Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(7): 102123, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35697073

RESUMEN

High levels of redox enzymes have been commonly observed in various types of human cancer, although whether and how the enzymes contribute to cancer malignancy and therapeutic resistance have yet to be understood. Peroxiredoxin IV (Prx4) is an antioxidant with bona fide peroxidase and molecular chaperone functions. Here, we report that Prx4 is highly expressed in prostate cancer patient specimens, as well as established prostate cancer cell lines, and that its levels can be further stimulated through the activation of androgen receptor signaling. We used lentivirus-mediated shRNA knockdown and CRISPR-Cas9 based KO techniques to establish Prx4-depleted prostate cancer cells, which showed delayed cell cycle progression, reduced rate of cell proliferation, migration, and invasion compared to control cells. In addition, we used proteome profiler phosphokinase arrays to identify signaling changes in Prx4-depleted cells; we found that loss of Prx4 results in insufficient phosphorylation of both Akt and its downstream kinase GSK3α/ß. Moreover, we demonstrate that Prx4-depleted cells are more sensitive to ionizing radiation as they display compromised ability to scavenge reactive oxygen species and increased accumulation of DNA damage. In mouse xenograft models, we show depletion of Prx4 leads to significant suppression of tumor growth, and tumors formed by Prx4-depleted cells respond more effectively to radiation therapy. Our findings suggest that increased levels of Prx4 contribute to the malignancy and radioresistance of prostate cancer through the activation of Akt/GSK3 signaling pathways. Therefore, strategies targeting Prx4 may be utilized to potentially inhibit tumor growth and overcome radioresistance in prostate cancer.


Asunto(s)
Neoplasias de la Próstata , Proteínas Proto-Oncogénicas c-akt , Animales , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Masculino , Ratones , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/radioterapia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
2.
Mol Carcinog ; 56(2): 325-336, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27148961

RESUMEN

Apurinic/apyrimidinic endonuclease 1 (APE1) is an essential protein crucial for repair of oxidized DNA damage not only in genomic DNA but also in mitochondrial DNA. Parkin, a tumor suppressor and Parkinson's disease (PD) associated gene, is an E3 ubiquitin ligase crucial for mitophagy. Although DNA damage is known to induce mitochondrial stress, Parkin's role in regulating DNA repair proteins has not been elucidated. In this study, we examined the possibility of Parkin-dependent ubiquitination of APE1. Ectopically expressed APE1 was degraded by Parkin and PINK1 via polyubiquitination in mouse embryonic fibroblast cells. PD-causing mutations in Parkin and PINK1 abrogated APE1 ubiquitination. Interaction of APE1 with Parkin was observed by co-immunoprecipitation, proximity ligation assay, and co-localization in the cytoplasm. N-terminal deletion of 41 amino acid residues in APE1 significantly reduced the Parkin-dependent APE1 degradation. These results suggested that Parkin directly ubiquitinated N-terminal Lys residues in APE1 in the cytoplasm. Modulation of Parkin and PINK1 activities under mitochondrial or oxidative stress caused moderate but statistically significant decrease of endogenous APE1 in human cell lines including SH-SY5Y, HEK293, and A549 cells. Analyses of glioblastoma tissues showed an inverse relation between the expression levels of APE1 and Parkin. These results suggest that degradation of endogenous APE1 by Parkin occur when cells are stressed to activate Parkin, and imply a role of Parkin in maintaining the quality of APE1, and loss of Parkin may contribute to elevated APE1 levels in glioblastoma. © 2016 Wiley Periodicals, Inc.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Células A549 , Reparación del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/análisis , Células HEK293 , Humanos , Mapas de Interacción de Proteínas , Proteínas Quinasas/análisis , Proteínas Quinasas/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/análisis
3.
BMC Genomics ; 17(1): 983, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27903243

RESUMEN

BACKGROUND: The COP9 signalosome, composed of eight subunits, is implicated in cancer genetics with its deneddylase activity to modulate cellular concentration of oncogenic proteins such as IkB and TGFß. However, its function in the normal cell physiology remains elusive. Primarily focusing on gene expression data of the normal tissues of the head and neck, the cancer genome atlas (TCGA) database was used to identify groups of genes that were expressed synergistically with the COP9 genes, particularly with the COPS5 (CSN5), which possesses the catalytic activity of COP9. RESULTS: Expressions of seven of the COP9 genes (COPS2, COPS3, COPS4, COPS5, COPS6, COPS7A, and COPS8) were found to be highly synergistic in the normal tissues. In contrast, the tumor tissues decreased the coordinated expression pattern of COP9 genes. Pathway analysis revealed a high coordination of the expression of the COPS5 and the other COP9 genes with mitochondria-related functional pathways, including genes encoding the respiratory chain complex. CONCLUSIONS: The results indicate that mRNA expression data for the matched normal tissues available in TCGA are statistically reliable, and are highly useful to assess novel associations of genes with functional pathways in normal physiology as well as in the cancer tissues. This study revealed the significant correlation between the expressions of the COP9 genes and those related to the mitochondrial activity.


Asunto(s)
Complejos Multiproteicos/genética , Neoplasias/genética , ARN/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Adulto , Anciano , Anciano de 80 o más Años , Complejo del Señalosoma COP9 , Mapeo Cromosómico , Bases de Datos Genéticas , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Persona de Mediana Edad , Boca/metabolismo , Complejos Multiproteicos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , ARN/aislamiento & purificación , Sitio de Iniciación de la Transcripción , Transcriptoma
4.
J Biol Chem ; 289(27): 18943-56, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24849605

RESUMEN

Over 70 different missense mutations, including a dominant mutation, in RPE65 retinoid isomerase are associated with distinct forms of retinal degeneration; however, the disease mechanisms for most of these mutations have not been studied. Although some mutations have been shown to abolish enzyme activity, the molecular mechanisms leading to the loss of enzymatic function and retinal degeneration remain poorly understood. Here we show that the 26 S proteasome non-ATPase regulatory subunit 13 (PSMD13), a newly identified negative regulator of RPE65, plays a critical role in regulating pathogenicity of three mutations (L22P, T101I, and L408P) by mediating rapid degradation of mutated RPE65s via a ubiquitination- and proteasome-dependent non-lysosomal pathway. These mutant RPE65s were misfolded and formed aggregates or high molecular complexes via disulfide bonds. Interaction of PSMD13 with mutant RPE65s promoted degradation of misfolded but not properly folded mutant RPE65s. Many mutations, including L22P, T101I, and L408P, were mapped on non-active sites. Although their activities were very low, these mutant RPE65s were catalytically active and could be significantly rescued at low temperature, whereas mutant RPE65s with a distinct active site mutation could not be rescued under the same conditions. Sodium 4-phenylbutyrate and glycerol displayed a significant synergistic effect on the low temperature rescue of the mutant RPE65s by promoting proper folding, reducing aggregation, and increasing membrane association. Our results suggest that a low temperature eye mask and sodium 4-phenylbutyrate, a United States Food and Drug Administration-approved oral medicine, may provide a promising "protein repair therapy" that can enhance the efficacy of gene therapy by reducing the cytotoxic effect of misfolded mutant RPE65s.


Asunto(s)
Dominio Catalítico , Enfermedad/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , cis-trans-Isomerasas/genética , cis-trans-Isomerasas/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glicerol/farmacología , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Fenilbutiratos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Epitelio Pigmentado de la Retina/citología , Temperatura , Ubiquitinación/efectos de los fármacos , cis-trans-Isomerasas/química
5.
J Biol Chem ; 288(12): 8445-8455, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23355472

RESUMEN

Non-coding apurinic/apyrimidinic (AP) sites in DNA form spontaneously and as DNA base excision repair intermediates are the most common toxic and mutagenic in vivo DNA lesion. For repair, AP sites must be processed by 5' AP endonucleases in initial stages of base repair. Human APE1 and bacterial Nfo represent the two conserved 5' AP endonuclease families in the biosphere; they both recognize AP sites and incise the phosphodiester backbone 5' to the lesion, yet they lack similar structures and metal ion requirements. Here, we determined and analyzed crystal structures of a 2.4 Å resolution APE1-DNA product complex with Mg(2+) and a 0.92 Å Nfo with three metal ions. Structural and biochemical comparisons of these two evolutionarily distinct enzymes characterize key APE1 catalytic residues that are potentially functionally similar to Nfo active site components, as further tested and supported by computational analyses. We observe a magnesium-water cluster in the APE1 active site, with only Glu-96 forming the direct protein coordination to the Mg(2+). Despite differences in structure and metal requirements of APE1 and Nfo, comparison of their active site structures surprisingly reveals strong geometric conservation of the catalytic reaction, with APE1 catalytic side chains positioned analogously to Nfo metal positions, suggesting surprising functional equivalence between Nfo metal ions and APE1 residues. The finding that APE1 residues are positioned to substitute for Nfo metal ions is supported by the impact of mutations on activity. Collectively, the results illuminate the activities of residues, metal ions, and active site features for abasic site endonucleases.


Asunto(s)
Proteínas Bacterianas/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , Desoxirribonucleasa IV (Fago T4-Inducido)/química , Thermotoga maritima/enzimología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , ADN/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Escherichia coli , Humanos , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , Unión Proteica , Estructura Secundaria de Proteína , Homología Estructural de Proteína
6.
Cells ; 13(2)2024 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-38247803

RESUMEN

Cancer-specific CD8+ cytotoxic T cells play important roles in preventing cancer growth, and IFN-γ, in addition to IL-12 and type I interferon, is critical for activating CD8+ cytotoxic T cells. We recently identified the capability of the amino-terminus region of dense granule protein 6 (GRA6Nt) of Toxoplasma gondii, an intracellular protozoan parasite, to activate IFN-γ production of microglia, a tissue-resident macrophage population. Therefore, in the present study, we examined whether recombinant GRA6Nt protein (rGRA6Nt) functions as an effective adjuvant to potently activate cancer-specific protective immunity using a murine model of MC38 colorectal cancer (CRC). When mice were immunized with non-replicable (either treated with mitomycin C or irradiated by X-ray) MC38 CRC cells in combination with rGRA6Nt adjuvant and received a challenge implantation of replication-capable MC38 tumor cells, those mice markedly inhibited the growth of the implanted tumors in association with a two-fold increase in CD8+ T cell density within the tumors. In addition, CD8+ T cells of the immunized mice secreted significantly increased amounts of granzyme B, a key mediator of the cytotoxic activity of CD8+ T cells, and IFN-γ in response to MC38 CRC cells in vitro when compared to the T cells from unimmunized mice. Notably, the protective effects of the immunization were specific to MC38 CRC cells, as the immunized mice did not exhibit a significantly inhibited growth of EL4 lymphoma tumors. These results indicate that rGRA6Nt is a novel and effective protein adjuvant when used in immunizations with non-replicable cancer cells to potently activate the protective immunity specifically against the cancer cells employed in the immunization.


Asunto(s)
Neoplasias Colorrectales , Parásitos , Animales , Ratones , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Inmunización , Adyuvantes Inmunológicos/farmacología , Adyuvantes Farmacéuticos
7.
Cancer Lett ; 596: 216993, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38801884

RESUMEN

Ribonucleotide Reductase (RNR) is a rate-limiting enzyme in the production of deoxyribonucleoside triphosphates (dNTPs), which are essential substrates for DNA repair after radiation damage. We explored the radiosensitization property of RNR and investigated a selective RRM2 inhibitor, 3-AP, as a radiosensitizer in the treatment of metastatic pNETs. We investigated the role of RNR subunit, RRM2, in pancreatic neuroendocrine (pNET) cells and responses to radiation in vitro. We also evaluated the selective RRM2 subunit inhibitor, 3-AP, as a radiosensitizer to treat pNET metastases in vivo. Knockdown of RNR subunits demonstrated that RRM1 and RRM2 subunits, but not p53R3, play significant roles in cell proliferation. RRM2 inhibition activated DDR pathways through phosphorylation of ATM and DNA-PK protein kinases but not ATR. RRM2 inhibition also induced Chk1 and Chk2 phosphorylation, resulting in G1/S phase cell cycle arrest. RRM2 inhibition sensitized pNET cells to radiotherapy and induced apoptosis in vitro. In vivo, we utilized pNET subcutaneous and lung metastasis models to examine the rationale for RNR-targeted therapy and 3-AP as a radiosensitizer in treating pNETs. Combination treatment significantly increased apoptosis of BON (human pNET) xenografts and significantly reduced the burden of lung metastases. Together, our results demonstrate that selective RRM2 inhibition induced radiosensitivity of metastatic pNETs both in vitro and in vivo. Therefore, treatment with the selective RRM2 inhibitor, 3-AP, is a promising radiosensitizer in the therapeutic armamentarium for metastatic pNETs.


Asunto(s)
Apoptosis , Proliferación Celular , Ratones Desnudos , Neoplasias Pancreáticas , Tolerancia a Radiación , Fármacos Sensibilizantes a Radiaciones , Ribonucleósido Difosfato Reductasa , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Ribonucleósido Difosfato Reductasa/genética , Ribonucleósido Difosfato Reductasa/antagonistas & inhibidores , Ribonucleósido Difosfato Reductasa/metabolismo , Animales , Línea Celular Tumoral , Fármacos Sensibilizantes a Radiaciones/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tolerancia a Radiación/efectos de los fármacos , Fosforilación , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/radioterapia , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/enzimología , Tumores Neuroendocrinos/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Ratones , Quinasa de Punto de Control 2/metabolismo , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/antagonistas & inhibidores , Femenino , Interferencia de ARN , Proteína Quinasa Activada por ADN
8.
Nucleic Acids Res ; 39(18): 8017-28, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21727086

RESUMEN

Apurinic/apyrimidinic endonuclease-1 (APE1) is a multifunctional DNA repair/gene regulatory protein in mammalian cells, and was recently reported to be phosphorylated at Thr233 by CDK5. We here report that ubiquitination of T233E APE1, a mimicry of phospho-T233 APE1, was markedly increased in multiple cell lines. Expression of CDK5 enhanced monoubiquitination of endogenous APE1. Polyubiquitinated APE1 was decreased when K48R ubiquitin was expressed, suggesting that polyubiquitination was mediated mainly through Lys48 of ubiquitin. The ubiquitination activity of MDM2, consistent in its role for APE1 ubiquitination, was increased for T233E APE1 compared to the wild-type APE1. In mouse embryonic fibroblasts lacking the MDM2 gene, ubiquitination of T233E APE1 was still observed probably because of the decreased degradation activity for monoubiquitinated APE1 and because of backup E3 ligases in the cells. Monoubiquitinated APE1 was present in the nucleus, and analyzing global gene expression profiles with or without induction of a ubiquitin-APE1 fusion gene suggested that monoubiquitination enhanced the gene suppression activity of APE1. These data reveal a delicate balance of ubiquitination and phosphorylation activities that alter the gene regulatory function of APE1.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Ubiquitinación , Sustitución de Aminoácidos , Animales , Línea Celular , Línea Celular Tumoral , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Ácido Glutámico/genética , Humanos , Lisina/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Treonina/genética
9.
Methods Mol Biol ; 2701: 231-242, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37574486

RESUMEN

Cells experience increased genome instability through the course of disease development including cancer initiation and progression. Point mutations, insertion/deletions, translocations, and amplifications of both coding and noncoding regions all contribute to cancer phenotypes. Copy number variation (CNV), i.e., changes of the number of copies of nuclear DNA, occurs in the genome of even normal somatic cells. Studies to understand the effects of CNV on tumor development, especially aspects concerning tumor aggressiveness and the influence on outcomes of therapeutic modalities, have been reignited by the breakthrough technologies of the molecular genomics. This section discusses the significance of analyzing CNVs that cause simultaneous increase/decrease of clusters of genes, using the expression profile of XRCC1 with its neighbor genes LIG1, PNKP, and POLD1 as an example. Methods for CNV assay at the individual gene level on formalin-fixed, paraffin-embedded (FFPE) tissues using the NanoString nCounter technology will then be described.


Asunto(s)
Variaciones en el Número de Copia de ADN , ADN , Genómica , Mutación INDEL , Reparación del ADN/genética , Adhesión en Parafina
10.
Cells ; 12(20)2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37887269

RESUMEN

Genome instability in cancer cells causes not only point mutations but also structural variations of the genome, including copy number variations (CNVs). It has recently been proposed that CNVs arise in cancer to adapt to a given microenvironment to survive. However, how CNV influences cellular resistance against ionizing radiation remains unknown. PRMT5 (protein arginine methyltransferase 5) and APE1 (apurinic/apyrimidinic endonuclease 1), which enhance repair of DNA double-strand breaks and oxidative DNA damage, are closely localized in the chromosome 14 of the human genome. In this study, the genomics data for the PRMT5 and APE1 genes, including their expression, CNVs, and clinical outcomes, were analyzed using TCGA's data set for oral squamous cell carcinoma patients. The two genes were found to share almost identical CNV values among cancer tissues from oral squamous cell carcinoma (OSCC) patients. Levels of expression of PRMT5 and APE1 in OSCC tissues are highly correlated in cancer but not in normal tissues, suggesting that regulation of PRMT5 and APE1 were overridden by the extent of CNV in the PRMT5-APE1 genome region. High expression levels of PRMT5 and APE1 were both associated with poor survival outcomes after radiation therapy. Simultaneous down-regulation of PRMT5 and APE1 synergistically hampered DNA double-strand break repair and sensitized OSCC cell lines to X-ray irradiation in vitro and in vivo. These results suggest that the extent of CNV in a particular genome region significantly influence the radiation resistance of cancer cells. Profiling CNV in the PRMT5-APE1 genome region may help us to understand the mechanism of the acquired radioresistance of tumor cells, and raises the possibility that simultaneous inhibition of PRMT5 and APE1 may increase the efficacy of radiation therapy.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas de Cabeza y Cuello , Variaciones en el Número de Copia de ADN/genética , Neoplasias de la Boca/genética , Neoplasias de la Boca/radioterapia , Línea Celular Tumoral , Radiación Ionizante , Tolerancia a Radiación/genética , ADN , Microambiente Tumoral , Proteína-Arginina N-Metiltransferasas/genética
11.
Cancers (Basel) ; 15(24)2023 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-38136418

RESUMEN

BACKGROUND: Magnetic resonance imaging (MRI) is a non-invasive imaging modality which, in conjunction with biopsies, provide a qualitative assessment of tumor response to treatment. Intravenous injection of contrast agents such as fluorine (19F) nanoemulsions labels systemic macrophages, which can, then, be tracked in real time with MRI. This method can provide quantifiable insights into the behavior of tumor-associated macrophages (TAMs) in the tumor microenvironment and macrophage recruitment during therapy. METHODS: Female mice received mammary fat pad injections of murine breast or colon cancer cell lines. The mice then received an intravenous 19F nanoemulsion injection, followed by a baseline 19F MRI. For each cancer model, half of the mice then received 8 Gy of localized radiation therapy (RT), while others remained untreated. The mice were monitored for two weeks for tumor growth and 9F signal using MRI. RESULTS: Across both cohorts, the RT-treated groups presented significant tumor growth reduction or arrest, contrary to the untreated groups. Similarly, the fluorine signal in treated groups increased significantly as early as four days post therapy. The fluorine signal change correlated to tumor volumes irrespective of time. CONCLUSION: These results demonstrate the potential of 19F MRI to non-invasively track macrophages during radiation therapy and its prognostic value with regard to tumor growth.

12.
Antioxid Redox Signal ; 38(1-3): 183-197, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35754343

RESUMEN

Aims: Though best known for its role in oxidative DNA damage repair, apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that regulates multiple host responses during oxidative stress, including the reductive activation of transcription factors. As knockout of the APE1-encoding gene, Apex1, is embryonically lethal, we sought to create a viable model with generalized inhibition of APE1 expression. Results: A hypomorphic (HM) mouse with decreased APE1 expression throughout the body was generated using a construct containing a neomycin resistance (NeoR) cassette knocked into the Apex1 site. Offspring were assessed for APE1 expression, breeding efficiency, and morphology with a focused examination of DNA damage in the stomach. Heterozygotic breeding pairs yielded 50% fewer HM mice than predicted by Mendelian genetics. APE1 expression was reduced up to 90% in the lungs, heart, stomach, and spleen. The HM offspring were typically smaller, and most had a malformed tail. Oxidative DNA damage was increased spontaneously in the stomachs of HM mice. Further, all changes were reversed when the NeoR cassette was removed. Primary gastric epithelial cells from HM mice differentiated more quickly and had more evidence of oxidative DNA damage after stimulation with Helicobacter pylori or a chemical carcinogen than control lines from wildtype mice. Innovation: A HM mouse with decreased APE1 expression throughout the body was generated and extensively characterized. Conclusion: The results suggest that HM mice enable studies of APE1's multiple functions throughout the body. The detailed characterization of the stomach showed that gastric epithelial cells from HM were more susceptible to DNA damage. Antioxid. Redox Signal. 38, 183-197.


Asunto(s)
Reparación del ADN , Estrés Oxidativo , Ratones , Animales , Daño del ADN , Oxidación-Reducción , Modelos Animales de Enfermedad , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Estómago , Endonucleasas/genética , Endonucleasas/metabolismo
13.
Brain Res ; 1782: 147840, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35183524

RESUMEN

Cranial radiation is important for treating both primary brain tumors and brain metastases. A potential delayed side effect of cranial radiation is neurocognitive function decline. Early detection of CNS injury might prevent further neuronal damage. Extracellular vesicles (EVs) have emerged as a potential diagnostic tool because of their unique membranous characteristics and cargos. We investigated whether EVs can be an early indicator of CNS injury by giving C57BJ/6 mice 10 Gy cranial IR. EVs were isolated from sera to quantify: 1) number of EVs using nanoparticle tracking analysis (NTA); 2) Glial fibrillary acidic protein (GFAP), an astrocyte marker; and 3) protein-bound 4-hydroxy-2-nonenal (HNE) adducts, an oxidative damage marker. Brain tissues were prepared for immunohistochemistry staining and protein immunoblotting. The results demonstrate: 1) increased GFAP levels (p < 0.05) in EVs, but not brain tissue, in the IR group; and 2) increased HNE-bound protein adduction levels (p < 0.05). The results support using EVs as an early indicator of cancer therapy-induced neuronal injury.


Asunto(s)
Lesiones Encefálicas , Vesículas Extracelulares , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Vesículas Extracelulares/metabolismo , Ratones , Neuronas/metabolismo , Proteínas/metabolismo
14.
Cell Mol Life Sci ; 67(21): 3609-20, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20711647

RESUMEN

A key issue in studying mammalian DNA base excision repair is how its component proteins respond to a plethora of cell-signaling mediators invoked by DNA damage and stress-inducing agents such as reactive oxygen species, and how the actions of individual BER proteins are attributed to cell survival or apoptotic/necrotic death. This article reviews the past and recent progress on posttranslational modification (PTM) of mammalian apurinic/apyrimidinic (AP) endonuclease 1 (APE1).


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Procesamiento Proteico-Postraduccional , Acetilación , Animales , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Humanos , Modelos Moleculares , Fosforilación , Ubiquitinación
15.
Nucleic Acids Res ; 37(12): 3946-58, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19401441

RESUMEN

Endonucleolytic cleavage of the coding region determinant (CRD) of c-myc mRNA appears to play a critical role in regulating c-myc mRNA turnover. Using (32)P-labeled c-myc CRD RNA as substrate, we have purified and identified two endoribonucleases from rat liver polysomes that are capable of cleaving the transcript in vitro. A 17-kDa enzyme was identified as RNase1. Apurinic/apyrimidinic (AP) DNA endonuclease 1 (APE1) was identified as the 35-kDa endoribonuclease that preferentially cleaves in between UA and CA dinucleotides of c-myc CRD RNA. APE1 was further confirmed to be the 35-kDa endoribonuclease because: (i) the endoribonuclease activity of the purified 35-kDa native enzyme was specifically immuno-depleted with APE1 monoclonal antibody, and (ii) recombinant human APE1 generated identical RNA cleavage patterns as the native liver enzyme. Studies using E96A and H309N mutants of APE1 suggest that the endoribonuclease activity for c-myc CRD RNA shares the same active center with the AP-DNA endonuclease activity. Transient knockdown of APE1 in HeLa cells led to increased steady-state level of c-myc mRNA and its half-life. We conclude that the ability to cleave RNA dinucleotides is a previously unidentified function of APE1 and it can regulate c-myc mRNA level possibly via its endoribonuclease activity.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endorribonucleasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/metabolismo , Animales , Anticuerpos Monoclonales , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/inmunología , Endorribonucleasas/química , Endorribonucleasas/aislamiento & purificación , Células HeLa , Humanos , Hígado/enzimología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Ratas , Proteínas Recombinantes/metabolismo , Ribonucleasa Pancreática/clasificación
16.
Med Phys ; 48(2): 881-889, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33283893

RESUMEN

PURPOSE: Accurate radiation dosimetry in radiobiological experiments is crucial for preclinical research in advancement of cancer treatment. Vendors of cell irradiators often perform calibration for end-users. However, calibration accuracy remains unclear due to missing detailed information on calibration equipment and procedures. In this study, we report our findings of a vender miscalibration of the radiation output and our investigation on the root cause of the discrepancy. METHODS: Independent calibration verification for a commercial preclinical orthovoltage irradiator was conducted. Initially, in the absence of ionization chambers calibrated at kV energy, radiochromic films (EBT3) was first calibrated at MV energy. Energy correction factors from literature were used to create an in-house kV dosimetry system. The miscalibration identified with the in-house kV EBT3 dosimetry was later confirmed by ADCL calibrated ionization chambers (Exradin A1SL and PTW 30013) at kV energy. Ionization chambers were suspended in-air following TG-61 recommendation for output calibration. To investigate the root cause of the miscalibration, additional measurements were performed with ionization chambers placed on the shelf. A validated Monte Carlo simulation code was also used to investigate the impact of placing the ionization chamber on the shelf instead of suspending it in air during the vendor-performed calibration process. RESULTS: Up to a 6% dosimetry error was observed when comparing the vendor calibrated output of the preclinical irradiator with our independent calibration check. Further investigation showed incorrect setups in the vendor's calibration procedure which may result in dose errors up to 11% from the backscatter of the shelf board during calibration, and up to 5% from omitting temperature and pressure corrections to ionization chamber readings. CONCLUSION: Our study revealed large dose calibration errors caused by incorrect setup and the omission of temperature/pressure correction in the vendor's calibration procedure. The findings also highlighted the importance of performing an independent check of the dose calibration for preclinical kV irradiators. More absolute dosimetry training is needed for both vendors and end users for establishing accurate absolute dosimetry.


Asunto(s)
Radiometría , Calibración , Método de Montecarlo
17.
Cells ; 10(5)2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34065268

RESUMEN

Patients with advanced-stage gastroenteropancreatic neuroendocrine tumors (GEP-NETs) have a poor overall prognosis despite chemotherapy and radiotherapy (e.g., peptide receptor radionuclide therapy (PRRT)). Better treatment options are needed to improve disease regression and patient survival. The purpose of this study was to examine a new treatment strategy by combining PI3K/mTOR dual inhibition and radiotherapy. First, we assessed the efficacy of two PI3K/mTOR dual inhibitors, PF-04691502 and PKI-402, to inhibit pAkt and increase apoptosis in NET cell lines (BON and QGP-1) and patient-derived tumor spheroids as single agents or combined with radiotherapy (XRT). Treatment with PF-04691502 decreased pAkt (Ser473) expression for up to 72 h compared with the control; in contrast, decreased pAkt expression was noted for less than 24 h with PKI-402. Simultaneous treatment with PF-04691502 and XRT did not induce apoptosis in NET cells; however, the addition of PF-04691502 48 h after XRT significantly increased apoptosis compared to PF-04691502 or XRT treatment alone. Our results demonstrate that schedule-dependent administration of a PI3K/mTOR inhibitor, combined with XRT, can enhance cytotoxicity by promoting the radiosensitivity of NET cells. Moreover, our findings suggest that radiotherapy, in combination with timed PI3K/mTOR inhibition, may be a promising therapeutic regimen for patients with GEP-NET.


Asunto(s)
Neoplasias Intestinales/tratamiento farmacológico , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/química , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Piridonas/farmacología , Pirimidinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Apoptosis , Proliferación Celular , Humanos , Neoplasias Intestinales/patología , Neoplasias Intestinales/radioterapia , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/radioterapia , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Neoplasias Gástricas/patología , Neoplasias Gástricas/radioterapia , Células Tumorales Cultivadas
18.
Nucleic Acids Res ; 36(5): 1555-66, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18208837

RESUMEN

The human AP-endonuclease (APE1/Ref-1), an essential multifunctional protein, plays a central role in the repair of oxidative base damage via the DNA base excision repair (BER) pathway. The mammalian AP-endonuclease (APE1) overexpression is often observed in tumor cells, and confers resistance to various anticancer drugs; its downregulation sensitizes tumor cells to those agents via induction of apoptosis. Here we show that wild type (WT) but not mutant p53 negatively regulates APE1 expression. Time-dependent decrease was observed in APE1 mRNA and protein levels in the human colorectal cancer line HCT116 p53(+/+), but not in the isogenic p53 null mutant after treatment with camptothecin, a DNA topoisomerase I inhibitor. Furthermore, ectopic expression of WTp53 in the p53 null cells significantly reduced both endogenous APE1 and APE1 promoter-dependent luciferase expression in a dose-dependent fashion. Chromatin immunoprecipitation assays revealed that endogenous p53 is bound to the APE1 promoter region that includes a Sp1 site. We show here that WTp53 interferes with Sp1 binding to the APE1 promoter, which provides a mechanism for the downregulation of APE1. Taken together, our results demonstrate that WTp53 is a negative regulator of APE1 expression, so that repression of APE1 by p53 could provide an additional pathway for p53-dependent induction of apoptosis in response to DNA damage.


Asunto(s)
Daño del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Humanos , Mutación , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Elementos de Respuesta , Factor de Transcripción Sp1/metabolismo , Proteína p53 Supresora de Tumor/genética
19.
Oral Oncol ; 111: 104949, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32801084

RESUMEN

OBJECTIVES: Recurrence rates for head and neck squamous cell carcinoma (HNSCC) approach 50% at 5 years. Current staging fails to identify patients with a worse prognosis who might benefit from intensified treatment, which warrants improved prognostic biomarkers. The purpose of this retrospective case study is to identify potential prognostic biomarkers in patients with HNSCC including APE1 (DNA repair/redox gene regulator), NRF2 and PPARGC1A (redox gene regulators), SOD3 and DCN (antioxidant proteins). MATERIALS AND METHODS: Differential protein expression between benign, carcinoma in situ (CIS), and invasive HNSCC tissue specimens from 77 patients was assessed using immunohistochemistry. Protein expression was analyzed with multivariate, pair-wise, and Kaplan-Meier survival analyses to identify potential prognostic biomarkers. Utilizing The Cancer Genome Atlas's transcriptome database, pair-wise and survival analysis was performed to identify potential prognostic biomarkers. RESULTS: APE1, NRF2, PPARGC1A, SOD3, and DCN expression in HNSCC in relation to, lymph node invasion, and patient survival were examined. Elevated APE1 protein expression in CIS corresponded with reduced survival (p = 0.0243). Increased APE1 gene expression in stage T4a HNSCC was associated with reduced patient survival (p < 0.015). Increased PPARGC1A in invasive tumor correlated with reduced survival (p = 0.0281). Patients with lymph node invasion at diagnosis had significantly increased APE1 protein in the primary sites (p < 0.05). Patients with poorly differentiated invasive tumors had reduced PPARGC1A in CIS proximal to the invasive tumor and had elevated DCN and SOD3 in proximal benign tissue (p < 0.05). CONCLUSIONS: The expression of APE1, DCN, and SOD3 is a potential prognostic signature that identifies patients with worsened survival.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Decorina/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Superóxido Dismutasa/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma in Situ/metabolismo , Carcinoma in Situ/mortalidad , Carcinoma in Situ/patología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Bases de Datos Genéticas , Decorina/genética , Femenino , Neoplasias de Cabeza y Cuello/mortalidad , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Ganglios Linfáticos/patología , Masculino , Persona de Mediana Edad , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Recurrencia Local de Neoplasia/mortalidad , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Pronóstico , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Superóxido Dismutasa/genética , Transcriptoma
20.
Phys Med Biol ; 64(5): 055001, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30673636

RESUMEN

The orthovoltage x-ray energy frequently used in radiation research is prone to dosimetry errors due to insufficient backscatter conditions. In many radiobiology studies, especially for cell irradiations, precise dose calculation algorithms such as Convolution-Superposition or Monte Carlo are impractical and as such, less accurate hand calculation methods are used for dose estimation. These dose estimation methods typically assume full backscatter conditions. The purpose of this study is to demonstrate the magnitude of the dose error that results from insufficient backscatter, and to provide lookup tables to account this issue. The beam spectra of several widely used commercial systems (XRAD-225, XRAD-320, SARRP) were used in Monte Carlo (MC) simulations on a series of phantom setups to investigate the impact of varying backscatter conditions on dosimetry. The depth dose curves for different field sizes, water phantom thicknesses and beam qualities were generated. In addition, depth dependent backscatter factors for different field sizes and different beam qualities were calculated. It is demonstrated that as much as a 50% dose difference exists for different backscatter conditions at the beam qualities studied. The choice of cell dish size as well as other changes in the experiment setup can have more than 10% impact on the dose. The impact of backscatter is reduced with a decrease in field size. Further, the thickness needed to provide full backscatter can be approximated as being equal to the field size. It is imperative to ensure full backscatter conditions during system and dosimeter calibration, or to use the look-up table provided in this study.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA