Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 4(1): 1420, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34934174

RESUMEN

Elevated aldehyde dehydrogenase (ALDH) activity correlates with poor outcome for many solid tumors as ALDHs may regulate cell proliferation and chemoresistance of cancer stem cells (CSCs). Accordingly, potent, and selective inhibitors of key ALDH enzymes may represent a novel CSC-directed treatment paradigm for ALDH+ cancer types. Of the many ALDH isoforms, we and others have implicated the elevated expression of ALDH1A3 in mesenchymal glioma stem cells (MES GSCs) as a target for the development of novel therapeutics. To this end, our structure of human ALDH1A3 combined with in silico modeling identifies a selective, active-site inhibitor of ALDH1A3. The lead compound, MCI-INI-3, is a selective competitive inhibitor of human ALDH1A3 and shows poor inhibitory effect on the structurally related isoform ALDH1A1. Mass spectrometry-based cellular thermal shift analysis reveals that ALDH1A3 is the primary binding protein for MCI-INI-3 in MES GSC lysates. The inhibitory effect of MCI-INI-3 on retinoic acid biosynthesis is comparable with that of ALDH1A3 knockout, suggesting that effective inhibition of ALDH1A3 is achieved with MCI-INI-3. Further development is warranted to characterize the role of ALDH1A3 and retinoic acid biosynthesis in glioma stem cell growth and differentiation.


Asunto(s)
Aldehído Oxidorreductasas/antagonistas & inhibidores , Glioma/metabolismo , Células Madre Neoplásicas/metabolismo , Tretinoina/metabolismo , Humanos
2.
NAR Cancer ; 3(4): zcab044, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34806016

RESUMEN

Elevated expression of the DNA damage response proteins PARP1 and poly(ADP-ribose) glycohydrolase (PARG) in glioma stem cells (GSCs) suggests that glioma may be a unique target for PARG inhibitors (PARGi). While PARGi-induced cell death is achieved when combined with ionizing radiation, as a single agent PARG inhibitors appear to be mostly cytostatic. Supplementation with the NAD+ precursor dihydronicotinamide riboside (NRH) rapidly increased NAD+ levels in GSCs and glioma cells, inducing PARP1 activation and mild suppression of replication fork progression. Administration of NRH+PARGi triggers hyperaccumulation of poly(ADP-ribose) (PAR), intra S-phase arrest and apoptosis in GSCs but minimal PAR induction or cytotoxicity in normal astrocytes. PAR accumulation is regulated by select PARP1- and PAR-interacting proteins. The involvement of XRCC1 highlights the base excision repair pathway in responding to replication stress while enhanced interaction of PARP1 with PCNA, RPA and ORC2 upon PAR accumulation implicates replication associated PARP1 activation and assembly with pre-replication complex proteins upon initiation of replication arrest, the intra S-phase checkpoint and the onset of apoptosis.

3.
DNA Repair (Amst) ; 93: 102930, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33087267

RESUMEN

The enzymes of the base excision repair (BER) pathway form DNA lesion-dependent, transient complexes that vary in composition based on the type of DNA damage. These protein sub-complexes facilitate substrate/product handoff to ensure reaction completion so as to avoid accumulation of potentially toxic DNA repair intermediates. However, in the mammalian cell, additional signaling molecules are required to fine-tune the activity of the BER pathway enzymes and to facilitate chromatin/histone reorganization for access to the DNA lesion for repair. These signaling enzymes include nicotinamide adenine dinucleotide (NAD+) dependent poly(ADP-ribose) polymerases (PARP1, PARP2) and class III deacetylases (SIRT1, SIRT6) that comprise a key PARP-NAD-SIRT axis to facilitate the regulation and coordination of BER in the mammalian cell. Here, we briefly describe the key nodes in the BER pathway that are regulated by this axis and highlight the cellular and organismal variation in NAD+ bioavailability that can impact BER signaling potential. We discuss how cellular NAD+ is required for BER to maintain genome stability and to mount a robust cellular response to DNA damage. Finally, we consider the dependence of BER on the PARP-NAD-SIRT axis for BER protein complex assembly.


Asunto(s)
Reparación del ADN , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Sirtuinas/metabolismo , Cromatina/metabolismo , ADN/metabolismo , Daño del ADN , Humanos , Poli(ADP-Ribosa) Polimerasas/metabolismo
4.
Sci Rep ; 10(1): 651, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959836

RESUMEN

Changes in nicotinamide adenine dinucleotide (NAD+) levels that compromise mitochondrial function trigger release of DNA damaging reactive oxygen species. NAD+ levels also affect DNA repair capacity as NAD+ is a substrate for PARP-enzymes (mono/poly-ADP-ribosylation) and sirtuins (deacetylation). The ecto-5'-nucleotidase CD73, an ectoenzyme highly expressed in cancer, is suggested to regulate intracellular NAD+ levels by processing NAD+ and its bio-precursor, nicotinamide mononucleotide (NMN), from tumor microenvironments, thereby enhancing tumor DNA repair capacity and chemotherapy resistance. We therefore investigated whether expression of CD73 impacts intracellular NAD+ content and NAD+-dependent DNA repair capacity. Reduced intracellular NAD+ levels suppressed recruitment of the DNA repair protein XRCC1 to sites of genomic DNA damage and impacted the amount of accumulated DNA damage. Further, decreased NAD+ reduced the capacity to repair DNA damage induced by DNA alkylating agents. Overall, reversal of these outcomes through NAD+ or NMN supplementation was independent of CD73. In opposition to its proposed role in extracellular NAD+ bioprocessing, we found that recombinant human CD73 only poorly processes NMN but not NAD+. A positive correlation between CD73 expression and intracellular NAD+ content could not be made as CD73 knockout human cells were efficient in generating intracellular NAD+ when supplemented with NAD+ or NMN.


Asunto(s)
5'-Nucleotidasa/metabolismo , 5'-Nucleotidasa/fisiología , Daño del ADN , Reparación del ADN , NAD/metabolismo , NAD/fisiología , Poli ADP Ribosilación , Poli(ADP-Ribosa) Polimerasas/fisiología , Microambiente Tumoral/genética , Microambiente Tumoral/fisiología , 5'-Nucleotidasa/genética , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Sirtuinas , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo
5.
Nucleic Acids Res ; 47(12): 6269-6286, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31287140

RESUMEN

Protein-protein interactions regulate many essential enzymatic processes in the cell. Somatic mutations outside of an enzyme active site can therefore impact cellular function by disruption of critical protein-protein interactions. In our investigation of the cellular impact of the T304I cancer mutation of DNA Polymerase ß (Polß), we find that mutation of this surface threonine residue impacts critical Polß protein-protein interactions. We show that proteasome-mediated degradation of Polß is regulated by both ubiquitin-dependent and ubiquitin-independent processes via unique protein-protein interactions. The ubiquitin-independent proteasome pathway regulates the stability of Polß in the cytosol via interaction between Polß and NAD(P)H quinone dehydrogenase 1 (NQO1) in an NADH-dependent manner. Conversely, the interaction of Polß with the scaffold protein X-ray repair cross complementing 1 (XRCC1) plays a role in the localization of Polß to the nuclear compartment and regulates the stability of Polß via a ubiquitin-dependent pathway. Further, we find that oxidative stress promotes the dissociation of the Polß/NQO1 complex, enhancing the interaction of Polß with XRCC1. Our results reveal that somatic mutations such as T304I in Polß impact critical protein-protein interactions, altering the stability and sub-cellular localization of Polß and providing mechanistic insight into how key protein-protein interactions regulate cellular responses to stress.


Asunto(s)
ADN Polimerasa beta/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Estrés Oxidativo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo , Línea Celular Tumoral , Cromatina/enzimología , Neoplasias del Colon/genética , ADN Polimerasa beta/química , ADN Polimerasa beta/genética , Estabilidad de Enzimas , Humanos , Mutación , NAD/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación
6.
Am J Cardiol ; 122(2): 229-234, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29751956

RESUMEN

Penile erection is a hemodynamic process consisting of 2 synchronized components in which the first (active) requires proper vascular endothelium functioning, whereas the second one (passive) is based on a veno-occlusive mechanism. Antihypertensive treatment reduces the passive component, often leading to the development of erectile dysfunction (ED), but lifestyle modifications can improve the sexual functioning. The study aimed to evaluate the association between blood pressure (BP) reduction caused by cardiovascular training and the intensity of ED in men with coronary heart disease. A total of 101 men (mean age 59.50 ± 7.93) with ED treated invasively for coronary heart disease and subjected to cardiac rehabilitation were enrolled. Patient characteristics, the International Index of Erectile Function 5 (IIEF-5) questionnaire (IIEF-5), and BP values were collected at baseline and after 6 months of cardiac rehabilitation and were analyzed. Cardiac rehabilitation led to a significant reduction of 5.08 mm Hg in systolic BP (p <0.001) and of 1.60 mm Hg in diastolic BP (p <0.001). The IIEF-5 score (EQ) significantly increased (median 15, interquartile range 11 to 19 vs median 18, interquartile range 12 to 21, p <0.001). Greater improvement in sexual performance was significantly negatively correlated with age, concentration of triglycerides, and high-density lipoprotein, whereas it was positively correlated with the presence of diabetes and baseline IIEF-5 score. After excluding patients with diabetes, a greater decrease in systolic BP was found to be significantly associated with greater improvement in erectile performance. In conclusion, a reduction of arterial BP caused by cardiac training is accompanied by improvement in erectile performance. This effect is the strongest in patients with hypertension and those with dyslipidemia.


Asunto(s)
Presión Arterial/fisiología , Rehabilitación Cardiaca/métodos , Enfermedad Coronaria/rehabilitación , Disfunción Eréctil/rehabilitación , Terapia por Ejercicio/métodos , Ejercicio Físico/fisiología , Erección Peniana/fisiología , Enfermedad Coronaria/complicaciones , Enfermedad Coronaria/fisiopatología , Electrocardiografía Ambulatoria , Disfunción Eréctil/etiología , Disfunción Eréctil/fisiopatología , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Tiempo
7.
Mol Cell Biol ; 38(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29483302

RESUMEN

Insulin receptor substrate 1 (IRS-1) is a common cytosolic adaptor molecule involved in signal transduction from insulin and insulin-like growth factor I (IGF-I) receptors. IRS-1 can also be found in the nucleus. We report here a new finding of unique IRS-1 nuclear structures, which we observed initially in glioblastoma biopsy specimens and glioblastoma xenografts. These nuclear structures can be reproduced in vitro by the ectopic expression of IRS-1 cDNA cloned in frame with the nuclear localization signal (NLS-IRS-1). In these structures, IRS-1 localizes at the periphery, while the center harbors a key autophagy protein, LC3. These new nuclear structures are highly dynamic, rapidly exchange IRS-1 molecules with the surrounding nucleoplasm, disassemble during mitosis, and require a growth stimulus for their reassembly and maintenance. In tumor cells engineered to express NLS-IRS-1, the IRS-1/LC3 nuclear structures repress autophagy induced by either amino acid starvation or rapamycin treatment. In this process, IRS-1 nuclear structures sequester LC3 inside the nucleus, possibly preventing its cytosolic translocation and the formation of new autophagosomes. This novel mechanism provides a quick and reversible way of inhibiting autophagy, which could counteract autophagy-induced cancer cell death under severe stress, including anticancer therapies.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/fisiología , Proteínas Adaptadoras Transductoras de Señales , Autofagia/fisiología , Núcleo Celular/fisiología , Supervivencia Celular/genética , Glioblastoma/metabolismo , Células HeLa , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/ultraestructura , Factor I del Crecimiento Similar a la Insulina/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Neoplasias , Fosfoproteínas , Receptor IGF Tipo 1/fisiología , Transducción de Señal
8.
Cell Cycle ; 17(3): 330-347, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29143560

RESUMEN

Fetal stem cells are a unique type of adult stem cells that have been suggested to be broadly multipotent with some features of pluripotency. Their clinical potential has been documented but their upgrade to full pluripotency could open up a wide range of cell-based therapies particularly suited for pediatric tissue engineering, longitudinal studies or disease modeling. Here we describe episomal reprogramming of mesenchymal stem cells from the human amnion to pluripotency (AM-iPSC) in chemically defined conditions. The AM-iPSC expressed markers of embryonic stem cells, readily formed teratomas with tissues of all three germ layers present and had a normal karyotype after around 40 passages in culture. We employed novel computational methods to determine the degree of pluripotency from microarray and RNA sequencing data in these novel lines alongside an iPSC and ESC control and found that all lines were deemed pluripotent, however, with variable scores. Differential expression analysis then identified several groups of genes that potentially regulate this variability in lines within the boundaries of pluripotency, including metallothionein proteins. By further studying this variability, characteristics relevant to cell-based therapies, like differentiation propensity, could be uncovered and predicted in the pluripotent stage.


Asunto(s)
Amnios/citología , Células Madre Pluripotentes Inducidas/citología , Biomarcadores/metabolismo , Forma de la Célula , Células Cultivadas , Redes Reguladoras de Genes , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Teratoma/patología , Transcripción Genética
9.
Arch Med Sci ; 13(2): 302-310, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28261282

RESUMEN

INTRODUCTION: Due to the pathogenetic association between erectile disorders and cardiovascular diseases, cardiologists consult many patients with erectile dysfunction (ED). The aim of the study was to evaluate sexual function in patients with coronary heart disease (CHD) and the use of sexual knowledge in cardiology practice, both current use and that expected by patients. MATERIAL AND METHODS: One thousand one hundred and thirty-six patients (average age: 60.73 ±9.20) underwent a dedicated survey which encompassed demographic data and the presence of modifiable ED risk factors. The presence of ED was assessed using the International Index of Erectile Function (IIEF-5) Questionnaire. RESULTS: Sexual problems were discussed by cardiologists with 45 (3.96%) patients. The frequency of initiating the topic was significantly associated with the respondents' education level (p = 0.0031); however, it was not associated with the patients' age, duration of CHD, presence of ED, or modifiable risk factors. Four hundred and sixteen (36.62%) respondents indicated that they expect their cardiologist to take an interest in their ED. Nine hundred and twenty-six (81.51%) patients claimed good sexual function to be important or very important to them. Attitude to sexual function was significantly associated with age (p < 0.0001), duration of CHD (p = 0.0018), education (p = 0.0011), presence of ED (p = 0.0041), diabetes (p = 0.0283) and hyperlipidaemia (p = 0.0014). CONCLUSIONS: The low frequency with which cardiologists initiate the topic of ED is in contrast to the expectations of patients with CHD. The majority of these patients regard good sexual maintenance as an important part of their life.

10.
J Cell Physiol ; 232(6): 1275-1286, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27607558

RESUMEN

Ethanol plays a detrimental role in the development of the brain. Multiple studies have shown that ethanol inhibits insulin-like growth factor I receptor (IGF-IR) function. Because the IGF-IR contributes to brain development by supporting neural growth, survival, and differentiation, we sought to determine the molecular mechanism(s) involved in ethanol's effects on this membrane-associated tyrosine kinase. Using multiple neuronal cell types, we performed Western blot, immunoprecipitation, and GST-pulldowns following acute (1-24 h) or chronic (3 weeks) treatment with ethanol. Surprisingly, exposure of multiple neuronal cell types to acute (up to 24 h) ethanol (50 mM) enhanced IGF-I-induced phosphorylation of extracellular regulated kinases (ERKs), without affecting IGF-IR tyrosine phosphorylation itself, or Akt phosphorylation. This acute increase in ERKs phosphorylation was followed by the expected inhibition of the IGF-IR signaling following 3-week ethanol exposure. We then expressed a GFP-tagged IGF-IR construct in PC12 cells and used them to perform fluorescence recovery after photobleaching (FRAP) analysis. Using these fluorescently labeled cells, we determined that 50 mM ethanol decreased the half-time of the IGF-IR-associated FRAP, which implied that cell membrane-associated signaling events could be affected. Indeed, co-immunoprecipitation and GST-pulldown studies demonstrated that the acute ethanol exposure increased the recruitment of p52-Shc to the Grb2-Shc complex, which is known to engage the Ras-Raf-ERKs pathway following IGF-1 stimulation. These experiments indicate that even a short and low-dose exposure to ethanol may dysregulate function of the receptor, which plays a critical role in brain development. J. Cell. Physiol. 232: 1275-1286, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Etanol/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Adaptadora GRB2/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Células PC12 , Fosforilación/efectos de los fármacos , Ratas , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
11.
PLoS One ; 11(7): e0157907, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27389192

RESUMEN

The renal podocyte plays an important role in maintaining the structural integrity of the glomerular basement membrane. We have previously reported that patients with idiopathic nephrotic syndrome (INS) have increased IL-2 production. We hypothesized that podocytes express an IL-2 receptor (IL-2R) and signaling through this receptor can result in podocyte injury. To confirm the presence of the IL-2R, we tested a conditionally immortalized murine podocyte cell line by flow cytometry, qPCR, and Western blot. To test for the presence of the IL-2R in vivo, immunohistochemical staining was performed on human renal biopsies in children with FSGS and control. Podocytes were stimulated with IL-2 in vitro, to study signaling events via the JAK/STAT pathway. The results showed that stimulation with IL-2 resulted in increased mRNA and protein expression of STAT 5a, phosphorylated STAT 5, JAK 3, and phosphorylated JAK 3. We then investigated for signs of cellular injury and the data showed that pro-apoptotic markers Bax and cFLIP were significantly increased following IL-2 exposure, whereas LC3 II was decreased. Furthermore, mitochondrial depolarization and apoptosis were both significantly increased following activation of the IL-2R. We used a paracellular permeability assay to monitor the structural integrity of a podocyte monolayer following IL-2 exposure. The results showed that podocytes exposed to IL-2 have increased albumin leakage across the monolayer. We conclude that murine podocytes express the IL-2R, and that activation through the IL-2R results in podocyte injury.


Asunto(s)
Síndrome Nefrótico/metabolismo , Podocitos/metabolismo , Receptores de Interleucina-2/metabolismo , Albúminas/metabolismo , Animales , Apoptosis , Biopsia , Niño , Modelos Animales de Enfermedad , Humanos , Inmunosupresores/uso terapéutico , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias/metabolismo , Fosforilación , Podocitos/citología , Reacción en Cadena de la Polimerasa , Transducción de Señal
12.
Artículo en Inglés | MEDLINE | ID: mdl-26869992

RESUMEN

Ketone bodies [beta-hydroxybutyrate (bHB) and acetoacetate] are mainly produced in the liver during prolonged fasting or starvation. bHB is a very efficient energy substrate for sustaining ATP production in peripheral tissues; importantly, its consumption is preferred over glucose. However, the majority of malignant cells, particularly cancer cells of neuroectodermal origin such as glioblastoma, are not able to use ketone bodies as a source of energy. Here, we report a novel observation that fenofibrate, a synthetic peroxisome proliferator-activated receptor alpha (PPARa) agonist, induces bHB production in melanoma and glioblastoma cells, as well as in neurospheres composed of non-transformed cells. Unexpectedly, this effect is not dependent on PPARa activity or its expression level. The fenofibrate-induced ketogenesis is accompanied by growth arrest and downregulation of transketolase, but the NADP/NADPH and GSH/GSSG ratios remain unaffected. Our results reveal a new, intriguing aspect of cancer cell biology and highlight the benefits of fenofibrate as a supplement to both canonical and dietary (ketogenic) therapeutic approaches against glioblastoma.

13.
J Biol Chem ; 290(52): 30931-46, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26534959

RESUMEN

The HIV-1 transactivator protein Tat is implicated in the neuronal damage that contributes to neurocognitive impairment affecting people living with HIV/AIDS. Aberrant splicing of TAU exon 10 results in tauopathies characterized by alterations in the proportion of TAU isoforms containing three (3R) or four (4R) microtubule-binding repeats. The splicing factor SC35/SRSF2 binds to nuclear RNA and facilitates the incorporation of exon 10 in the TAU molecule. Here, we utilized clinical samples, an animal model, and neuronal cell cultures and found that Tat promotes TAU 3R up-regulation through increased levels of phosphorylated SC35, which is retained in nuclear speckles. This mechanism involved Tat-mediated increased expression of DYRK1A and was prevented by DYRK1A silencing. In addition, we found that Tat associates with TAU RNA, further demonstrating that Tat interferes with host RNA metabolism in the absence of viral infection. Altogether, our data unravel a novel mechanism of Tat-mediated neuronal toxicity through dysregulation of the SC35-dependent alternative splicing of TAU exon 10. Furthermore, the increased immunostaining of DYRK1A in HIV+ brains without pathology points at dysregulation of DYRK1A as an early event in the neuronal complications of HIV infection.


Asunto(s)
Exones , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/biosíntesis , Ribonucleoproteínas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Infecciones por VIH/genética , VIH-1/genética , Humanos , Ratones , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Ribonucleoproteínas/genética , Factores de Empalme Serina-Arginina , Regulación hacia Arriba , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Proteínas tau/genética , Quinasas DyrK
14.
J Biol Chem ; 290(13): 8067-80, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25645911

RESUMEN

Glioblastoma is one of the most aggressive brain tumors. We have previously found up-regulation of growth differentiation factor 15 (GDF15) in glioblastoma cells treated with the anticancer agent fenofibrate. Sequence analysis of GDF15 revealed the presence of a microRNA, miR-3189, in the single intron. We then asked whether miR-3189 was expressed in clinical samples and whether it was functional in glioblastoma cells. We found that expression of miR-3189-3p was down-regulated in astrocytoma and glioblastoma clinical samples compared with control brain tissue. In vitro, the functionality of miR-3189-3p was tested by RNA-binding protein immunoprecipitation, and miR-3189-3p coimmunoprecipitated with Argonaute 2 together with two of its major predicted gene targets, the SF3B2 splicing factor and the guanine nucleotide exchange factor p63RhoGEF. Overexpression of miR-3189-3p resulted in a significant inhibition of cell proliferation and migration through direct targeting of SF3B2 and p63RhoGEF, respectively. Interestingly, miR-3189-3p levels were increased by treatment of glioblastoma cells with fenofibrate, a lipid-lowering drug with multiple anticancer activities. The attenuated expression of miR-3189-3p in clinical samples paralleled the elevated expression of SF3B2, which could contribute to the activation of SF3B2 growth-promoting pathways in these tumors. Finally, miR-3189-3p-mediated inhibition of tumor growth in vivo further supported the function of this microRNA as a tumor suppressor.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , MicroARNs/genética , Animales , Secuencia de Bases , Sitios de Unión , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Glioblastoma/genética , Glioblastoma/patología , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Ratones Desnudos , Trasplante de Neoplasias , Interferencia de ARN , Factores de Empalme de ARN , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Factores de Intercambio de Guanina Nucleótido Rho/biosíntesis , Factores de Intercambio de Guanina Nucleótido Rho/genética
15.
Mol Cell Biol ; 35(1): 182-98, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25332241

RESUMEN

Fenofibrate (FF) is a common lipid-lowering drug and a potent agonist of the peroxisome proliferator-activated receptor alpha (PPARα). FF and several other agonists of PPARα have interesting anticancer properties, and our recent studies demonstrate that FF is very effective against tumor cells of neuroectodermal origin. In spite of these promising anticancer effects, the molecular mechanism(s) of FF-induced tumor cell toxicity remains to be elucidated. Here we report a novel PPARα-independent mechanism explaining FF's cytotoxicity in vitro and in an intracranial mouse model of glioblastoma. The mechanism involves accumulation of FF in the mitochondrial fraction, followed by immediate impairment of mitochondrial respiration at the level of complex I of the electron transport chain. This mitochondrial action sensitizes tested glioblastoma cells to the PPARα-dependent metabolic switch from glycolysis to fatty acid ß-oxidation. As a consequence, prolonged exposure to FF depletes intracellular ATP, activates the AMP-activated protein kinase-mammalian target of rapamycin-autophagy pathway, and results in extensive tumor cell death. Interestingly, autophagy activators attenuate and autophagy inhibitors enhance FF-induced glioblastoma cytotoxicity. Our results explain the molecular basis of FF-induced glioblastoma cytotoxicity and reveal a new supplemental therapeutic approach in which intracranial infusion of FF could selectively trigger metabolic catastrophe in glioblastoma cells.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Fenofibrato/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Astrocitos/citología , Neoplasias Encefálicas/metabolismo , Muerte Celular , Línea Celular Tumoral , Transporte de Electrón , Femenino , Glioblastoma/metabolismo , Glucólisis , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Trasplante de Neoplasias , Oxígeno/metabolismo , Consumo de Oxígeno , PPAR alfa/metabolismo , Transducción de Señal
16.
Neoplasia ; 16(10): 874-82, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25379024

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with a median survival of 12 to 15 months after diagnosis. Acquired chemoresistance, high systemic toxicity, and low penetration of the blood brain barrier by many anticancer drugs contribute to the failure of anti-GBM therapies. To circumvent some of these obstacles, we tested a novel prodrug approach to evaluate anti-GBM efficacy by utilizing serum albumin-binding doxorubicin (Doxo), aldoxorubicin (Aldoxo), which is less toxic, is released from albumin in an acidic environment and accumulates in tumor tissues. A human GBM cell line that expresses a luciferase reporter (U87-luc) was stereotactically injected into the left striatum of the brain of immunodeficient mice. Following initial tumor growth for 12 days, mice were injected once a week in the tail-vein with Aldoxo [24 mg/kg or 18 mg/kg of doxorubicin equivalents-3/4 maximum tolerated dose (MTD)], Doxo [6 mg/kg (3/4 MTD)], or vehicle. Aldoxo-treated mice demonstrated significantly slower growth of the tumor when compared to vehicle-treated or Doxo-treated mice. Five out of eight Aldoxo-treated mice remained alive more than 60 days with a median survival of 62 days, while the median survival of vehicle- and Doxo-treated mice was only 26 days. Importantly, Aldoxo-treated mice exhibited high levels of Doxo within the tumor tissue, accompanied by low tumor cell proliferation (Ki67) and abundant intratumoral programmed cell death (cleaved caspase-3). Effective accumulation of Aldoxo in brain tumor tissues but not normal brain, its anti-tumor efficacy, and low toxicity, provide a strong rationale for evaluating this novel drug conjugate as a treatment for patients afflicted with GBM.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacología , Glioblastoma/tratamiento farmacológico , Hidrazonas/farmacología , Administración Intravenosa , Animales , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Femenino , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Hidrazonas/farmacocinética , Dosis Máxima Tolerada , Ratones Desnudos , Factores de Tiempo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Cancer ; 134(12): 2853-64, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24259296

RESUMEN

The accumulation of myeloid-derived suppressor cells (MDSC) in tumor-bearing hosts is a hallmark of malignancy-associated inflammation and a major mediator for the induction of T cell suppression in cancer. MDSC can be divided phenotypically into granulocytic (G-MDSC) and monocytic (Mo-MDSC) subgroups. Several mechanisms mediate the induction of T cell anergy by MDSC; however, the specific role of these pathways in the inhibitory activity of MDSC subpopulations remains unclear. Therefore, we aimed to determine the effector mechanisms by which subsets of tumor-infiltrating MDSC block T cell function. We found that G-MDSC had a higher ability to impair proliferation and expression of effector molecules in activated T cells, as compared to Mo-MDSC. Interestingly, both MDSC subgroups inhibited T cells through nitric oxide (NO)-related pathways, but expressed different effector inhibitory mechanisms. Specifically, G-MDSC impaired T cells through the production of peroxynitrites (PNT), while Mo-MDSC suppressed by the release of NO. The production of PNT in G-MDSC depended on the expression of gp91(phox) and endothelial NO synthase (eNOS), while inducible NO synthase (iNOS) mediated the generation of NO in Mo-MDSC. Deletion of eNOS and gp91(phox) or scavenging of PNT blocked the suppressive function of G-MDSC and induced anti-tumoral effects, without altering Mo-MDSC inhibitory activity. Furthermore, NO-scavenging or iNOS knockdown prevented Mo-MDSC function, but did not affect PNT production or suppression by G-MDSC. These results suggest that MDSC subpopulations utilize independent effector mechanisms to regulate T cell function. Inhibition of these pathways is expected to specifically block MDSC subsets and overcome immune suppression in cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Granulocitos/inmunología , Monocitos/inmunología , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , NADPH Oxidasas/genética , Neoplasias/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/genética , Nitritos/metabolismo , Ácido Peroxinitroso/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/inmunología
18.
J Cell Physiol ; 228(11): 2127-38, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23558788

RESUMEN

Polycyclic aromatic hydrocarbons (PAHs) are the products of incomplete combustion of organic materials, which are present in cigarette smoke, deep-fried food, and in natural crude oil. Since PAH-metabolites form DNA adducts and cause oxidative DNA damage, we asked if these environmental carcinogens could affect transforming potential of the human Polyomavirus JC oncoprotein, T-antigen (JCV T-antigen). We extracted DMSO soluble PAHs from Deepwater Horizon oil spill in the Gulf of Mexico (oil-PAHs), and detected several carcinogenic PAHs. The oil-PAHs were tested in exponentially growing cultures of normal mouse fibroblasts (R508), and in R508 stably expressing JCV T-antigen (R508/T). The oil-PAHs were cytotoxic only at relatively high doses (1:50-1:100 dilution), and at 1:500 dilution the growth and cell survival rates were practically unaffected. This non-toxic dose triggered however, a significant accumulation of reactive oxygen species (ROS), caused oxidative DNA damage and the formation of DNA double strand breaks (DSBs). Although oil-PAHs induced similar levels of DNA damage in R508 and R508/T cells, only T-antigen expressing cells demonstrated inhibition of high fidelity DNA repair by homologous recombination (HRR). In contrast, low-fidelity repair by non-homologous end joining (NHEJ) was unaffected. This potential mutagenic shift between DNA repair mechanisms was accompanied by a significant increase in clonal growth of R508/T cells chronically exposed to low doses of the oil-PAHs. Our results indicate for the first time carcinogenic synergy in which oil-PAHs trigger oxidative DNA damage and JCV T-antigen compromises DNA repair fidelity.


Asunto(s)
Antígenos Virales de Tumores/genética , Virus JC/genética , Mutagénesis/efectos de los fármacos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Fraccionamiento Químico , Cromatografía Líquida de Alta Presión , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Dimetilsulfóxido/química , Histonas/metabolismo , Humanos , Ratones , Estrés Oxidativo/efectos de los fármacos , Petróleo
19.
Am J Physiol Renal Physiol ; 303(12): F1629-40, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23019230

RESUMEN

Candidate genes have been identified that confer increased risk for diabetic glomerulosclerosis (DG). Mice heterozygous for the Akita (Ins2(+/C96Y)) diabetogenic mutation with a second mutation introduced at the bradykinin 2 receptor (B2R(-/-)) locus express a disease phenotype that approximates human DG. Src homology 2 domain transforming protein 1 (p66) controls mitochondrial metabolism and cellular responses to oxidative stress, aging, and apoptosis. We generated p66-null Akita mice to test whether inactivating mutations at the p66 locus will rescue kidneys of Akita mice from disease-causing mutations at the Ins2 and B2R loci. Here we show null mutations at the p66 and B2R loci interact with the Akita (Ins2(+/C96Y)) mutation, independently and in combination, inducing divergent phenotypes in the kidney. The B2R(-/-) mutation induces detrimental phenotypes, as judged by increased systemic and renal levels of oxidative stress, histology, and urine albumin excretion, whereas the p66-null mutation confers a powerful protection phenotype. To elucidate the mechanism(s) of the protection phenotype, we turned to our in vitro system. Experiments with cultured podocytes revealed previously unrecognized cross talk between p66 and the redox-sensitive transcription factor p53 that controls hyperglycemia-induced ROS metabolism, transcription of p53 target genes (angiotensinogen, angiotensin II type-1 receptor, and bax), angiotensin II generation, and apoptosis. RNA-interference targeting p66 inhibits all of the above. Finally, protein levels of p53 target genes were upregulated in kidneys of Akita mice but unchanged in p66-null Akita mice. Taken together, p66 is a potential molecular target for therapeutic intervention in DG.


Asunto(s)
Diabetes Mellitus/genética , Insulina/genética , Mutación/genética , Fenotipo , Receptor de Bradiquinina B2/genética , Proteínas Adaptadoras de la Señalización Shc/genética , Animales , Apoptosis , Células Cultivadas , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Hiperglucemia/metabolismo , Hiperglucemia/patología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/metabolismo , Podocitos/patología , Receptor de Bradiquinina B2/deficiencia , Receptor de Bradiquinina B2/metabolismo , Proteínas Adaptadoras de la Señalización Shc/deficiencia , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Proteína p53 Supresora de Tumor/metabolismo
20.
Cell Cycle ; 11(14): 2660-71, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22732497

RESUMEN

Anti-neoplastic potential of calorie restriction or ligand-induced activation of peroxisome proliferator activated receptors (PPARs) has been demonstrated in multiple studies; however, mechanism(s) by which tumor cells respond to these stimuli remain to be elucidated. One of the potent agonists of PPARα, fenofibrate, is a commonly used lipid-lowering drug with low systemic toxicity. Fenofibrate-induced PPARα transcriptional activity is expected to shift energy metabolism from glycolysis to fatty acid ß-oxidation, which in the long-term, could target weak metabolic points of glycolysis-dependent glioblastoma cells. The results of this study demonstrate that 25 µM fenofibrate can effectively repress malignant growth of primary glial tumor cells and glioblastoma cell lines. This cytostatic action involves G(1) arrest accompanied by only a marginal level of apoptotic cell death. Although the cells treated with 25 µM fenofibrate remain arrested, the cells treated with 50 µM fenofibrate undergo massive apoptosis, which starts after 72 h of the treatment. This delayed apoptotic event was preceded by FoxO3A nuclear accumulation, FoxO3A phosphorylation on serine residue 413, its elevated transcriptional activity and expression of FoxO-dependent apoptotic protein, Bim. siRNA-mediated inhibition of FoxO3A attenuated fenofibrate-induced apoptosis, indicating a direct involvement of this transcription factor in the fenofibrate action against glioblastoma. These properties of fenofibrate, coupled with its low systemic toxicity, make it a good candidate in support of conventional therapies against glial tumors.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Fenofibrato/toxicidad , Factores de Transcripción Forkhead/metabolismo , Hipolipemiantes/toxicidad , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Nucléolo Celular/metabolismo , Metabolismo Energético , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , PPAR gamma/agonistas , PPAR gamma/metabolismo , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...