoblastoma/metabolismo , Homologia de Sequência de Aminoácidos , Espectrometria de Massas em Tandem/métodos
12.
BMC Cancer ; 14: 599, 2014 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-25134433

RESUMO

BACKGROUND: The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway is activated in cells exposed to various stimuli, including those originating on the cell surface or in the nucleus. Activated NF-κB signaling is thought to enhance cell survival in response to these stimuli, which include chemotherapy and radiation. In the present effort, we determined which anticancer drugs preferentially activate NF-κB in colon cancer cells. METHODS: NF-κB reporter cells were established and treated with 5-fluorouracil (5-FU, DNA/RNA damaging), oxaliplatin (DNA damaging), camptothecin (CTP, topoisomerase inhibitor), phleomycin (radiomimetic), or erlotinib (EGFR inhibitor). The activation of NF-κB was assessed by immunofluorescence for p65 translocation, luciferase assays, and downstream targets of NF-κB activation (cIAP2, and Bcl-XL) were evaluated by immunoblotting, by ELISA (CXCL8 and IL-6 in culture supernatants), or by gene expression analysis. RESULTS: Colon cancer cells responded variably to different classes of therapeutic agents, and these agents initiated variable responses among different cell types. CPT activated NF-κB in SW480 colon cancer cells in a dose-dependent manner, but not in HCT116 cells that were either wild-type or deficient for p53. In SW480 colon cancer cells, NF-κB activation by CPT was accompanied by secretion of the cytokine CXCL8, but not by up-regulation of the anti-apoptotic genes, cIAP2 or Bcl-XL. On the contrary, treatment of HCT116 cells with CPT resulted in up-regulation of CXCR2, a receptor for CXCL8, without an increase in cytokine levels. In SW480 cells, NF-κB reporter activity, but not cytokine secretion, was inhibited by SM-7368, an NF-κB inhibitor. CONCLUSION: The results show that, in response to cancer therapeutic agents, NF-κB activation varies with the cellular make up and that drug-induced NF-κB activation may be functionally uncoupled from anti-apoptotic outcomes found for other stimuli. Some cancer cells in a heterogeneous tumor tissue may, under therapeutic pressure, release soluble factors that have paracrine activity on neighboring cells that express the cognate receptors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Camptotecina/farmacologia , Camptotecina/uso terapêutico , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Cloridrato de Erlotinib , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Compostos Organoplatínicos/farmacologia , Compostos Organoplatínicos/uso terapêutico , Oxaliplatina , Fleomicinas/farmacologia , Fleomicinas/uso terapêutico , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico
13.
Bioengineered ; 4(5): 322-31, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23644277

RESUMO

To improve the general secretion ability of the biotechnologically relevant fungus Ashbya gossypii, random mutagenesis with ethyl methane sulfonate (EMS) was performed. The selection and screening strategy followed revealed mutants with improved secretion of heterologous Trichoderma reesei endoglucanase I (EGI), native α-amylase and/or native ß-glucosidase. One mutant, S436, presented 1.4- to 2-fold increases in all extracellular enzymatic activities measured, when compared with the parent strain, pointing to a global improvement in protein secretion. Three other mutants exhibited 2- to 3-fold improvements in only one (S397, B390) or two (S466) of the measured activities.   A targeted genetic approach was also followed. Two homologs of the Saccharomyces cerevisiae GAS1, AgGAS1A (AGL351W) and AgGAS1B (AGL352W), were deleted from the A. gossypii genome. For both copies deletion, a new antibiotic marker cassette conferring resistance to phleomycin, BLE3, was constructed. GAS1 encodes an ß-1,3-glucanosyltransglycosylase involved in cell wall assembly. Higher permeability of the cell wall was expected to increase the protein secretion capacity. However, total protein secreted to culture supernatants and secreted EGI activity did not increase in the Aggas1AΔ mutants. Deletion of the AgGAS1B copy affected cellular morphology and resulted in severe retardation of growth, similarly to what has been reported for GAS1-defficient yeast. Thus, secretion could not be tested in these mutants.


Assuntos
Parede Celular/genética , Celulase/genética , Proteínas Fúngicas/genética , Saccharomycetales/genética , alfa-Amilases/genética , beta-Glucosidase/genética , Parede Celular/enzimologia , Celulase/metabolismo , Farmacorresistência Fúngica , Metanossulfonato de Etila/farmacologia , Proteínas Fúngicas/metabolismo , Marcadores Genéticos , Engenharia Metabólica/métodos , Mutagênese , Mutação , Fleomicinas/farmacologia , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Saccharomycetales/efeitos dos fármacos , Saccharomycetales/enzimologia , Trichoderma/química , Trichoderma/enzimologia , Trichoderma/genética , alfa-Amilases/metabolismo , beta-Glucosidase/metabolismo
14.
Parasitology ; 140(4): 547-59, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23388105

RESUMO

We have previously shown that the subunit 1 of Leishmania amazonensis RPA (LaRPA-1) alone binds the G-rich telomeric strand and is structurally different from other RPA-1. It is analogous to telomere end-binding proteins described in model eukaryotes whose homologues were not identified in the protozoan´s genome. Here we show that LaRPA-1 is involved with damage response and telomere protection although it lacks the RPA1N domain involved with the binding with multiple checkpoint proteins. We induced DNA double-strand breaks (DSBs) in Leishmania using phleomycin. Damage was confirmed by TUNEL-positive nuclei and triggered a G1/S cell cycle arrest that was accompanied by nuclear accumulation of LaRPA-1 and RAD51 in the S phase of hydroxyurea-synchronized parasites. DSBs also increased the levels of RAD51 in non-synchronized parasites and of LaRPA-1 and RAD51 in the S phase of synchronized cells. More LaRPA-1 appeared immunoprecipitating telomeres in vivo and associated in a complex containing RAD51, although this interaction needs more investigation. RAD51 apparently co-localized with few telomeric clusters but it did not immunoprecipitate telomeric DNA. These findings suggest that LaRPA-1 and RAD51 work together in response to DNA DSBs and at telomeres, upon damage, LaRPA-1 works probably to prevent loss of single-stranded DNA and to assume a capping function.


Assuntos
Quebras de DNA de Cadeia Dupla , Leishmania/genética , Leishmania/metabolismo , Proteínas de Protozoários/metabolismo , Telômero/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Leishmania/efeitos dos fármacos , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fleomicinas/farmacologia
15.
Methods Mol Biol ; 845: 131-47, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22328372

RESUMO

The following transformation protocol is based on homologous recombination that occurs between a gene disruption or gene replacement construct and a target gene of Coccidioides. The DNA constructs employed contain either the gene that encodes for hygromycin B or phleomycin resistance, which are present in the pAN7.1 or pAN8.1 plasmid vectors, respectively. Hygromycin B or phleomycin are used to select for transformants at concentrations that inhibit growth of the parental strain. Coccidioides protoplasts generated from germinated arthroconidia are used for the transformation experiments. The plasmid DNA constructs are taken up by the protoplasts in the presence of calcium and polyethylene glycol. Twenty to 100 transformants/µg DNA can be obtained in each transformation experiment. Approximately 5-10% of the transformation events are homologous recombinations. Coccidioides cells in all developmental stages, including arthroconidia, are multinucleate. Since all Coccidioides nuclei are haploid, only one run of transformation is sufficient to create a mutant strain. However, the transformed protoplasts develop into heterokaryotic cells that typically contain both the parental and mutated nuclei. To isolate a homokaryotic strain, we perform multiple subcultures of the single colonies which contain heterokaryotic cells on selection plates with hygromycin B or phleomycin to enrich for the mutated nuclei. Homokaryotic mutants can be obtained after three to four subcultures of isolated colonies. In this protocol, we describe the methodology for preparation of Coccidioides protoplasts, transformation and isolation of homokaryotic mutants.


Assuntos
Cinamatos/farmacologia , Coccidioides/efeitos dos fármacos , Coccidioides/genética , Farmacorresistência Fúngica/genética , Deleção de Genes , Marcadores Genéticos/genética , Higromicina B/análogos & derivados , Fleomicinas/farmacologia , Farmacorresistência Fúngica/efeitos dos fármacos , Marcadores Genéticos/efeitos dos fármacos , Higromicina B/farmacologia , Mutagênese/genética
16.
Mycologia ; 103(5): 1019-27, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21498556

RESUMO

A growth assay was established for the barley pathogen Rhynchosporium commune with EGFP-tagged fungal mutants. This assay was used to study the effect of four antibiotics (hygromycin B, nourseothricin, kanamycin, phleomycin) and a herbicide (phosphinothricin) on fungal development. Fitting the growth curves to the modified Gompertz model allowed calculating growth parameters, such as lag periods of fungal colony formation and mycelial growth rates as well as EC(50) values. Except kanamycin all compounds were efficient inhibitors so that the corresponding resistance-conferring genes can be used as markers for selection of fungal transformation-based mutants. In addition the assay was used to quantify the inhibitory activity of a barley secondary metabolite, the indole alkaloid gramine.


Assuntos
Antifúngicos/farmacologia , Ascomicetos/efeitos dos fármacos , Ascomicetos/crescimento & desenvolvimento , Hordeum/microbiologia , Agrobacterium tumefaciens/metabolismo , Alcaloides/farmacologia , Aminobutiratos/farmacologia , Ascomicetos/patogenicidade , Proteínas de Fluorescência Verde , Herbicidas/farmacologia , Higromicina B/farmacologia , Alcaloides Indólicos , Canamicina/farmacologia , Micélio/efeitos dos fármacos , Micélio/crescimento & desenvolvimento , Fleomicinas/farmacologia , Estreptotricinas/farmacologia
17.
Mol Cell Biol ; 31(10): 1959-71, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21402778

RESUMO

Double-strand breaks (DSBs) in chromosomal DNA elicit a rapid signaling response through the ATM protein kinase. ATM corresponds to Tel1 in budding yeast. Here we show that the catalytic activity of Tel1 is altered by protein binding at DNA ends via the Mre11-Rad50-Xrs2 (MRX) complex. Like ATM, Tel1 is activated through interaction with the MRX complex and DNA ends. In vivo, Tel1 activation is enhanced in sae2Δ or mre11-3 mutants after camptothecin treatment; both of these mutants are defective in the removal of topoisomerase I from DNA. In contrast, an sae2Δ mutation does not stimulate Tel1 activation after expression of the EcoRI endonuclease, which generates "clean" DNA ends. In an in vitro system, tethering of Fab fragments to DNA ends inhibits MRX-mediated DNA end processing but enhances Tel1 activation. The mre11-3 mutation abolishes DNA end-processing activity but does not affect the ability to enhance Tel1 activation. These results support a model in which MRX controls Tel1 activation by recognizing protein-bound DNA ends.


Assuntos
DNA Fúngico/genética , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas Mutadas de Ataxia Telangiectasia , Camptotecina/farmacologia , Proteínas de Ciclo Celular/genética , Quebras de DNA de Cadeia Dupla , DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/metabolismo , DNA Fúngico/metabolismo , Proteínas de Ligação a DNA/genética , Endodesoxirribonucleases/genética , Ativação Enzimática , Exodesoxirribonucleases/genética , Histonas/metabolismo , Fragmentos Fab das Imunoglobulinas , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Fleomicinas/farmacologia , Proteínas Serina-Treonina Quinases/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
18.
Mutat Res ; 689(1-2): 12-20, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20450923

RESUMO

The Fanconi anaemia (FA) FANCG protein is an integral component of the FA nuclear core complex that is required for monoubiquitylation of FANCD2. FANCG is also part of another protein complex termed D1-D2-G-X3 that contains FANCD2 and the homologous recombination repair proteins BRCA2 (FANCD1) and XRCC3. Formation of the D1-D2-G-X3 complex is mediated by serine-7 phosphorylation of FANCG and occurs independently of the FA core complex and FANCD2 monoubiquitylation. FANCG contains seven tetratricopeptide repeat (TPR) motifs that mediate protein-protein interactions and here we show that mutation of several of the TPR motifs at a conserved consensus residue ablates the in vivo binding activity of FANCG. Expression of mutated TPR1, TPR2, TPR5 and TPR6 in Chinese hamster fancg mutant NM3 fails to functionally complement its hypersensitivities to mitomycin C (MMC) and phleomycin and fails to restore FANCD2 monoubiquitylation. Using co-immunoprecipitation analysis, we demonstrate that these TPR-mutated FANCG proteins fail to interact with BRCA2, XRCC3, FANCA or FANCF. The interactions of other proteins in the D1-D2-G-X3 complex are also absent, including the interaction of BRCA2 with both the monoubiquitylated (FANCD2-L) and non-ubiquitylated (FANCD2-S) isoforms of FANCD2. Interestingly, a mutation of TPR7 (R563E), that complements the MMC and phleomycin hypersensitivity of human FA-G EUFA316 cells, fails to complement NM3, despite the mutated FANCG protein co-precipitating with FANCA, BRCA2 and XRCC3. Whilst interaction of TPR7-mutated FANCG with FANCF does appear to be reduced in NM3, FANCD2 is monoubiquitylated suggesting that sub-optimal interactions of FANCG in the core complex and the D1-D2-G-X3 complex are responsible for the observed MMC- and phleomycin-hypersensitivity, rather than a defect in FANCD2 monoubiquitylation. Our data demonstrate that FANCG functions as a mediator of protein-protein interactions and is vital for the assembly of multi-protein complexes including the FA core complex and the D1-D2-G-X3 complex.


Assuntos
Motivos de Aminoácidos , Proteína BRCA2/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação G da Anemia de Fanconi/química , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fleomicinas/farmacologia , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Mutação , Recombinação Genética , Transfecção , Ubiquitinação
19.
Mutagenesis ; 25(3): 271-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20130020

RESUMO

Radiation-induced bystander effects have been evaluated extensively, including the involvement of the mitogen-activated protein kinase (MAPK) pathways. However, few studies have examined the ability of chemicals to induce bystander effects, and the molecular mechanisms involved in chemical bystander effects have not been investigated. We have previously demonstrated the ability of mitomycin C (MMC) and phleomycin (PHL) to induce bystander effects in normal human lymphoblastoid cells. Here, we demonstrate changes in the expression of MAPK target genes following bystander exposure to MMC or PHL or ionizing radiation. The expression changes of 18 genes, which code for proteins that are downstream targets of MAPK proteins, were evaluated at various time points following direct or bystander exposure to MMC, PHL and ionizing radiation. The 18 genes were analysed as groups belonging to one of the seven possible combinations of the three MAPK pathways. We observed statistically significant changes in expression of several genes following exposure to each agent. However, when the expression changes were analysed in the bystander cells alone, significant increases in expression of MAPK target genes were observed for MMC- and radiation-induced bystander effects but not for PHL. PHL is an acknowledged radiomimetic agent; however, in the present study, PHL responses did not resemble those of radiation. These results provide evidence for bystander-induced changes in MAPK proteins and downstream targets and suggest that the bystander effects are a part of a general stress response.


Assuntos
Efeito Espectador/efeitos dos fármacos , Efeito Espectador/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitomicina/farmacologia , Fleomicinas/farmacologia , Efeito Espectador/genética , Linhagem Celular , Dano ao DNA , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Radiação Ionizante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Elk-1 do Domínio ets/genética , Proteínas Elk-1 do Domínio ets/metabolismo
20.
Mutat Res ; 686(1-2): 15-29, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20034502

RESUMO

Many studies have examined bystander effects induced by ionizing radiation, however few have evaluated the ability of chemicals to induce similar effects. We previously reported the ability of two chemicals, mitomycin C (MMC) and phleomycin (PHL) to induce bystander effects in normal human lymphoblastoid cell lines. The focus of the current study was to determine the involvement of the MAPK proteins in bystander effects induced by physical and chemical DNA damaging agents and to evaluate the effects of MAPK inhibition on bystander-induced caspase 3/7 activation. The phosphorylation levels of the MAPK proteins ERK1/2, JNK, and p38, were measured from 1 to 24h following direct or bystander exposure to MMC, PHL or radiation. We observed transient phosphorylation, at early time points, of all 3 proteins in bystander cells. We also evaluated the effect of MAPK inhibition on bystander-induced caspase 3/7 activity to determine the role of MAPK proteins in bystander-induced apoptosis. We observed bystander-induced activation of caspase 3/7 in bystander cells. Inhibition of MAPK proteins resulted in a decrease in caspase 3/7 activity at the early time points, and the caspase activity increased (in the case of ERK inhibition) or returned to basal levels (in the case of JNK or p38 inhibition) between 12 and 24h. PHL is considered to be a radiomimetic agent, however in the present study PHL behaved more like a chemical and not like radiation in terms of MAPK phosphorylation. These results point to the involvement of MAPK proteins in the bystander effect induced by radiation and chemicals and provide additional evidence that this response is not limited to radiation but is a generalized stress response in cells.


Assuntos
Efeito Espectador/efeitos dos fármacos , Efeito Espectador/efeitos da radiação , Reagentes de Ligações Cruzadas/farmacologia , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Mitomicina/farmacologia , Fleomicinas/farmacologia , Radiação Ionizante , Linfócitos T/efeitos dos fármacos , Linfócitos T/efeitos da radiação , Caspase 3/metabolismo , Linhagem Celular Transformada , Dano ao DNA , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA