Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 12(7): e1006107, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27437695

RESUMEN

XRN2 is a 5'-3' exoribonuclease implicated in transcription termination. Here we demonstrate an unexpected role for XRN2 in the DNA damage response involving resolution of R-loop structures and prevention of DNA double-strand breaks (DSBs). We show that XRN2 undergoes DNA damage-inducible nuclear re-localization, co-localizing with 53BP1 and R loops, in a transcription and R-loop-dependent process. XRN2 loss leads to increased R loops, genomic instability, replication stress, DSBs and hypersensitivity of cells to various DNA damaging agents. We demonstrate that the DSBs that arise with XRN2 loss occur at transcriptional pause sites. XRN2-deficient cells also exhibited an R-loop- and transcription-dependent delay in DSB repair after ionizing radiation, suggesting a novel role for XRN2 in R-loop resolution, suppression of replication stress, and maintenance of genomic stability. Our study highlights the importance of regulating transcription-related activities as a critical component in maintaining genetic stability.


Asunto(s)
Daño del ADN , Replicación del ADN , Exorribonucleasas/genética , Exorribonucleasas/fisiología , Transcripción Genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Antineoplásicos/química , Núcleo Celular/metabolismo , ADN Helicasas , Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Genómica , Células HeLa , Humanos , Microscopía Fluorescente , Enzimas Multifuncionales , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Plásmidos/metabolismo , ARN Helicasas/metabolismo , ARN Interferente Pequeño/metabolismo
2.
J Biol Chem ; 292(44): 18203-18216, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-28916726

RESUMEN

Many cancer treatments, such as those for managing recalcitrant tumors like pancreatic ductal adenocarcinoma, cause off-target toxicities in normal, healthy tissue, highlighting the need for more tumor-selective chemotherapies. ß-Lapachone is bioactivated by NAD(P)H:quinone oxidoreductase 1 (NQO1). This enzyme exhibits elevated expression in most solid cancers and therefore is a potential cancer-specific target. ß-Lapachone's therapeutic efficacy partially stems from the drug's induction of a futile NQO1-mediated redox cycle that causes high levels of superoxide and then peroxide formation, which damages DNA and causes hyperactivation of poly(ADP-ribose) polymerase, resulting in extensive NAD+/ATP depletion. However, the effects of this drug on energy metabolism due to NAD+ depletion were never described. The futile redox cycle rapidly consumes O2, rendering standard assays of Krebs cycle turnover unusable. In this study, a multimodal analysis, including metabolic imaging using hyperpolarized pyruvate, points to reduced oxidative flux due to NAD+ depletion after ß-lapachone treatment of NQO1+ human pancreatic cancer cells. NAD+-sensitive pathways, such as glycolysis, flux through lactate dehydrogenase, and the citric acid cycle (as inferred by flux through pyruvate dehydrogenase), were down-regulated by ß-lapachone treatment. Changes in flux through these pathways should generate biomarkers useful for in vivo dose responses of ß-lapachone treatment in humans, avoiding toxic side effects. Targeting the enzymes in these pathways for therapeutic treatment may have the potential to synergize with ß-lapachone treatment, creating unique NQO1-selective combinatorial therapies for specific cancers. These findings warrant future studies of intermediary metabolism in patients treated with ß-lapachone.


Asunto(s)
Antineoplásicos/farmacología , Metabolismo Energético/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , Naftoquinonas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Profármacos/farmacología , Activación Metabólica , Antineoplásicos/metabolismo , Biomarcadores/metabolismo , Isótopos de Carbono , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclo del Ácido Cítrico/efectos de los fármacos , Daño del ADN , Inhibidores Enzimáticos/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Metabolómica/métodos , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/metabolismo , Análisis de Componente Principal , Profármacos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
3.
Br J Cancer ; 119(8): 928-936, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30318513

RESUMEN

BACKGROUND: NAD(P)H:quinone oxidoreductase 1 (NQO1) is a two-electron oxidoreductase expressed in multiple tumour types. ARQ 761 is a ß-lapachone (ß-lap) analogue that exploits the unique elevation of NQO1 found in solid tumours to cause tumour-specific cell death. METHODS: We performed a 3+3 dose escalation study of 3 schedules (weekly, every other week, 2/3 weeks) of ARQ 761 in patients with refractory advanced solid tumours. Tumour tissue was analysed for NQO1 expression. After 20 patients were analysed, enrolment was restricted to patients with NQO1-high tumours (H-score ≥ 200). RESULTS: A total of 42 patients were treated. Median number of prior lines of therapy was 4. Maximum tolerated dose was 390 mg/m2 as a 2-h infusion every other week. Dose-limiting toxicity was anaemia. The most common treatment-related adverse events were anaemia (79%), fatigue (45%), hypoxia (33%), nausea (17%), and vomiting (17%). Transient grade 3 hypoxia, reflecting possible methemoglobinaemia, occurred in 26% of patients. Among 32 evaluable patients, best response was stable disease (n = 12); 6 patients had tumour shrinkage. There was a trend towards improved efficacy in NQO1-high tumours (P = 0.06). CONCLUSIONS: ARQ 761 has modest single-agent activity, which appears associated with tumour NQO1 expression. Principal toxicities include anaemia and possible methemoglobinaemia.


Asunto(s)
Apoptosis/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/análisis , NAD(P)H Deshidrogenasa (Quinona)/biosíntesis , Naftoquinonas/uso terapéutico , Necrosis/inducido químicamente , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Naftoquinonas/química , Especies Reactivas de Oxígeno/metabolismo
4.
Nucleic Acids Res ; 44(4): 1718-31, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26819409

RESUMEN

Ku70-binding protein 5 (Kub5)-Hera (K-H)/RPRD1B maintains genetic integrity by concomitantly minimizing persistent R-loops and promoting repair of DNA double strand breaks (DSBs). We used tandem affinity purification-mass spectrometry, co-immunoprecipitation and gel-filtration chromatography to define higher-order protein complexes containing K-H scaffolding protein to gain insight into its cellular functions. We confirmed known protein partners (Ku70, RNA Pol II, p15RS) and discovered several novel associated proteins that function in RNA metabolism (Topoisomerase 1 and RNA helicases), DNA repair/replication processes (PARP1, MSH2, Ku, DNA-PKcs, MCM proteins, PCNA and DNA Pol δ) and in protein metabolic processes, including translation. Notably, this approach directed us to investigate an unpredicted involvement of K-H in DNA mismatch repair (MMR) where K-H depletion led to concomitant MMR deficiency and compromised global microsatellite stability. Mechanistically, MMR deficiency in K-H-depleted cells was a consequence of reduced stability of the core MMR proteins (MLH1 and PMS2) caused by elevated basal caspase-dependent proteolysis. Pan-caspase inhibitor treatment restored MMR protein loss. These findings represent a novel mechanism to acquire MMR deficiency/microsatellite alterations. A significant proportion of colon, endometrial and ovarian cancers exhibit k-h expression/copy number loss and may have severe mutator phenotypes with enhanced malignancies that are currently overlooked based on sporadic MSI+ screening.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Inestabilidad Genómica , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Antígenos Nucleares/genética , Proteínas de Ciclo Celular/genética , Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo I/genética , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Autoantígeno Ku , Complejos Multiproteicos/genética , Proteínas de Neoplasias/genética , Neoplasias/metabolismo , ARN Helicasas/genética , ARN Polimerasa II/genética , Proteínas Represoras/genética
5.
J Surg Oncol ; 116(1): 83-88, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28346693

RESUMEN

Novel, tumor-selective therapies are needed to increase the survival rate of pancreatic cancer patients. K-Ras-mutant-driven NAD(P)H:quinone oxidoreductase 1 (NQO1) is over-expressed in pancreatic tumor versus associated normal tissue, while catalase expression is lowered compared to levels in associated normal pancreas tissue. ARQ761 undergoes a robust, futile redox cycle in NQO1+ cancer cells, producing massive hydrogen peroxide (H2 O2 ) levels; normal tissues are spared by low NQO1 and high catalase expression. DNA damage created by ARQ761 in pancreatic cancer cells "hyperactivates" PARP1, causing metabolic catastrophe and NAD ± keresis cell death. NQO1: catalase levels (high in tumor, low in normal tissue) are an attractive therapeutic window to treat pancreatic cancer. Based on a growing body of literature, we are leading a clinical trial to evaluate the combination of ARQ761 and chemotherapy in patients with pancreatic cancer.


Asunto(s)
NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , Naftoquinonas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Albúminas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ensayos Clínicos Fase I como Asunto , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Humanos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Paclitaxel/farmacología , Neoplasias Pancreáticas/metabolismo , Gemcitabina
6.
PLoS Genet ; 10(6): e1004419, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24922507

RESUMEN

Translesion synthesis (TLS) enables DNA replication through damaged bases, increases cellular DNA damage tolerance, and maintains genomic stability. The sliding clamp PCNA and the adaptor polymerase Rev1 coordinate polymerase switching during TLS. The polymerases Pol η, ι, and κ insert nucleotides opposite damaged bases. Pol ζ, consisting of the catalytic subunit Rev3 and the regulatory subunit Rev7, then extends DNA synthesis past the lesion. Here, we show that Rev7 binds to the transcription factor TFII-I in human cells. TFII-I is required for TLS and DNA damage tolerance. The TLS function of TFII-I appears to be independent of its role in transcription, but requires homodimerization and binding to PCNA. We propose that TFII-I bridges PCNA and Pol ζ to promote TLS. Our findings extend the general principle of component sharing among divergent nuclear processes and implicate TLS deficiency as a possible contributing factor in Williams-Beuren syndrome.


Asunto(s)
Daño del ADN/genética , Reparación del ADN/genética , Replicación del ADN/genética , Proteínas Mad2/metabolismo , Factores de Transcripción TFII/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/biosíntesis , ADN Polimerasa Dirigida por ADN/biosíntesis , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Proteínas Mad2/biosíntesis , Proteínas Mad2/genética , Proteínas Nucleares/biosíntesis , Nucleotidiltransferasas/biosíntesis , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Transcripción TFII/biosíntesis , Factores de Transcripción TFII/metabolismo
7.
Nucleic Acids Res ; 42(8): 4996-5006, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24589584

RESUMEN

Functions of Kub5-Hera (In Greek Mythology Hera controlled Artemis) (K-H), the human homolog of the yeast transcription termination factor Rtt103, remain undefined. Here, we show that K-H has functions in both transcription termination and DNA double-strand break (DSB) repair. K-H forms distinct protein complexes with factors that repair DSBs (e.g. Ku70, Ku86, Artemis) and terminate transcription (e.g. RNA polymerase II). K-H loss resulted in increased basal R-loop levels, DSBs, activated DNA-damage responses and enhanced genomic instability. Significantly lowered Artemis protein levels were detected in K-H knockdown cells, which were restored with specific K-H cDNA re-expression. K-H deficient cells were hypersensitive to cytotoxic agents that induce DSBs, unable to reseal complex DSB ends, and showed significantly delayed γ-H2AX and 53BP1 repair-related foci regression. Artemis re-expression in K-H-deficient cells restored DNA-repair function and resistance to DSB-inducing agents. However, R loops persisted consistent with dual roles of K-H in transcription termination and DSB repair.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Reparación del ADN , Proteínas de Neoplasias/metabolismo , Terminación de la Transcripción Genética , Animales , Antineoplásicos/toxicidad , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Células Cultivadas , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN , Endonucleasas , Inestabilidad Genómica , Humanos , Ratones , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética
8.
Crit Rev Eukaryot Gene Expr ; 24(1): 15-28, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24579667

RESUMEN

Poly (ADP-ribose) polymerases (PARPs) are a family of related enzymes that share the ability to catalyze the transfer of ADP-ribose to target proteins. PARPs play an important role in various cellular processes, including modulation of chromatin structure, transcription, replication, recombination, and DNA repair. The role of PARP proteins in DNA repair is of particular interest, in view of the finding that certain tumors defective in homologous recombination mechanisms, may rely on PARP-mediated DNA repair for survival, and are sensitive to its inhibition. PARP inhibitors may also increase tumor sensitivity to DNA-damaging agents. Clinical trials of PARP inhibitors are investigating the utility of these approaches in cancer. The hyperactivation of PARP has also been shown to result in a specific programmed cell death pathway involving NAD+/ATP depletion, mu-calpain activation, loss of mitochondrial membrane potential, and the release of apoptosis inducing factor. Hyperactivation of the PARP pathway may be exploited to selectively kill cancer cells. Other PARP forms, including tankyrase 1 (PARP 5a), which plays an important role in enhancing telomere elongation by telomerase, have been found to be potential targets in cancer therapy. The PARP pathway and its inhibition thus offers a number of opportunities for therapeutic intervention in both cancer and other disease states.


Asunto(s)
Neoplasias/terapia , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Reparación del ADN , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Humanos , Terapia Molecular Dirigida , Nanomedicina , Naftoquinonas/farmacología , Necrosis/enzimología , Necrosis/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Factores de Transcripción/metabolismo
9.
bioRxiv ; 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38798459

RESUMEN

Background: Triple negative breast cancer (TNBC), characterized by the lack of three canonical receptors, is unresponsive to commonly used hormonal therapies. One potential TNBC-specific therapeutic target is NQO1, as it is highly expressed in many TNBC patients and lowly expressed in non-cancer tissues. DNA damage induced by NQO1 bioactivatable drugs in combination with Rucaparib-mediated inhibition of PARP1-dependent DNA repair synergistically induces cell death. Methods: To gain a better understanding of the mechanisms behind this synergistic effect, we used global proteomics, phosphoproteomics, and thermal proteome profiling to analyze changes in protein abundance, phosphorylation and protein thermal stability. Results: Very few protein abundance changes resulted from single or dual agent treatment; however, protein phosphorylation and thermal stability were impacted. Histone H2AX was among several proteins identified to have increased phosphorylation when cells were treated with the combination of IB-DNQ and Rucaparib, validating that the drugs induced persistent DNA damage. Thermal proteome profiling revealed destabilization of H2AX following combination treatment, potentially a result of the increase in phosphorylation. Kinase substrate enrichment analysis predicted altered activity for kinases involved in DNA repair and cell cycle following dual agent treatment. Further biophysical analysis of these two processes revealed alterations in SWI/SNF complex association and tubulin / p53 interactions. Conclusions: Our findings that the drugs target DNA repair and cell cycle regulation, canonical cancer treatment targets, in a way that is dependent on increased expression of a protein selectively found to be upregulated in cancers without impacting protein abundance illustrate that multi-omics methodologies are important to gain a deeper understanding of the mechanisms behind treatment induced cancer cell death.

10.
Ann Surg Oncol ; 20(6): 2073-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23328973

RESUMEN

BACKGROUND: Perioperative blood transfusion in pancreatic cancer patients is linked to decreased survival; however, a causal mechanism has not been determined. Previously we have shown that the plasma fraction of stored packed red blood cells (pRBCs) promotes pancreas cancer progression and associated morbidity. We hypothesize these untoward effects will be mitigated by use of a hemoglobin-based oxygen carrier (HBOC). METHODS: Cytokines and growth factors were measured in the plasma fraction from stored pRBCs and in an HBOC via cytokine array followed by formal enzyme-linked immunosorbent assay (ELISA). In an immunocompetent murine model, pancreas cancer progression was determined in vivo by bioluminescence, tumor weight, and number of metastases. RESULTS: Elevated levels of epidermal growth factor (EGF), platelet-derived growth factor BB (PDGF-BB), and regulated upon activation, normal T cell expressed and secreted (RANTES) were present in the plasma fraction of stored pRBCs, but were not found in the HBOC. Intravenous delivery of plasma fraction to mice with pancreatic cancer resulted in increased bioluminescence activity compared with mice that received HBOC. Metastatic events and pancreatic primary tumor weights were significantly higher in animals receiving plasma fraction from stored pRBCs compared with animals receiving HBOC. CONCLUSIONS: Intravenous receipt of the acellular plasma fraction of stored pRBCs promotes pancreatic cancer progression in an immunocompetent mouse model. These untoward events are mitigated by use of an HBOC.


Asunto(s)
Sustitutos Sanguíneos/farmacología , Citocinas/farmacología , Transfusión de Eritrocitos , Hemoglobinas/farmacología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Plasma/química , Análisis de Varianza , Animales , Becaplermina , Sustitutos Sanguíneos/química , Sustitutos Sanguíneos/uso terapéutico , Quimiocina CCL5/análisis , Citocinas/análisis , Progresión de la Enfermedad , Factor de Crecimiento Epidérmico/análisis , Transfusión de Eritrocitos/efectos adversos , Hemoglobinas/química , Hemoglobinas/uso terapéutico , Humanos , Ratones , Metástasis de la Neoplasia , Análisis por Matrices de Proteínas , Proteínas Proto-Oncogénicas c-sis/análisis
11.
Proc Natl Acad Sci U S A ; 107(6): 2485-90, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20080667

RESUMEN

A single nucleotide polymorphism in the DAB2IP gene is associated with risk of aggressive prostate cancer (PCa), and loss of DAB2IP expression is frequently detected in metastatic PCa. However, the functional role of DAB2IP in PCa remains unknown. Here, we show that the loss of DAB2IP expression initiates epithelial-to-mesenchymal transition (EMT), which is visualized by repression of E-cadherin and up-regulation of vimentin in both human normal prostate epithelial and prostate carcinoma cells as well as in clinical prostate-cancer specimens. Conversely, restoring DAB2IP in metastatic PCa cells reversed EMT. In DAB2IP knockout mice, prostate epithelial cells exhibited elevated mesenchymal markers, which is characteristic of EMT. Using a human prostate xenograft-mouse model, we observed that knocking down endogenous DAB2IP in human carcinoma cells led to the development of multiple lymph node and distant organ metastases. Moreover, we showed that DAB2IP functions as a scaffold protein in regulating EMT by modulating nuclear beta-catenin/T-cell factor activity. These results show the mechanism of DAB2IP in EMT and suggest that assessment of DAB2IP may provide a prognostic biomarker and potential therapeutic target for PCa metastasis.


Asunto(s)
Células Epiteliales/patología , Mesodermo/patología , Neoplasias de la Próstata/patología , Proteínas Activadoras de ras GTPasa/fisiología , Animales , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Células Epiteliales/metabolismo , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Mesodermo/metabolismo , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción TCF/metabolismo , Transfección , Trasplante Heterólogo , Vimentina/genética , Vimentina/metabolismo , beta Catenina/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
12.
J Biol Chem ; 286(10): 8655-8665, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21209102

RESUMEN

Fibrosis is a pathological process characterized by infiltration and proliferation of mesenchymal cells in interstitial space. A substantial portion of these cells is derived from residing non-epithelial and/or epithelial cells that have acquired the ability to migrate and proliferate. The mesenchymal transition is also observed in cancer cells to confer the ability to metastasize. Here, we show that renal fibrosis induced by unilateral ureteral obstruction and metastasis of human cancer xenografts are suppressed by administration of secreted Klotho protein to mice. Klotho is a single-pass transmembrane protein expressed in renal tubular epithelial cells. The extracellular domain of Klotho is secreted by ectodomain shedding. Secreted Klotho protein directly binds to the type-II TGF-ß receptor and inhibits TGF-ß1 binding to cell surface receptors, thereby inhibiting TGF-ß1 signaling. Klotho suppresses TGF-ß1-induced epithelial-to-mesenchymal transition (EMT) responses in cultured cells, including decreased epithelial marker expression, increased mesenchymal marker expression, and/or increased cell migration. In addition to TGF-ß1 signaling, secreted Klotho has been shown to inhibit Wnt and IGF-1 signaling that can promote EMT. These results have raised the possibility that secreted Klotho may function as an endogenous anti-EMT factor by inhibiting multiple growth factor signaling pathways simultaneously.


Asunto(s)
Glucuronidasa/metabolismo , Neoplasias Renales/metabolismo , Riñón/metabolismo , Neoplasias Experimentales/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Regulación Neoplásica de la Expresión Génica/genética , Glucuronidasa/genética , Células HEK293 , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Riñón/patología , Neoplasias Renales/genética , Neoplasias Renales/patología , Proteínas Klotho , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/genética , Trasplante Heterólogo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
13.
J Biol Chem ; 286(46): 40083-90, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21953454

RESUMEN

Expression of the clusterin (CLU) gene results in the synthesis of a conventional secretory isoform set (pre- and mature secretory clusterin proteins, psCLU/sCLU), as well as another set of intracellular isoforms, appearing in the cytoplasm (pre-nuclear CLU, pnCLU) and in the nucleus as an ∼55-kDa mature nuclear clusterin (nCLU) form. These two isoform sets have opposing cell functions: pro-survival and pro-death, respectively. Although much is known about the regulation and function of sCLU as a pro-survival factor, the regulation and function of endogenous nCLU in cell death are relatively unexplored. Here, we show that depletion of endogenous nCLU protein using siRNA specific to its truncated mRNA increased clonogenic survival of ionizing radiation (IR)-exposed cells. nCLU-mediated apoptosis was Bax-dependent, and lethality correlated with accumulation of mature nCLU protein. nCLU accumulation was regulated by CRM1 because binding between CRM1 and nCLU proteins was significantly diminished by leptomycin B (LMB), and nuclear levels of nCLU protein were significantly enhanced by LMB and IR co-treatment. Moreover, LMB treatment significantly enhanced IR-induced nCLU-mediated cell death responses. Importantly, bax(-/-) and bax(-/-)/bak(-/-) double knock-out cells were resistant to nCLU-mediated cell death, whereas bak(-/-) or wild-type bax(+/+)/bak(+/+) cells were hypersensitive. The regulation of nCLU by CRM1 nuclear export/import may explain recent clinical results showing that highly malignant tumors have lost the ability to accumulate nCLU levels, thereby avoiding growth inhibition and cell death.


Asunto(s)
Apoptosis/efectos de la radiación , Núcleo Celular/metabolismo , Clusterina/metabolismo , Rayos gamma , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/efectos de la radiación , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Núcleo Celular/genética , Clusterina/genética , Ácidos Grasos Insaturados/farmacología , Humanos , Carioferinas/genética , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína Exportina 1
14.
Genes Chromosomes Cancer ; 50(4): 275-83, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21319262

RESUMEN

Using a custom CGH-like oligonucleotide array to measure the global microsatellite content in the genomes of 72 cancer, cancer-free, and high risk patient and cell line samples (56 germline DNA and 16 in tumor or tumor cell line DNA) we found a unique, reproducible, and statistically significant pattern of 18 motif-specific microsatellite families (out of 962 possible 1-6 mer repeats) in breast cancer patient germline and tumor DNA, but not in germline DNA of cancer-free volunteer controls or in breast cancer patients with BRCA1/2 mutations. These high-similarity A/T rich repetitive motifs were also more pronounced in the germlines and tumors of colon cancer tumor patients (3/6 samples) and microsatellite unstable colon cancer cell lines; however, germline DNA of sporadic breast cancer patients exhibited the largest global content shift for those motifs with extreme AT/GC ratios. These results indicate that global microsatellite variability is complex, suggest the existence of a previously unknown genomic destabilization mechanism in breast cancer patients' germline DNA, and warrant further testing of such microsatellite variability as a predictor of future breast cancer development.


Asunto(s)
Secuencia Rica en At , Neoplasias de la Mama/genética , Inestabilidad de Microsatélites , Repeticiones de Microsatélite/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , ADN de Neoplasias/genética , Femenino , Genes BRCA1 , Genes BRCA2 , Variación Genética , Humanos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos
15.
Nutrients ; 14(15)2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35893874

RESUMEN

The compound ß-lapachone, a naturally derived naphthoquinone, has been utilized as a potent medicinal nutrient to improve health. Over the last twelve years, numerous reports have demonstrated distinct associations of ß-lapachone and NAD(P)H: quinone oxidoreductase 1 (NQO1) protein in the amelioration of various diseases. Comprehensive research of NQO1 bioactivity has clearly confirmed the tumoricidal effects of ß-lapachone action through NAD+-keresis, in which severe DNA damage from reactive oxygen species (ROS) production triggers a poly-ADP-ribose polymerase-I (PARP1) hyperactivation cascade, culminating in NAD+/ATP depletion. Here, we report a novel combination strategy with aminooxyacetic acid (AOA), an aspartate aminotransferase inhibitor that blocks the malate-aspartate shuttle (MAS) and synergistically enhances the efficacy of ß-lapachone metabolic perturbation in NQO1+ breast cancer. We evaluated metabolic turnover in MDA-MB-231 NQO1+, MDA-MB-231 NQO1-, MDA-MB-468, and T47D cancer cells by measuring the isotopic labeling of metabolites from a [U-13C]glucose tracer. We show that ß-lapachone treatment significantly hampers lactate secretion by ~85% in NQO1+ cells. Our data demonstrate that combinatorial treatment decreases citrate, glutamate, and succinate enrichment by ~14%, ~50%, and ~65%, respectively. Differences in citrate, glutamate, and succinate fractional enrichments indicate synergistic effects on central metabolism based on the coefficient of drug interaction. Metabolic modeling suggests that increased glutamine anaplerosis is protective in the case of MAS inhibition.


Asunto(s)
Ácido Aminooxiacético , Neoplasias de la Mama , Naftoquinonas , Ácido Aminooxiacético/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Citratos , Femenino , Glutamatos/metabolismo , Humanos , NAD/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/farmacología , Naftoquinonas/uso terapéutico , Succinatos/metabolismo
16.
Nat Cell Biol ; 5(4): 320-9, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12652308

RESUMEN

Bax induces mitochondrial-dependent cell death signals in mammalian cells. However, the mechanism of how Bax is kept inactive has remained unclear. Yeast-based functional screening of Bax inhibitors from mammalian cDNA libraries identified Ku70 as a new Bax suppressor. Bax-mediated apoptosis was suppressed by overexpression of Ku70 in mammalian cells, but enhanced by downregulation of Ku70. We found that Ku70 interacts with Bax, and that the carboxyl terminus of Ku70 and the amino terminus of Bax are required for this interaction. Bax is known to translocate from the cytosol to mitochondria when cells receive apoptotic stimuli. We found that Ku70 blocks the mitochondrial translocation of Bax. These results suggest that in addition to its previously recognized DNA repair activity in the nucleus, Ku70 has a cytoprotective function in the cytosol that controls the localization of Bax.


Asunto(s)
Apoptosis/fisiología , Proteínas de Arabidopsis/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Eucariotas/metabolismo , Mitocondrias/metabolismo , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Proto-Oncogénicas/deficiencia , Animales , Proteínas de Arabidopsis/genética , Sitios de Unión/genética , Citosol/metabolismo , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Ratones , Mutación/genética , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Recombinantes de Fusión , Transducción de Señal/genética , Proteína X Asociada a bcl-2
17.
Proteome Sci ; 9: 53, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21917185

RESUMEN

An improved version of quantitative protein array platform utilizing linear Quantum dot signaling for systematically measuring protein levels and phosphorylation states is presented. The signals are amplified linearly by a confocal laser Quantum dot scanner resulting in ~1000-fold more sensitivity than traditional Western blots, but are not linear by the enzyme-based amplification. Software is developed to facilitate the quantitative readouts of signaling network activities. Kinetics of EGFRvIII mutant signaling was analyzed to quantify cross-talks between EGFR and other signaling pathways.

18.
Cell Syst ; 12(1): 68-81.e11, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33476554

RESUMEN

Redox cofactor production is integral toward antioxidant generation, clearance of reactive oxygen species, and overall tumor response to ionizing radiation treatment. To identify systems-level alterations in redox metabolism that confer resistance to radiation therapy, we developed a bioinformatics pipeline for integrating multi-omics data into personalized genome-scale flux balance analysis models of 716 radiation-sensitive and 199 radiation-resistant tumors. These models collectively predicted that radiation-resistant tumors reroute metabolic flux to increase mitochondrial NADPH stores and reactive oxygen species (ROS) scavenging. Simulated genome-wide knockout screens agreed with experimental siRNA gene knockdowns in matched radiation-sensitive and radiation-resistant cancer cell lines, revealing gene targets involved in mitochondrial NADPH production, central carbon metabolism, and folate metabolism that allow for selective inhibition of glutathione production and H2O2 clearance in radiation-resistant cancers. This systems approach represents a significant advancement in developing quantitative genome-scale models of redox metabolism and identifying personalized metabolic targets for improving radiation sensitivity in individual cancer patients.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias , Humanos , NADP/química , NADP/genética , Neoplasias/genética , Neoplasias/radioterapia , Oxidación-Reducción , Especies Reactivas de Oxígeno/química
19.
Carcinogenesis ; 31(10): 1889-96, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20663777

RESUMEN

DNA double-strand breaks (DSBs) are the most deleterious lesion inflicted by ionizing radiation. Although DSBs are potentially carcinogenic, it is not clear whether complex DSBs that are refractory to repair are more potently tumorigenic compared with simple breaks that can be rapidly repaired, correctly or incorrectly, by mammalian cells. We previously demonstrated that complex DSBs induced by high-linear energy transfer (LET) Fe ions are repaired slowly and incompletely, whereas those induced by low-LET gamma rays are repaired efficiently by mammalian cells. To determine whether Fe-induced DSBs are more potently tumorigenic than gamma ray-induced breaks, we irradiated 'sensitized' murine astrocytes that were deficient in Ink4a and Arf tumor suppressors and injected the surviving cells subcutaneously into nude mice. Using this model system, we find that Fe ions are potently tumorigenic, generating tumors with significantly higher frequency and shorter latency compared with tumors generated by gamma rays. Tumor formation by Fe-irradiated cells is accompanied by rampant genomic instability and multiple genomic changes, the most interesting of which is loss of the p15/Ink4b tumor suppressor due to deletion of a chromosomal region harboring the CDKN2A and CDKN2B loci. The additional loss of p15/Ink4b in tumors derived from cells that are already deficient in p16/Ink4a bolsters the hypothesis that p15 plays an important role in tumor suppression, especially in the absence of p16. Indeed, we find that reexpression of p15 in tumor-derived cells significantly attenuates the tumorigenic potential of these cells, indicating that p15 loss may be a critical event in tumorigenesis triggered by complex DSBs.


Asunto(s)
Inhibidor p15 de las Quinasas Dependientes de la Ciclina/fisiología , Roturas del ADN de Doble Cadena , Neoplasias/etiología , Animales , Células Cultivadas , Aberraciones Cromosómicas , Deleción Cromosómica , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Reparación del ADN , Inestabilidad Genómica , Humanos , Ratones
20.
Clin Cancer Res ; 15(1): 131-9, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19118040

RESUMEN

PURPOSE: beta-Lapachone (ARQ 501, a formulation of beta-lapachone complexed with hydroxypropyl-beta-cyclodextrin) is a novel anticancer agent with selectivity against prostate cancer cells overexpressing the NAD(P)H:quinone oxidoreductase-1 enzyme. Lack of solubility and an efficient drug delivery strategy limits this compound in clinical applications. In this study, we aimed to develop beta-lapachone-containing polymer implants (millirods) for direct implantation into prostate tumors to test the hypothesis that the combination of a tumor-specific anticancer agent with site-specific release of the agent will lead to significant antitumor efficacy. EXPERIMENTAL DESIGN: Survival assays in vitro were used to test the killing effect of beta-lapachone in different prostate cancer cells. beta-Lapachone release kinetics from millirods was determined in vitro and in vivo. PC-3 prostate tumor xenografts in athymic nude mice were used for antitumor efficacy studies in vivo. RESULTS: beta-Lapachone killed three different prostate cancer cell lines in an NAD(P)H:quinone oxidoreductase-1-dependent manner. Upon incorporation of solid-state inclusion complexes of beta-lapachone with hydroxypropyl-beta-cyclodextrin into poly(D,L-lactide-co-glycolide) millirods, beta-lapachone release kinetics in vivo showed a burst release of approximately 0.5 mg within 12 hours and a subsequently sustained release of the drug ( approximately 0.4 mg/kg/d) comparable with that observed in vitro. Antitumor efficacy studies showed significant tumor growth inhibition by beta-lapachone millirods compared with controls (P < 0.0001; n = 10 per group). Kaplan-Meier survival curves showed that tumor-bearing mice treated with beta-lapachone millirods survived nearly 2-fold longer than controls, without observable systemic toxicity. CONCLUSIONS: Intratumoral delivery of beta-lapachone using polymer millirods showed the promising therapeutic potential for human prostate tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Implantes de Medicamentos/administración & dosificación , Naftoquinonas/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Portadores de Fármacos/administración & dosificación , Implantes de Medicamentos/uso terapéutico , Humanos , Masculino , Ratones , Ratones Desnudos , Naftoquinonas/uso terapéutico , Polímeros/farmacología , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA