Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Anal Biochem ; 633: 114394, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34610334

RESUMEN

Human apurinic/pyrimidine endonuclease 1 (APE1) played a critical role in the occurrence, progress and prognosis of tumors through overexpression and subcellular localization. Thus, it has become an important target for enhancing the sensitivity of tumor cells to radiotherapy and chemotherapy. Therefore, detecting and imaging its intracellular activity is of great significance for inhibitor discovery, cancer diagnosis and therapy. In this work, using DNA-based nanoprobe, we developed a new method for monitor intracellular APE1 activity. The detecting system was consisted by single fluorophore labeled hairpin probe and reduced graphene oxide (rGO). The in vitro result showed that a liner response of the detection method ranged from 0.02 U/mL to 2 U/mL with a limit of detection of 0.02 U/mL. Furthermore, this strategy possessing high specificity was successfully applied for APE1-related inhibitor screening using intracellular fluorescence imaging. Panaxytriol, an effective inhibitor of APE1 activity, was screened from traditional Chinese medicine (TCM) and its effect on APE1 activity was monitored in real time in A549 cells. In summary, this sensitive and specific APE1 detection technology is expected to provide an assistance for APE1-related inhibitor screening and diseases diagnosis.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/análisis , ADN/química , Grafito/química , Nanopartículas/química , Células A549 , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Evaluación Preclínica de Medicamentos , Enediinos/farmacología , Alcoholes Grasos/farmacología , Humanos , Imagen Óptica , Factores de Tiempo
2.
Antiviral Res ; 167: 98-103, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31034848

RESUMEN

Curcumin, a polyphenol, is the main bioactive compound in dietary spice turmeric curcuma longa. It possesses anti-inflammatory, anti-oxidant and anti-neoplastic properties and shows potentials in treating or preventing particular diseases such as oxidative and inflammatory conditions, metabolic syndrome, arthritis, anxiety, hyperlipidemia and cancers. The diverse range and potential health beneficial effects has generated enthusiasm leading to intensive investigation into the phytochemical. However, a concern has been also raised if curcumin has a promiscuous bioassay profile and is a Pan-Assay INterference compound (PAINS). Here we present evidence indicating that curcumin is not a PAINS, but an inhibitor to APE1 redox function that affects many genes and pathways. This discovery explains the wide range of effects of curcumin on diverse human diseases and predicts a potential application in treatment of viral infection and virus-associated cancer. As a proof-of-concept, we demonstrated that curcumin is able to efficiently block Kaposi's sarcoma-associated herpesvirus replication and inhibit the pathogenic processes of angiogenesis and cell invasion.


Asunto(s)
Curcumina/farmacología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Herpesvirus Humano 8/efectos de los fármacos , Extractos Vegetales/farmacología , Antivirales/farmacología , Línea Celular , Curcuma , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Inhibidores Enzimáticos/farmacología , Herpesvirus Humano 8/patogenicidad , Humanos , Replicación Viral/efectos de los fármacos
3.
Physiol Plant ; 166(2): 494-512, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30035320

RESUMEN

Mitochondria are one of the major sites of reactive oxygen species (ROS) production in the plant cell. ROS can damage DNA, and this damage is in many organisms mainly repaired by the base excision repair (BER) pathway. We know very little about DNA repair in plants especially in the mitochondria. Combining proteomics, bioinformatics, western blot and enzyme assays, we here demonstrate that the complete BER pathway is found in mitochondria isolated from potato (Solanum tuberosum) tubers. The enzyme activities of three DNA glycosylases and an apurinic/apyrimidinic (AP) endonuclease (APE) were characterized with respect to Mg2+ dependence and, in the case of the APE, temperature sensitivity. Evidence for the presence of the DNA polymerase and the DNA ligase, which complete the repair pathway by replacing the excised base and closing the gap, was also obtained. We tested the effect of oxidative stress on the mitochondrial BER pathway by incubating potato tubers under hypoxia. Protein carbonylation increased significantly in hypoxic tuber mitochondria indicative of increased oxidative stress. The activity of two BER enzymes increased significantly in response to this oxidative stress consistent with the role of the BER pathway in the repair of oxidative damage to mitochondrial DNA.


Asunto(s)
Reparación del ADN/genética , ADN Mitocondrial/genética , ADN de Plantas/genética , Solanum tuberosum/genética , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Solanum tuberosum/metabolismo
4.
J Neuroinflammation ; 14(1): 243, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29233148

RESUMEN

BACKGROUND: The production of reactive oxygen species (ROS) during pneumococcal meningitis (PM) leads to severe DNA damage in the neurons and is the major cause of cell death during infection. Hence, the use of antioxidants as adjuvant therapy has been investigated. Previous studies have demonstrated the possible participation of apurinic/apyrimidinic endonuclease (APE1) during PM. The aims of this study were to investigate the APE1 expression in the cortical and hippocampal tissues of infant Wistar rats infected with Streptococcus pneumoniae and its association with cell death and understand the role of vitamin B6 (vitB6) as a protective factor against cell death. METHODS: APE1 expression and oxidative stress markers were analyzed at two-time points, 20 and 24 h post infection (p.i.), in the cortex (CX) and hippocampus (HC) of rats supplemented with vitB6. Statistical analyses were performed by the nonparametric Kruskal-Wallis test using Dunn's post test. RESULTS: Our results showed high protein levels of APE1 in CX and HC of infected rats. In the CX, at 20 h p.i., vitB6 supplementation led to the reduction of expression of APE1 and apoptosis-inducing factor, while no significant changes in the transcript levels of caspase-3 were observed. Furthermore, levels of carbonyl content and glutamate in the CX were reduced by vitB6 supplementation at the same time point of 20 h p.i.. Since our data showed a significant effect of vitB6 on the CX at 20 h p.i. rather than that at 24 h p.i., we evaluated the effect of administering a second dose of vitB6 at 18 h p.i. and sacrifice at 24 h p.i.. Reduction in the oxidative stress and APE1 levels were observed, although the latter was not significant. Although the levels of APE1 was not significantly changed in the HC with vitB6 adjuvant therapy, vitB6 supplementation prevented the formation of the truncated form of APE1 (34 kDa) that is associated with apoptosis. CONCLUSIONS: Our data suggest that PM affects APE1 expression, which can be modulated by vitB6. Additionally, vitB6 contributes to the reduction of glutamate and ROS levels. Besides the potential to reduce cell death and oxidative stress during neuroinflammation, vitB6 showed enhanced effect on the CX than on the HC during PM.


Asunto(s)
Antioxidantes/farmacología , Encéfalo/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Meningitis Neumocócica/metabolismo , Vitamina B 6/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Reparación del ADN , Meningitis Neumocócica/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar
5.
Sci Rep ; 7(1): 13867, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29066842

RESUMEN

Oxidative stress initiates harmful cellular responses, such as DNA damage and protein denaturation, triggering a series of cardiovascular disorders. Systematic investigations of the transcription factors (TFs) involved in oxidative stress can help reveal the underlying molecular mechanisms and facilitate the discovery of effective therapeutic targets in related diseases. In this study, an integrated strategy which integrated RNA-seq-based transcriptomics techniques and a newly developed concatenated tandem array of consensus TF response elements (catTFREs)-based proteomics approach and then combined with a network pharmacology analysis, was developed and this integrated strategy was used to investigate critical TFs in the protection of Yixin-shu (YXS), a standardized medical product used for ischaemic heart disease, against hydrogen peroxide (H2O2)-induced damage in cardiomyocytes. Importantly, YXS initiated biological process such as anti-apoptosis and DNA repair to protect cardiomyocytes from H2O2-induced damage. By using the integrated strategy, DNA-(apurinic or apyrimidinic site) lyase (Apex1), pre B-cell leukemia transcription factor 3 (Pbx3), and five other TFs with their functions involved in anti-oxidation, anti-apoptosis and DNA repair were identified. This study offers a new understanding of the mechanism underlying YXS-mediated protection against H2O2-induced oxidative stress in cardiomyocytes and reveals novel targets for oxidative stress-related diseases.


Asunto(s)
Citoprotección/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Peróxido de Hidrógeno/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Evaluación Preclínica de Medicamentos , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Miocitos Cardíacos/citología , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos , Transcriptoma/efectos de los fármacos
6.
Free Radic Biol Med ; 112: 480-493, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28822748

RESUMEN

Oxidative stress plays a vital role in many pathological processes of the cardiovascular diseases. However, the underlying mechanism remains unclear, especially on a transcription factor (TF) level. In this study, a new method, concatenated tandem array of consensus transcription factor response elements (catTFREs), and an Illumina-based RNA-seq technology were integrated to systematically investigate the role of TFs in hydrogen peroxide (H2O2)-induced oxidative stress in cardiomyocytes; the damage was then rescued by Danhong injection (DHI), a Chinese standardized product approved for cardiovascular diseases treatment. The overall gene expression revealed cell apoptosis and DNA repair were vital for cardiomyocytes in resisting oxidative stress. By comprehensively integrating the transcription activity of TFs and their downstream target genes, an important TFs-target network were constructed and 13 TFs were identified as critical TFs in DHI-mediated protection in H2O2-induced oxidative stress. By using the integrated approach, seven TFs of these 13 TFs were also identified in melatonin-mediated protection in H2O2-induced damage. Furthermore, the transcription activity of DNA-(apurinic or apyrimidinic site) lyase (Apex1), Myocyte-specific enhancer factor 2D (Mef2d) and Pre B-cell leukemia transcription factor 3 (Pbx3) was further verified in pluripotent stem cell-derived cardiomyocytes. This research offers a new understanding of cardiomyocytes in response to H2O2-induced oxidative stress and reveals additional potential therapeutic targets. The combination of two parallel omics datasets (corresponding to the transcriptome and proteome) can reduce the noise in high-throughput data and reveal the fundamental changes of the biological process, making it suitable and reliable for investigation of critical targets in many other complicated pathological processes.


Asunto(s)
Cardiotónicos/farmacología , Reparación del ADN/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Peróxido de Hidrógeno/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Factores de Transcripción/genética , Transcriptoma , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Peróxido de Hidrógeno/farmacología , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Melatonina/farmacología , Análisis por Micromatrices , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Elementos de Respuesta , Factores de Transcripción/metabolismo
7.
Int J Oncol ; 49(6): 2331-2340, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27748797

RESUMEN

Lung cancer is the leading cause of cancer-related deaths worldwide and is associated with a very poor outcome. Oxymatrine exerts antitumor effects by inducing apoptosis and inhibiting the proliferation of different cancer cells; however, the anticancer effects and mechanism of action of oxymatrine have not been evaluated sufficiently in human lung cancer cells. Thus, the present study aimed to investigate the anticancer effects of oxymatrine in human lung cancer cells and identify the molecular mechanisms underlying these effects. MTT assays demonstrated that oxymatrine significantly inhibited the proliferation of A549 and H1299 cells in a time- and dose-dependent manner. In addition, flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling assays suggested that oxymatrine treatment may induce lung cancer cell apoptosis in a dose-dependent manner. Furthermore, we detected that oxymatrine induced a significant increase in DNA damage and the expression of PARP and phosphorylated H2AX, and a significant decrease in that of nuclear APE1 and AP endonuclease activity in A549 cells. APE1 knockdown cells (APE1shRNA) plus oxymatrine treatment reduced cells proliferation and induced apoptosis more seriously than control shRNA cells. This appeared to be a consequence of an increase in the number of apurinic/apyrimidinic (AP) sites, DNA damage, PARP and H2AX phosphorylation, which together resulted in the induction of apoptosis. In contrast, the sensitizing effects of APE1 overexpression plus oxymatrine treatment did not occur in APEOE cells. These findings reveal a potential mechanism of action for oxymatrine-induced apoptosis and suggest that oxymatrine is a promising potential therapeutic agent for the treatment of lung cancer.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Quinolizinas/farmacología , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Histonas/biosíntesis , Humanos , Neoplasias Pulmonares/patología , Medicina Tradicional China , Poli(ADP-Ribosa) Polimerasa-1/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/genética
8.
J Pharmacol Exp Ther ; 359(2): 300-309, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27608656

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) is a potentially debilitating side effect of a number of chemotherapeutic agents. There are currently no U.S. Food and Drug Administration-approved interventions or prevention strategies for CIPN. Although the cellular mechanisms mediating CIPN remain to be determined, several lines of evidence support the notion that DNA damage caused by anticancer therapies could contribute to the neuropathy. DNA damage in sensory neurons after chemotherapy correlates with symptoms of CIPN. Augmenting apurinic/apyrimidinic endonuclease (APE)-1 function in the base excision repair pathway reverses this damage and the neurotoxicity caused by anticancer therapies. This neuronal protection is accomplished by either overexpressing APE1 or by using a first-generation targeted APE1 small molecule, E3330 [(2E)-2-[(4,5-dimethoxy-2-methyl-3,6-dioxo-1,4-cyclohexadien-1-yl)methylene]-undecanoic acid; also called APX3330]. Although E3330 has been approved for phase 1 clinical trials (Investigational New Drug application number IND125360), we synthesized novel, second-generation APE1-targeted molecules and determined whether they would be protective against neurotoxicity induced by cisplatin or oxaliplatin while not diminishing the platins' antitumor effect. We measured various endpoints of neurotoxicity using our ex vivo model of sensory neurons in culture, and we determined that APX2009 [(2E)-2-[(3-methoxy-1,4-dioxo-1,4-dihydronaphthalen-2-yl)methylidene]-N,N-diethylpentanamide] is an effective small molecule that is neuroprotective against cisplatin and oxaliplatin-induced toxicity. APX2009 also demonstrated a strong tumor cell killing effect in tumor cells and the enhanced tumor cell killing was further substantiated in a more robust three-dimensional pancreatic tumor model. Together, these data suggest that the second-generation compound APX2009 is effective in preventing or reversing platinum-induced CIPN while not affecting the anticancer activity of platins.


Asunto(s)
Antineoplásicos/efectos adversos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/prevención & control , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/efectos adversos , Sistema Enzimático del Citocromo P-450/metabolismo , Daño del ADN , Evaluación Preclínica de Medicamentos , Humanos , Modelos Moleculares , Conformación Molecular , Compuestos Organoplatinos/efectos adversos , Oxaliplatino , Enfermedades del Sistema Nervioso Periférico/enzimología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/patología
9.
Antioxid Redox Signal ; 24(15): 855-66, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-26650895

RESUMEN

AIMS: The sodium-iodide symporter (NIS) mediates the uptake of I(-) by the thyroid follicular cell and is essential for thyroid hormone biosynthesis. Nis expression is stimulated by thyroid-stimulating hormone (TSH) and also requires paired box 8 (Pax8) to bind to its promoter. Pax8 binding activity depends on its redox state by a mechanism involving thioredoxin/thioredoxin reductase-1 (Txn/TxnRd1) reduction of apurinic/apyrimidinic endonuclease 1 (Ape1). In this study, we investigate the role of Se in Nis expression. RESULTS: Selenium increases TSH-induced Nis expression and activity in rat thyroid cells. The stimulatory effect of Se occurs at the transcriptional level and is only observed for Nis promoters containing a Pax8 binding site in the Nis upstream enhancer, suggesting that Pax8 is involved in this effect. In fact, Se increases Pax8 expression and its DNA-binding capacity, and in Pax8-silenced rat thyroid cells, Nis is not Se responsive. By inhibiting Ape1 and TxnRd1 functions, we found that both enzymes are crucial for TSH and TSH plus Se stimulation of Pax8 activity and mediate the Nis response to Se treatment. INNOVATION: We describe that Se increases Nis expression and activity. We demonstrate that this effect is dependent on the redox functions of Ape1 and Txn/TxnRd1 through control of the DNA binding activity of Pax8. CONCLUSION: Nis expression is controlled by Txn/Ape1 through a TSH/Se-dependent mechanism. These findings open a new field of study regarding the regulation of Nis activity in thyroid cells. Antioxid. Redox Signal. 24, 855-866.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Factor de Transcripción PAX8/metabolismo , Selenio/fisiología , Simportadores/genética , Tiorredoxinas/fisiología , Tirotropina/fisiología , Animales , Línea Celular , Glutatión Peroxidasa/metabolismo , Oxidación-Reducción , Unión Proteica , Ratas , Simportadores/metabolismo , Tiorredoxina Reductasa 1/metabolismo , Transcripción Genética , Activación Transcripcional , Glutatión Peroxidasa GPX1
10.
Drug Des Devel Ther ; 8: 2147-60, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25395832

RESUMEN

Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a multifunctional protein possessing both DNA repair and redox regulatory activities. It has been shown that blocking redox function leads to genotoxic, antiangiogenic, cytostatic, and proapoptotic effects in cells. Therefore, the selective inhibitors against APE1's redox function can be served as potential pharmaceutical candidates in cancer therapeutics. In the present study, we identified the biological specificity of the Chinese herbal compound tanshinone IIA (T2A) in blocking the redox function of APE1. Using dual polarization interferometry, the direct interaction between APE1 and T2A was observed with a KD value at subnanomolar level. In addition, we showed that T2A significantly compromised the growth of human cervical cancer and colon cancer cells. Furthermore, the growth-inhibitory or proapoptotic effect of T2A was diminished in APE1 knockdown or redox-deficient cells, suggesting that the cytostatic effect of T2A might be specifically through inhibiting the redox function of APE1. Finally, T2A pretreatment enhanced the cytotoxicity of ionizing radiation or other chemotherapeutic agents in human cervical cancer and colon cancer cell lines. The data presented herein suggest T2A as a promising bioactive inhibitor of APE1 redox activity.


Asunto(s)
Abietanos/farmacología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Células Cultivadas , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Relación Dosis-Respuesta a Droga , Células HCT116 , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Simulación del Acoplamiento Molecular , Oxidación-Reducción/efectos de los fármacos , Relación Estructura-Actividad
11.
Lasers Med Sci ; 29(6): 1953-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24930134

RESUMEN

Although red laser lights lie in the region of non-ionizing radiations in the electromagnetic spectrum, there are doubts whether absorption of these radiations causes lesions in the DNA molecule. Our aim was to investigate the expression of the genes involved with base excision and nucleotide excision repair pathways in skin tissue submitted to burn injury and exposed to low-level red laser. Wistar rats were divided as follows: control group-rats burned and not irradiated, laser group-rats burned and irradiated 1 day after injury for five consecutive days, and later laser group-rats injured and treated 4 days after injury for five consecutive days. Irradiation was performed according to a clinical protocol (20 J/cm(2), 100 mW, continuous wave emission mode). The animals were sacrificed on day 10, and scarred tissue samples were withdrawn for total RNA extraction, complementary DNA (cDNA) synthesis, and evaluation of gene expression by quantitative polymerase chain reaction. Low-level red laser exposure (1) reduces the expression of APE1 messenger (mRNA), (2) increases the expression of OGG1 mRNA, (3) reduces the expression of XPC mRNA, and (4) increases the expression of XPA mRNA both in laser and later laser groups. Red laser exposure at therapeutic fluences alters the expression of genes related to base excision and nucleotide excision pathways of DNA repair during wound healing of burned skin.


Asunto(s)
Quemaduras/etiología , Reparación del ADN , Terapia por Luz de Baja Intensidad/efectos adversos , Piel/efectos de la radiación , Cicatrización de Heridas/efectos de la radiación , Animales , Proteínas Portadoras/metabolismo , ADN Glicosilasas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Perfilación de la Expresión Génica , Rayos Láser , Masculino , Ratas , Ratas Wistar , Piel/metabolismo
12.
Redox Biol ; 2: 485-94, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24624338

RESUMEN

The effectiveness of current treatment for age related macular degeneration (AMD) by targeting one molecule is limited due to its multifactorial nature and heterogeneous pathologies. Treatment strategy to target multiple signaling pathways or pathological components in AMD pathogenesis is under investigation for better clinical outcome. Inhibition of the redox function of apurinic endonuclease 1/redox factor-1 (APE1) was found to suppress endothelial angiogenesis and promote neuronal cell recovery, thereby may serve as a potential treatment for AMD. In the current study, we for the first time have found that a specific inhibitor of APE1 redox function by a small molecule compound E3330 regulates retinal pigment epithelium (RPEs) cell response to oxidative stress. E3330 significantly blocked sub-lethal doses of oxidized low density lipoprotein (oxLDL) induced proliferation decline and senescence advancement of RPEs. At the same time, E3330 remarkably decreased the accumulation of intracellular reactive oxygen species (ROS) and down-regulated the productions of monocyte chemoattractant protein-1 (MCP-1) and vascular endothelial growth factor (VEGF), as well as attenuated the level of nuclear factor-κB (NF-κB) p65 in RPEs. A panel of stress and toxicity responsive transcription factors that were significantly upregulated by oxLDL was restored by E3330, including Nrf2/Nrf1, p53, NF-κB, HIF1, CBF/NF-Y/YY1, and MTF-1. Further, a single intravitreal injection of E3330 effectively reduced the progression of laser-induced choroidal neovascularization (CNV) in mouse eyes. These data revealed that E3330 effectively rescued RPEs from oxidative stress induced senescence and dysfunctions in multiple aspects in vitro, and attenuated laser-induced damages to RPE-Bruch׳s membrane complex in vivo. Together with its previously established anti-angiogenic and neuroprotection benefits, E3330 is implicated for potential use for AMD treatment.


Asunto(s)
Benzoquinonas/administración & dosificación , Neovascularización Coroidal/tratamiento farmacológico , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Fármacos Neuroprotectores/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Propionatos/administración & dosificación , Epitelio Pigmentado de la Retina/metabolismo , Animales , Senescencia Celular/efectos de los fármacos , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inyecciones Intravítreas , Ratones , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología
13.
Nat Commun ; 4: 2674, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24154628

RESUMEN

Cellular responses to DNA damage involve distinct DNA repair pathways, such as mismatch repair (MMR) and base excision repair (BER). Using Caenorhabditis elegans as a model system, we present genetic and molecular evidence of a mechanistic link between processing of DNA damage and activation of autophagy. Here we show that the BER AP endonucleases APN-1 and EXO-3 function in the same pathway as MMR, to elicit DNA-directed toxicity in response to 5-fluorouracil, a mainstay of systemic adjuvant treatment of solid cancers. Immunohistochemical analyses suggest that EXO-3 generates the DNA nicks required for MMR activation. Processing of DNA damage via this pathway, in which both BER and MMR enzymes are required, leads to induction of autophagy in C. elegans and human cells. Hence, our data show that MMR- and AP endonuclease-dependent processing of 5-fluorouracil-induced DNA damage leads to checkpoint activation and induction of autophagy, whose hyperactivation contributes to cell death.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Reparación de la Incompatibilidad de ADN , Reparación del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Endodesoxirribonucleasas/genética , Animales , Antimetabolitos Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Roturas del ADN de Cadena Simple/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endodesoxirribonucleasas/metabolismo , Fluorouracilo/farmacología , Regulación de la Expresión Génica , Humanos , Transducción de Señal
14.
Neurol Sci ; 34(3): 305-12, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22391677

RESUMEN

Redox effector factor (Ref-1) is a sensitive marker for oxidative cellular injury. The aim of this study was to investigate the effects of acupuncture on hippocampal Ref-1 expression in cerebral multi-infarction rats. The rats with reference memory impairment were randomly allocated to three groups: impaired group, acupuncture group and placebo acupuncture group. Moreover, normal group and sham-operated group were set as control groups. Morris water maze test showed that cerebral multi-infarction rats did not present significant changes in spatial working memory performance. Further investigation by immunohistochemistry revealed that acupunctural treatment significantly increased the expression of Ref-1 in the hippocampus of the impaired rats. These findings suggested that the spatial working memory was unaffected in the cerebral multi-infarction rats although spatial reference memory deficits were detected in our previous study; in addition, acupuncture could increase the Ref-1 expression, consequently exerting the anti-oxidant effects.


Asunto(s)
Acupuntura , Infarto Cerebral/patología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Regulación de la Expresión Génica/fisiología , Hipocampo/metabolismo , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Conducta Exploratoria , Masculino , Aprendizaje por Laberinto , Memoria a Corto Plazo/fisiología , Ratas , Ratas Wistar
15.
PLoS One ; 7(10): e47974, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23110144

RESUMEN

The major human apurinic/apyrimidinic endonuclease APE1 plays a pivotal role in the repair of base damage via participation in the DNA base excision repair (BER) pathway. Increased activity of APE1, often observed in tumor cells, is thought to contribute to resistance to various anticancer drugs, whereas down-regulation of APE1 sensitizes cells to DNA damaging agents. Thus, inhibiting APE1 repair endonuclease function in cancer cells is considered a promising strategy to overcome therapeutic agent resistance. Despite ongoing efforts, inhibitors of APE1 with adequate drug-like properties have yet to be discovered. Using a kinetic fluorescence assay, we conducted a fully-automated high-throughput screen (HTS) of the NIH Molecular Libraries Small Molecule Repository (MLSMR), as well as additional public collections, with each compound tested as a 7-concentration series in a 4 µL reaction volume. Actives identified from the screen were subjected to a panel of confirmatory and counterscreen tests. Several active molecules were identified that inhibited APE1 in two independent assay formats and exhibited potentiation of the genotoxic effect of methyl methanesulfonate with a concomitant increase in AP sites, a hallmark of intracellular APE1 inhibition; a number of these chemotypes could be good starting points for further medicinal chemistry optimization. To our knowledge, this represents the largest-scale HTS to identify inhibitors of APE1, and provides a key first step in the development of novel agents targeting BER for cancer treatment.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Daño del ADN , Reparación del ADN/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Células HeLa , Humanos , Metilmetanosulfonato/antagonistas & inhibidores , Metilmetanosulfonato/farmacología , Estructura Molecular , Relación Estructura-Actividad
16.
Biochemistry ; 51(31): 6246-59, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22788932

RESUMEN

The repair of abasic sites that arise in DNA from hydrolytic depurination/depyrimidination of the nitrogenous bases from the sugar-phosphate backbone and the action of DNA glycosylases on deaminated, oxidized, and alkylated bases are critical to cell survival. Apurinic/apyrimidinic endonuclease-1/redox effector factor-1 (APE-1; aka APE1/ref-1) is responsible for the initial removal of abasic lesions as part of the base excision repair pathway. Deletion of APE-1 activity is embryonic lethal in animals and is lethal in cells. Potential inhibitors of the repair function of APE-1 were identified based upon molecular modeling of the crystal structure of the APE-1 protein. We describe the characterization of several unique nanomolar inhibitors using two complementary biochemical screens. The most active molecules all contain a 2-methyl-4-amino-6,7-dioxolo-quinoline structure that is predicted from the modeling to anchor the compounds in the endonuclease site of the protein. The mechanism of action of the selected compounds was probed by fluorescence and competition studies, which indicate, in a specific case, direct interaction between the inhibitor and the active site of the protein. It is demonstrated that the inhibitors induce time-dependent increases in the accumulation of abasic sites in cells at levels that correlate with their potency to inhibit APE-1 endonuclease excision. The inhibitor molecules also potentiate by 5-fold the toxicity of a DNA methylating agent that creates abasic sites. The molecules represent a new class of APE-1 inhibitors that can be used to probe the biology of this critical enzyme and to sensitize resistant tumor cells to the cytotoxicity of clinically used DNA damaging anticancer drugs.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Secuencia de Bases , Dominio Catalítico , Línea Celular Tumoral , ADN/genética , ADN/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/toxicidad , Humanos , Simulación del Acoplamiento Molecular , Oxidación-Reducción/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/toxicidad
17.
Free Radic Biol Med ; 53(7): 1478-88, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22709585

RESUMEN

Oxidative stress and mitochondrial dysfunction have been implicated in the pathology of HD; however, the precise mechanisms by which mutant huntingtin modulates levels of oxidative damage in turn resulting in mitochondrial dysfunction are not known. We hypothesize that mutant huntingtin increases oxidative mtDNA damage leading to mitochondrial dysfunction. We measured nuclear and mitochondrial DNA lesions and mitochondrial bioenergetics in the STHdhQ7 and STHdhQ111 in vitro striatal model of HD. Striatal cells expressing mutant huntingtin show higher basal levels of mitochondrial-generated ROS and mtDNA lesions and a lower spare respiratory capacity. Silencing of APE1, the major mammalian apurinic/apyrimidinic (AP) endonuclease that participates in the base excision repair (BER) pathway, caused further reductions of spare respiratory capacity in the mutant huntingtin-expressing cells. Localization experiments show that APE1 increases in the mitochondria of wild-type Q7 cells but not in the mutant huntingtin Q111 cells after treatment with hydrogen peroxide. Moreover, these results are recapitulated in human HD striata and HD skin fibroblasts that show significant mtDNA damage (increased lesion frequency and mtDNA depletion) and significant decreases in spare respiratory capacity, respectively. These data suggest that mtDNA is a major target of mutant huntingtin-associated oxidative stress and may contribute to subsequent mitochondrial dysfunction and that APE1 (and, by extension, BER) is an important target in the maintenance of mitochondrial function in HD.


Asunto(s)
ADN Mitocondrial/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Enfermedad de Huntington/metabolismo , Mitocondrias/metabolismo , Recambio Mitocondrial , Neostriado/metabolismo , Animales , Autopsia , Núcleo Celular/metabolismo , Daño del ADN , ADN Mitocondrial/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Expresión Génica , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Peróxido de Hidrógeno/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mutación , Neostriado/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Cultivo Primario de Células
18.
Pharmacol Res ; 65(5): 523-30, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22402395

RESUMEN

The anti-inflammatory properties of the flavonol quercetin have been intensively investigated using in vitro cell systems and are to a great extent reflected by changes in the expression of inflammatory markers. However, information relating to the degree at which quercetin affects inflammatory gene expression in vivo is limited. Recently, micro RNAs (miRNAs) have been identified as powerful post-transcriptional gene regulators. The effect of quercetin on miRNA regulation in vivo is largely unknown. Laboratory mice were fed for six weeks with control or quercetin enriched high fat diets and biomarkers of inflammation as well as hepatic levels of miRNAs previously involved in inflammation (miR-125b) and lipid metabolism (miR-122) were determined. We found lower mRNA steady state levels of the inflammatory genes interleukin 6, C-reactive protein, monocyte chemoattractant protein 1, and acyloxyacyl hydrolase in quercetin fed mice. In addition we found evidence for an involvement of redox factor 1, a modulator of nuclear factor κB signalling, on the attenuation of inflammatory gene expression mediated by dietary quercetin. Furthermore, the results demonstrate that hepatic miR-122 and miR-125b concentrations were increased by dietary quercetin supplementation and may therefore contribute to the gene-regulatory activity of quercetin in vivo.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Quercetina/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Proteína C-Reactiva/genética , Hidrolasas de Éster Carboxílico/genética , Quimiocina CCL2/genética , Suplementos Dietéticos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hepatitis Animal/tratamiento farmacológico , Hepatitis Animal/genética , Hepatitis Animal/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-6/genética , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
19.
J Bacteriol ; 193(17): 4495-508, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21665970

RESUMEN

Hydrolytic deamination of cytosine to uracil in DNA is increased in organisms adapted to high temperatures. Hitherto, the uracil base excision repair (BER) pathway has only been described in two archaeons, the crenarchaeon Pyrobaculum aerophilum and the euryarchaeon Archaeoglobus fulgidus, which are hyperthermophiles and use single-nucleotide replacement. In the former the apurinic/apyrimidinic (AP) site intermediate is removed by the sequential action of a 5'-acting AP endonuclease and a 5'-deoxyribose phosphate lyase, whereas in the latter the AP site is primarily removed by a 3'-acting AP lyase, followed by a 3'-phosphodiesterase. We describe here uracil BER by a cell extract of the thermoacidophilic euryarchaeon Thermoplasma acidophilum, which prefers a similar short-patch repair mode as A. fulgidus. Importantly, T. acidophilumcell extract also efficiently executes ATP/ADP-stimulated long-patch BER in the presence of deoxynucleoside triphosphates, with a repair track of ∼15 nucleotides. Supplementation of recombinant uracil-DNA glycosylase (rTaUDG; ORF Ta0477) increased the formation of short-patch at the expense of long-patch repair intermediates, and additional supplementation of recombinant DNA ligase (rTalig; Ta1148) greatly enhanced repair product formation. TaUDG seems to recruit AP-incising and -excising functions to prepare for rapid single-nucleotide insertion and ligation, thus excluding slower and energy-costly long-patch BER.


Asunto(s)
Proteínas Arqueales/metabolismo , Reparación del ADN , Thermoplasma/genética , Uracil-ADN Glicosidasa/metabolismo , Adenosina Difosfato/genética , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Proteínas Arqueales/genética , ADN Ligasas/genética , ADN Ligasas/metabolismo , ADN Recombinante , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Electroforesis en Gel de Poliacrilamida , Regulación de la Expresión Génica Arqueal , Genes Arqueales , Liasas de Fósforo-Oxígeno/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Thermoplasma/enzimología , Uracil-ADN Glicosidasa/genética
20.
Pharm Res ; 27(6): 1115-27, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20309614

RESUMEN

PURPOSE: Genistein, the major bioactive isoflavone of soybeans, acts as a radiosensitizer for prostate cancer (PCa) both in vitro and in vivo. However, pure genistein promoted increased metastasis to lymph nodes. A mixture of soy isoflavones (genistein, daidzein, glycitein) did not cause increased metastasis, but potentiated radiotherapy. We tested whether daidzein could negate genistein-induced metastasis. METHODS: Mice bearing PC-3 prostate tumors were treated with daidzein, genistein or both, and with tumor irradiation. Primary tumors and metastases were evaluated. The effects of each isoflavone and soy were compared in vitro using PC-3 (AR-) and C4-2B (AR+) androgen-independent PCa cell lines. RESULTS: Daidzein did not increase metastasis to lymph nodes and acted as a radiosensitizer for prostate tumors. Daidzein inhibited cell growth and enhanced radiation in vitro but at doses higher than genistein or soy. Daidzein caused milder effects on inhibition of expression and/or activities of APE1/Ref-1, HIF-1alpha and NF-kappaB in PC-3 and C4-2B cells. CONCLUSIONS: Daidzein could be the component of soy that protects against genistein-induced metastasis. Daidzein inhibited cell growth and synergized with radiation, affecting APE1/Ref-1, NF-kappaB and HIF-1alpha, but at lower levels than genistein and soy, in AR+ and AR- PCa cells, suggesting it is an AR-independent mechanism.


Asunto(s)
Antineoplásicos/uso terapéutico , Genisteína/efectos adversos , Glycine max , Isoflavonas/uso terapéutico , Extractos Vegetales/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/efectos adversos , Andrógenos/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , ADN/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Ganglios Linfáticos/efectos de la radiación , Masculino , Ratones , FN-kappa B/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/secundario , Glycine max/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA