Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Genes Cells ; 21(7): 717-27, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27184081

RESUMO

The identification of novel synthetic ligands for G protein-coupled receptors (GPCRs) is important not only for understanding human physiology, but also for the development of novel drugs, especially for orphan GPCRs for which endogenous ligands are unknown. One of the orphan GPCR subfamilies, Super conserved Receptor Expressed in Brain (SREB), consists of GPR27, GPR85 and GPR173 and is expressed in the central nervous system. We report herein the identification of inverse agonists for the SREB family without their agonists. We carried out an in vitro screening of 5472 chemical compounds from the RIKEN NPDepo chemical library. The binding of [(35) S]GTPγS to the GPR173-Gsα fusion protein expressed in Sf9 cells was measured and resulted in the identification of 8 novel GPR173 inverse agonists. The most potent compound showed an IC50 of approximately 8 µm. The identified compounds were also antagonists for other SREB members, GPR27 and GPR85. These results indicated that the SREB family could couple Gs-type G proteins, and SREB-Gsα fusion proteins showed significant constitutive activities. Moreover, a molecular model of GPR173 was constructed using the screening results. The combination of computational and biological methods will provide a unique approach to ligand identification for orphan GPCRs and brain research.


Assuntos
Simulação de Acoplamento Molecular/métodos , Proteínas do Tecido Nervoso/química , Receptores Acoplados a Proteínas G/química , Bibliotecas de Moléculas Pequenas/química , Encéfalo/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Expressão Gênica , Humanos , Ligantes , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Interface Usuário-Computador
2.
J Hum Genet ; 61(8): 679-85, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27169690

RESUMO

In successive reports from 2014 to 2015, X-ray repair cross-complementing protein 4 (XRCC4) has been identified as a novel causative gene of primordial dwarfism. XRCC4 is indispensable for non-homologous end joining (NHEJ), the major pathway for repairing DNA double-strand breaks. As NHEJ is essential for V(D)J recombination during lymphocyte development, it is generally believed that abnormalities in XRCC4 cause severe combined immunodeficiency. Contrary to expectations, however, no overt immunodeficiency has been observed in patients with primordial dwarfism harboring XRCC4 mutations. Here, we describe the various XRCC4 mutations that lead to disease and discuss their impact on NHEJ and V(D)J recombination.


Assuntos
Proteínas de Ligação a DNA/genética , Nanismo Hipofisário/genética , Nanismo Hipofisário/imunologia , Estudos de Associação Genética , Mutação , Animais , Reparo do DNA por Junção de Extremidades , DNA Ligase Dependente de ATP/metabolismo , Proteínas de Ligação a DNA/metabolismo , Suscetibilidade a Doenças , Nanismo Hipofisário/diagnóstico , Estabilidade Enzimática , Humanos , Imunidade/genética , Síndromes de Imunodeficiência , Fenótipo , Ligação Proteica
3.
Genes (Basel) ; 14(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36833347

RESUMO

Natural antioxidants derived from plants exert various physiological effects, including antitumor effects. However, the molecular mechanisms of each natural antioxidant have not yet been fully elucidated. Identifying the targets of natural antioxidants with antitumor properties in vitro is costly and time-consuming, and the results thus obtained may not reliably reflect in vivo conditions. Therefore, to enhance understanding regarding the antitumor effects of natural antioxidants, we focused on DNA, one of the targets of anticancer drugs, and evaluated whether antioxidants, e.g., sulforaphane, resveratrol, quercetin, kaempferol, and genistein, which exert antitumor effects, induce DNA damage using gene-knockout cell lines derived from human Nalm-6 and HeLa cells pretreated with the DNA-dependent protein kinase inhibitor NU7026. Our results suggested that sulforaphane induces single-strand breaks or DNA strand crosslinks and that quercetin induces double-strand breaks. In contrast, resveratrol showed the ability to exert cytotoxic effects other than DNA damage. Our results also suggested that kaempferol and genistein induce DNA damage via unknown mechanisms. Taken together, the use of this evaluation system facilitates the analysis of the cytotoxic mechanisms of natural antioxidants.


Assuntos
Antioxidantes , Quebras de DNA de Cadeia Dupla , Humanos , Antioxidantes/farmacologia , Quempferóis , Resveratrol , Quercetina , Células HeLa , Genisteína , DNA
4.
FEBS J ; 290(22): 5313-5321, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37530740

RESUMO

Arsenic is a carcinogen that can cause skin, lung, and bladder cancer. While DNA double-strand breaks (DSBs) have been implicated in arsenic-induced carcinogenesis, the exact mechanism remains unclear. In this study, we performed genetic analysis to examine the impact of arsenic trioxide (As2 O3 ) on four different DSB repair pathways using the human pre-B cell line Nalm-6. Random integration analysis showed that As2 O3 does not negatively affect non-homologous end joining or polymerase theta-mediated end joining. In contrast, chromosomal DSB repair analysis revealed that As2 O3 decreases the efficiency of homologous recombination (HR) and, less prominently, single-strand annealing. Consistent with this finding, As2 O3 decreased gene-targeting efficiency, owing to a significant reduction in the frequency of HR-mediated targeted integration. To further verify the inhibitory effect of arsenic on HR, we examined cellular sensitivity to olaparib and camptothecin, which induce one-ended DSBs requiring HR for precise repair. Intriguingly, we found that As2 O3 significantly enhances sensitivity to those anticancer agents in HR-proficient cells. Our results suggest that arsenic-induced genomic instability is attributed to HR suppression, providing valuable insights into arsenic-associated carcinogenesis and therapeutic options.


Assuntos
Arsênio , Quebras de DNA de Cadeia Dupla , Humanos , Reparo do DNA , Recombinação Homóloga , Reparo do DNA por Junção de Extremidades , DNA , Carcinogênese
5.
Genes (Basel) ; 12(7)2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34356107

RESUMO

The DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a member of the phosphatidylinositol 3-kinase-related kinase family, phosphorylates serine and threonine residues of substrate proteins in the presence of the Ku complex and double-stranded DNA. Although it has been established that DNA-PKcs is involved in non-homologous end-joining, a DNA double-strand break repair pathway, the mechanisms underlying DNA-PKcs activation are not fully understood. Nevertheless, the findings of numerous in vitro and in vivo studies have indicated that DNA-PKcs contains two autophosphorylation clusters, PQR and ABCDE, as well as several autophosphorylation sites and conformational changes associated with autophosphorylation of DNA-PKcs are important for self-activation. Consistent with these features, an analysis of transgenic mice has shown that the phenotypes of DNA-PKcs autophosphorylation mutations are significantly different from those of DNA-PKcs kinase-dead mutations, thereby indicating the importance of DNA-PKcs autophosphorylation in differentiation and development. Furthermore, there has been notable progress in the high-resolution analysis of the conformation of DNA-PKcs, which has enabled us to gain a visual insight into the steps leading to DNA-PKcs activation. This review summarizes the current progress in the activation of DNA-PKcs, focusing in particular on autophosphorylation of this kinase.


Assuntos
Proteína Quinase Ativada por DNA/metabolismo , Proteína Quinase Ativada por DNA/fisiologia , Fosforilação/genética , Animais , Diferenciação Celular/genética , DNA/metabolismo , Dano ao DNA/genética , Reparo do DNA por Junção de Extremidades/fisiologia , Reparo do DNA/genética , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , Camundongos Transgênicos , Fosforilação/fisiologia
6.
Nucleic Acids Res ; 36(19): 6333-42, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18835848

RESUMO

In higher animal cells, the principal limitation of gene-targeting technology is the extremely low efficiency of targeted integration, which occurs three to four orders of magnitude less frequently than random integration. Assuming that random integration mechanistically involves non-homologous end-joining (NHEJ), inactivation of this pathway should reduce random integration and may enhance gene targeting. To test this possibility, we examined the frequencies of random and targeted integration in NHEJ-deficient chicken DT40 and human Nalm-6 cell lines. As expected, loss of NHEJ resulted in drastically reduced random integration in DT40 cells. Unexpectedly, however, this was not the case for Nalm-6 cells, indicating that NHEJ is not the sole mechanism of random integration. Nevertheless, we present evidence that NHEJ inactivation can lead to enhanced gene targeting through a reduction of random integration and/or an increase in targeted integration by homologous recombination. Most intriguingly, our results show that, in the absence of functional NHEJ, random integration of targeting vectors occurs more frequently than non-targeting vectors (harboring no or little homology to the host genome), implying that suppression of NHEJ-independent random integration events is needed to greatly enhance gene targeting in animal cells.


Assuntos
Marcação de Genes/métodos , Recombinação Genética , Animais , Linhagem Celular Tumoral , Galinhas/genética , DNA/metabolismo , DNA Ligases/genética , Deleção de Genes , Vetores Genéticos , Humanos
7.
FEBS J ; 287(2): 377-385, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31330087

RESUMO

Mammalian cells possess multiple pathways for repairing various types of DNA damage. Although the molecular mechanisms of each DNA repair pathway have been analyzed by biochemical analysis and cell biological analysis, interplay between different pathways has not been fully elucidated. In this study, using human Nalm-6-mutant cell lines, we analyzed the relationship between the base excision repair factor DNA polymerase ß (POLß) and DNA ligase IV (LIG4), which is essential for DNA double-strand break (DSB) repair by non-homologous end-joining (NHEJ). We found that cells lacking both POLß and LIG4 grew significantly more slowly than either single mutant, indicating cooperative functions of the two proteins in normal cell growth. To further investigate the genetic interaction between POLß and LIG4, we examined DNA damage sensitivity of the mutant cell lines. Our results suggested that NHEJ acts as a backup pathway for repairing alkylation damage (when converted into DSBs) in the absence of POLß. Surprisingly, despite the critical role of POLß in alkylation damage repair, cells lacking POLß exhibited increased resistance to camptothecin (a topoisomerase I inhibitor that induces DNA single-strand breaks), irrespective of the presence or absence of LIG4. A LIG4-independent increased resistance associated with POLß loss was also observed with ionizing radiation; however, cells lacking both POLß and LIG4 were more radiosensitive than either single mutant. Taken together, our findings provide novel insight into the complex interplay between different DNA repair pathways.


Assuntos
Reparo do DNA por Junção de Extremidades , DNA Ligase Dependente de ATP/genética , DNA Polimerase beta/genética , Camptotecina/toxicidade , Linhagem Celular , Dano ao DNA , DNA Ligase Dependente de ATP/metabolismo , DNA Polimerase beta/metabolismo , Resistência a Medicamentos , Humanos , Mutação , Tolerância a Radiação , Inibidores da Topoisomerase/toxicidade
8.
Biochem Biophys Res Commun ; 382(3): 492-6, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19285952

RESUMO

DNA transfection can be too inefficient to establish a desired number of stable transfectants, particularly in lymphocytes; however, this could be circumvented by increasing the absolute frequency of random integration. In this paper, we show that treating cells with topoisomerase II inhibitor following electroporation greatly (approximately 10- to 20-fold) enhances random integration of input DNA in a human pre-B lymphocyte cell line, Nalm-6. With the use of various kinds of topoisomerase II-targeting agents, we also present evidence that topoisomerase IIalpha inhibition is critical for the enhancement of random integration, while the contribution of topoisomerase IIbeta may be negligible. As topoisomerase IIalpha is highly expressed in vigorously growing cells, our results show that topoisomerase IIalpha inhibition provides a promising way of enhancing random integration in virtually all cultured cell lines.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , DNA/genética , Inibidores Enzimáticos/farmacologia , Células Precursoras de Linfócitos B/efeitos dos fármacos , Recombinação Genética/efeitos dos fármacos , Inibidores da Topoisomerase II , Transfecção , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Linhagem Celular , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Humanos , Células Precursoras de Linfócitos B/enzimologia , Recombinação Genética/genética
9.
J Biochem ; 166(4): 317-322, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31119277

RESUMO

Recent studies have revealed that not only proton-sensing channels, but also one family of G protein-coupled receptors (GPCRs) comprising OGR1, GPR4, G2A and TDAG8 are responsible for the sensing of extracellular protons, or pH. Here, we report that two other GPCRs, GPR31 and GPR151, were also activated in acidic condition. Elevated pH of assay mixtures resulted in a remarkable increase in [35S]GTPγS binding by GPR31-Giα and GPR151-Giα fusion proteins in a narrow range between pH 6 and 5. Our reporter gene assays with CHO cells expressing recombinant GPR31 or GPR151 also showed that activation was maximal at pH ∼5.8. Although these results from in vitro and cellular assays revealed slightly different pH sensitivities, all of our results indicated that GPR31 and GPR151 sensed extracellular protons equally well as other proton-sensing GPCRs.

10.
Biochem Biophys Res Commun ; 372(4): 662-7, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18538133

RESUMO

Non-homologous end joining (NHEJ) plays a major role in the repair of ionizing radiation-induced DNA double-strand breaks (DSBs), especially during the G1-phase of the cell cycle. Using a flow cytometric cell sorter, we fractionated G1- and S/G2-phase cells based on size to assess the DSB-repair activity in NHEJ factor-deficient DT40 and Nalm-6 cell lines. Colony formation assays revealed that the X-ray sensitivities of the G1-enriched populations correctly reflected the DSB-repair activities of both the DT40 and Nalm-6 cell lines. Furthermore, as assessed by gamma-H2AX foci formation, the sorted cells exhibited less DNA damage than chemically synchronized cells. Given that it does not use fluorescent labeling or chemical agents, this method of cell sorting is simpler and less toxic than other methods, making it applicable to a variety of cell lines, including those that cannot be synchronized by standard chemical treatments.


Assuntos
Separação Celular/métodos , Dano ao DNA , Reparo do DNA/genética , Citometria de Fluxo/métodos , Fase G1/efeitos da radiação , Tolerância a Radiação/genética , Proliferação de Células/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla , Fase G1/efeitos dos fármacos , Células HeLa , Histonas/análise , Humanos , Recombinação Genética/genética , Raios X
11.
DNA Cell Biol ; 27(1): 55-61, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17941805

RESUMO

Artemis is a recently identified factor involved in V(D)J recombination and nonhomologous end joining (NHEJ) of DNA double-strand break (DSB) repair. Here, we performed targeted disruption of the Artemis gene (ARTEMIS) in the human pre-B cell line Nalm-6. Unexpectedly, we found that cells lacking Artemis exhibit increased sensitivity to low doses, but not high doses, of ionizing radiation. We also show that ARTEMIS-deficient cells are hypersensitive to the topoisomerase II inhibitor etoposide, but to a much lesser extent than cells lacking DNA ligase IV, a critical component of NHEJ. Unlike DNA ligase IV-deficient cells, ARTEMIS-deficient cells were not hypersensitive to ICRF-193, a topoisomerase II inhibitor that does not stabilize topoisomerase II-DNA cleavable complexes. Collectively, our results suggest that Artemis only partially participates in the NHEJ pathway to repair DSBs in human somatic cells.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Inibidores Enzimáticos/farmacologia , Proteínas Nucleares/fisiologia , Southern Blotting , Western Blotting , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias , Primers do DNA/química , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA , Dicetopiperazinas , Relação Dose-Resposta à Radiação , Endonucleases , Etoposídeo/farmacologia , Citometria de Fluxo , Humanos , Piperazinas/farmacologia , Células Precursoras de Linfócitos B/efeitos dos fármacos , Células Precursoras de Linfócitos B/metabolismo , Células Precursoras de Linfócitos B/efeitos da radiação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tolerância a Radiação , Radiação Ionizante , Recombinação Genética/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidores da Topoisomerase II
12.
Methods Mol Biol ; 435: 17-29, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370065

RESUMO

Gene targeting provides a powerful means for studying gene function by a reverse genetic approach. Despite recent rapid progress in gene knockdown technologies, gene knockout studies using human somatic cells will be of greater importance for analyzing the functions of human genes in greater detail. Although the frequency of gene targeting is typically very low in human cultured cells, we have recently shown that a human precursor B cell line, Nalm-6, exceptionally allows for high-efficiency gene targeting by homologous recombination. In addition, we have developed a quick and simplified method to construct gene-targeting vectors, which is applicable to all sequenced organisms as well as embryonic stem cells. The combination of the simplified vector construction technology and the highly efficient gene-knockout system using Nalm-6 cells has enabled us to disrupt virtually any locus of the human genome within one month. Our system will greatly facilitate gene-knockout studies in human cells.


Assuntos
Marcação de Genes/métodos , Células Precursoras de Linfócitos B/metabolismo , Antígenos Nucleares/genética , Sequência de Bases , Linhagem Celular , Ensaio de Unidades Formadoras de Colônias , Primers do DNA/genética , Reparo do DNA , Proteínas de Ligação a DNA/genética , Eletroporação , Vetores Genéticos , Genoma Humano , Humanos , Autoantígeno Ku , Plasmídeos/genética , Recombinação Genética
13.
FEBS J ; 284(17): 2748-2763, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28622436

RESUMO

Gene targeting by homologous recombination provides the definitive tool for analyzing gene function. Promoterless vectors, which do not possess a promoter to drive marker gene expression, confer higher targeting efficiencies than conventional vectors due to the reduced number of drug-resistant clones. We here show that gene-targeting efficiency is typically ≥ 25% with the use of exon-trapping-type promoterless vectors in a human diploid cell line, Nalm-6. The efficiency of exon-trapping gene targeting was correlated with the level of target gene expression when a 2A peptide sequence was linked to the marker gene. Intriguingly, total arm length was not necessarily a determinant of targeting efficiency, as longer arms tend to enhance both homologous (targeted) and nonhomologous (nontargeted) integration of the vector; rather, the presence of an exon in the 5' arm led to a decreased targeting efficiency. Strikingly, loss of Rad54 did not severely affect the targeting efficiency of exon-trap vectors. Moreover, additional deletion of the Rad54 paralog Rad54B had limited impact on the high-efficiency gene targeting. These results indicate that targeted integration occurs in human cells even when both Rad54 and Rad54B are missing. These studies provide additional important insight into the contribution of various DNA repair factors on the targeting mechanics.


Assuntos
DNA Helicases/genética , Técnicas de Inativação de Genes , Proteínas Nucleares/genética , Linhagem Celular , Proteínas de Ligação a DNA , Éxons , Loci Gênicos , Vetores Genéticos , Humanos , Regiões Promotoras Genéticas
14.
Eur J Pharmacol ; 814: 130-137, 2017 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-28823924

RESUMO

GPBA is a G protein-coupled receptor that is activated by bile acids. Because activation of GPBA leads to increased cAMP levels and secretion of incretins and insulin, GPBA has been proposed as a promising drug target for the treatment of metabolic syndrome. Previously, we have developed a ligand-screening system to identify novel agonists of GPBA by means of a fusion protein of GPBA with G protein stimulatory α subunit (Gsα) and by a [35S]GTPγS-binding assay. To express the GPBA-Gsα fusion protein, transgenic silkworms were employed in this study, and cell membrane fractions were prepared from their fat body or pupae. We applied them to the screening of a chemical library that contains 10,625 compounds from the RIKEN Natural Products Depository (NPDepo). Eventually, a unique partial agonist, GUM2, was successfully identified. Our results indicated that the GPCR-Gα fusion proteins were beneficial for ligand identification and that the transgenic silkworms were useful for large-scale production of GPCRs. In HEK293 cells transiently expressing GPBA, GUM2 showed 50% effective concentration (EC50) of 3.5 ± 2.4µM and induced GPBA internalization as effectively as did an endogenous agonist, TLC. We also confirmed that GUM2 stimulates insulin secretion in MIN6 cells. Moreover, a single 2mg/kg dose of GUM2 significantly reduced blood glucose levels in mice during an intraperitoneal glucose tolerance test even though GUM2 is only a partial agonist with a low intrinsic activity. We concluded that GUM2 is a good candidate for research on GPBA signaling under physiological conditions and for the development of GPBA-targeting therapeutic compounds.


Assuntos
Produtos Biológicos/farmacologia , Glicemia/metabolismo , Teste de Tolerância a Glucose , Receptores Acoplados a Proteínas G/agonistas , Animais , Células HEK293 , Humanos , Insulina/metabolismo , Secreção de Insulina , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Larva/metabolismo , Camundongos , Pupa/metabolismo
15.
DNA Repair (Amst) ; 31: 29-40, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25973742

RESUMO

Exposure of cells to ionizing radiation or radiomimetic drugs generates DNA double-strand breaks that are processed either by homologous recombination repair (HRR), or by canonical, DNA-PKcs-dependent non-homologous end-joining (C-NHEJ). Chemical or genetic inactivation of factors involved in C-NHEJ or HRR, but also their local failure in repair proficient cells, promotes an alternative, error-prone end-joining pathway that serves as backup (A-EJ). There is evidence for the involvement of Artemis endonuclease, a protein deficient in a human radiosensitivity syndrome associated with severe immunodeficiency (RS-SCID), in the processing of subsets of DSBs by HRR or C-NHEJ. It is thought that within HRR or C-NHEJ Artemis processes DNA termini at complex DSBs. Whether Artemis has a role in A-EJ remains unknown. Here, we analyze using pulsed-field gel electrophoresis (PFGE) and specialized reporter assays, DSB repair in wild-type pre-B NALM-6 lymphocytes, as well as in their Artemis(-/-), DNA ligase 4(-/-) (LIG4(-/-)), and LIG4(-/-)/Artemis(-/-) double mutant counterparts, under conditions allowing evaluation of A-EJ. Our results substantiate the suggested roles of Artemis in C-NHEJ and HRR, but also demonstrate a role for the protein in A-EJ that is confirmed in Artemis deficient normal human fibroblasts. We conclude that Artemis is a nuclease participating in DSB repair by all major repair pathways.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Reparo do DNA , Endonucleases/genética , Ciclo Celular , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , DNA Ligase Dependente de ATP , DNA Ligases/genética , DNA Ligases/metabolismo , Proteínas de Ligação a DNA , Eletroforese em Gel de Campo Pulsado , Endonucleases/deficiência , Endonucleases/metabolismo , Fase G2 , Humanos , Recombinação Genética , Imunodeficiência Combinada Severa
16.
Eur J Pharmacol ; 767: 193-200, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26476280

RESUMO

Opioids are the most effective and widely used drugs for pain treatment. Morphine is an archetypal opioid and is an opioid receptor agonist. Unfortunately, the clinical usefulness of morphine is limited by adverse effects such as analgesic tolerance and addiction. Therefore, it is important to study the development of novel opioid agonists as part of pain control. The analgesic effects of opioids are mediated by three opioid receptors, namely opioid µ-, δ-, and κ-receptors. They belong to the G protein-coupled receptor superfamily and are coupled to Gi proteins. In the present study, we developed a ligand screening system to identify novel opioid µ-receptor agonists that measures [(35)S]GTPγS binding to cell membrane fractions prepared from the fat body of transgenic silkworms expressing µ-receptor-Gi1α fusion protein. We screened the RIKEN Natural Products Depository (NPDepo) chemical library, which contains 5848 compounds, and analogs of hit compounds. We successfully identified a novel, structurally unique compound, that we named GUM1, with agonist activity for the opioid µ-receptor (EC50 of 1.2 µM). The Plantar Test (Hargreaves' Method) demonstrated that subcutaneous injection of 3mg/kg of GUM1 into wild-type rats significantly extended latency time. This extension was also observed in a rat model of morphine tolerance and was inhibited by pre-treatment of naloxone. The unique molecular skeleton of GUM1 makes it an attractive molecule for further ligand-opioid receptor binding studies.


Assuntos
Benzilaminas/agonistas , Benzilaminas/farmacologia , Produtos Biológicos/farmacologia , Piranos/agonistas , Piranos/farmacologia , Receptores Opioides mu/agonistas , Analgésicos Opioides/agonistas , Analgésicos Opioides/farmacologia , Animais , Animais Geneticamente Modificados , Bombyx , Tolerância a Medicamentos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Masculino , Medição da Dor/efeitos dos fármacos , Ensaio Radioligante , Ratos , Receptores Opioides mu/genética , Radioisótopos de Enxofre/metabolismo
17.
Int J Biochem Cell Biol ; 35(1): 86-94, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12467650

RESUMO

Ku protein, a heterodimer of 70kDa (Ku70) and 86kDa (Ku86) polypeptides, is involved in non-homologous DNA end-joining (NHEJ) of DNA double-strand break repair and V(D)J recombination in combination with the catalytic component of DNA-dependent protein kinase (p470). Although Ku protein is known to be ubiquitously present in eukaryotic cells, it was previously reported to be absent in mature neutrophils. Using a mixture of protease inhibitors in the isolation procedure of neutrophils from human peripheral blood, we were able to detect Ku in the neutrophils by immunoblot and flow-cytometric analyses. Transcripts of Ku70 and Ku86 genes were also detected by the reverse transcriptase-polymerase chain reaction (RT-PCR), and Ku protein was shown to be localized in the nucleus of neutrophils as a heterodimer. Like poly(ADP-ribose) polymerase-1, neither mRNA nor protein of p470 was detected in the neutrophils. These results suggest that Ku is involved independently of p470 in DNA metabolism and signal transduction.


Assuntos
Antígenos Nucleares/metabolismo , DNA Helicases , Proteínas de Ligação a DNA/metabolismo , Neutrófilos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Antígenos Nucleares/genética , Núcleo Celular/metabolismo , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/genética , Dimerização , Citometria de Fluxo/métodos , Expressão Gênica , Humanos , Autoantígeno Ku , Proteínas Nucleares , Poli(ADP-Ribose) Polimerases/genética , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Proteínas Serina-Treonina Quinases/genética
19.
PLoS One ; 9(9): e108236, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25250686

RESUMO

Random integration of targeting vectors into the genome is the primary obstacle in human somatic cell gene targeting. Non-homologous end-joining (NHEJ), a major pathway for repairing DNA double-strand breaks, is thought to be responsible for most random integration events; however, absence of DNA ligase IV (LIG4), the critical NHEJ ligase, does not significantly reduce random integration frequency of targeting vector in human cells, indicating robust integration events occurring via a LIG4-independent mechanism. To gain insights into the mechanism and robustness of LIG4-independent random integration, we employed various types of targeting vectors to examine their integration frequencies in LIG4-proficient and deficient human cell lines. We find that the integration frequency of targeting vector correlates well with the length of homology arms and with the amount of repetitive DNA sequences, especially SINEs, present in the arms. This correlation was prominent in LIG4-deficient cells, but was also seen in LIG4-proficient cells, thus providing evidence that LIG4-independent random integration occurs frequently even when NHEJ is functionally normal. Our results collectively suggest that random integration frequency of conventional targeting vectors is substantially influenced by homology arms, which typically harbor repetitive DNA sequences that serve to facilitate LIG4-independent random integration in human cells, regardless of the presence or absence of functional NHEJ.


Assuntos
DNA Ligases/metabolismo , Marcação de Genes , Linhagem Celular , Reparo do DNA por Junção de Extremidades , DNA Ligase Dependente de ATP , DNA Ligases/genética , Deleção de Genes , Marcação de Genes/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos
20.
PLoS One ; 8(8): e72253, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967291

RESUMO

Nonhomologous end-joining (NHEJ) and homologous recombination (HR) are two major pathways for repairing DNA double-strand breaks (DSBs); however, their respective roles in human somatic cells remain to be elucidated. Here we show using a series of human gene-knockout cell lines that NHEJ repairs nearly all of the topoisomerase II- and low-dose radiation-induced DNA damage, while it negatively affects survival of cells harbouring replication-associated DSBs. Intriguingly, we find that loss of DNA ligase IV, a critical NHEJ ligase, and Artemis, an NHEJ factor with endonuclease activity, independently contribute to increased resistance to replication-associated DSBs. We also show that loss of Artemis alleviates hypersensitivity of DNA ligase IV-null cells to low-dose radiation- and topoisomerase II-induced DSBs. Finally, we demonstrate that Artemis-null human cells display increased gene-targeting efficiencies, particularly in the absence of DNA ligase IV. Collectively, these data suggest that DNA ligase IV and Artemis act cooperatively to promote NHEJ, thereby suppressing HR. Our results point to the possibility that HR can only operate on accidental DSBs when NHEJ is missing or abortive, and Artemis may be involved in pathway switching from incomplete NHEJ to HR.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , DNA Ligases/metabolismo , Recombinação Homóloga , Proteínas Nucleares/metabolismo , Linhagem Celular , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos da radiação , DNA Ligase Dependente de ATP , DNA Ligases/deficiência , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA , Endonucleases , Técnicas de Inativação de Genes , Marcação de Genes , Recombinação Homóloga/efeitos dos fármacos , Recombinação Homóloga/efeitos da radiação , Humanos , Mutação , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Inibidores da Topoisomerase II/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA