Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 177(2): 446-462.e16, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30951671

RESUMEN

Poor reproducibility within and across studies arising from lack of knowledge regarding the performance of extracellular RNA (exRNA) isolation methods has hindered progress in the exRNA field. A systematic comparison of 10 exRNA isolation methods across 5 biofluids revealed marked differences in the complexity and reproducibility of the resulting small RNA-seq profiles. The relative efficiency with which each method accessed different exRNA carrier subclasses was determined by estimating the proportions of extracellular vesicle (EV)-, ribonucleoprotein (RNP)-, and high-density lipoprotein (HDL)-specific miRNA signatures in each profile. An interactive web-based application (miRDaR) was developed to help investigators select the optimal exRNA isolation method for their studies. miRDar provides comparative statistics for all expressed miRNAs or a selected subset of miRNAs in the desired biofluid for each exRNA isolation method and returns a ranked list of exRNA isolation methods prioritized by complexity, expression level, and reproducibility. These results will improve reproducibility and stimulate further progress in exRNA biomarker development.


Asunto(s)
Ácidos Nucleicos Libres de Células/aislamiento & purificación , MicroARN Circulante/aislamiento & purificación , ARN/aislamiento & purificación , Adulto , Líquidos Corporales/química , Línea Celular , Vesículas Extracelulares/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , MicroARNs/aislamiento & purificación , MicroARNs/metabolismo , ARN/metabolismo , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN/métodos
2.
Cell ; 177(2): 463-477.e15, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30951672

RESUMEN

To develop a map of cell-cell communication mediated by extracellular RNA (exRNA), the NIH Extracellular RNA Communication Consortium created the exRNA Atlas resource (https://exrna-atlas.org). The Atlas version 4P1 hosts 5,309 exRNA-seq and exRNA qPCR profiles from 19 studies and a suite of analysis and visualization tools. To analyze variation between profiles, we apply computational deconvolution. The analysis leads to a model with six exRNA cargo types (CT1, CT2, CT3A, CT3B, CT3C, CT4), each detectable in multiple biofluids (serum, plasma, CSF, saliva, urine). Five of the cargo types associate with known vesicular and non-vesicular (lipoprotein and ribonucleoprotein) exRNA carriers. To validate utility of this model, we re-analyze an exercise response study by deconvolution to identify physiologically relevant response pathways that were not detected previously. To enable wide application of this model, as part of the exRNA Atlas resource, we provide tools for deconvolution and analysis of user-provided case-control studies.


Asunto(s)
Comunicación Celular/fisiología , ARN/metabolismo , Adulto , Líquidos Corporales/química , Ácidos Nucleicos Libres de Células/metabolismo , MicroARN Circulante/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Masculino , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN/métodos , Programas Informáticos
3.
FASEB J ; 28(1): 230-43, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24025729

RESUMEN

Uterine glands and their secretions are indispensable for endometrial function and fertility; however, the mechanisms regulating their development and function are not well understood. Forkhead transcription factor box A2 (FOXA2) is uniquely expressed in the glandular epithelial (GE) cells of the uterus, and conditional deletion of Foxa2 after birth impedes uterine gland development. An integrative approach was used here to define the FOXA2 cistrome in the murine uterus. Genome-wide mapping of FOXA2 binding sites was combined with transcriptomic analyses of isolated GE and Foxa2-deleted uteri. ChIP-Seq analyses found the number of FOXA2 target genes was substantially greater in the adult (8893) than neonatal uterus (1101). In the neonatal uterus, FOXA2-bound and GE-expressed genes (469) were enriched for developmentally related processes, including cell cycle, cell junction, focal adhesion, and WNT signaling. In the adult uterus, FOXA2-bound and GE-expressed genes (3730) were enriched for functional processes, including metabolic pathways, focal adhesion, bacterial invasion of epithelial cells, and WNT signaling. Analysis of the uterine FOXA2 cistrome provides novel insights into mechanisms governing endometrial gland development and function, which are important to understand fundamental aspects of uterine differentiation, regeneration and disease.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Factor Nuclear 3-beta del Hepatocito/metabolismo , Análisis por Micromatrices/métodos , Útero/metabolismo , Animales , Femenino , Inmunohistoquímica , Ratones
4.
Biol Reprod ; 90(1): 7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24227756

RESUMEN

Fibroblast growth factors (FGFs) and their receptors (FGFRs) regulate luminal epithelial (LE) cell proliferation in the adult mouse uterus. This study tested the hypothesis that FGFR2 has a biological role in postnatal development and function of the uterus by conditionally deleting Fgfr2 after birth using progesterone receptor (Pgr)-Cre mice. Adult Fgfr2 mutant female mice were initially subfertile and became infertile with increasing parity. No defects in uterine gland development were observed in conditional Fgfr2 mutant mice. In the adult, Fgfr2 mutant mice possessed a histologically normal reproductive tract with the exception of the uterus. The LE of the Fgfr2 mutant uterus was stratified, but no obvious histological differences were observed in the glandular epithelium, stroma, or myometrium. Within the stratified LE, cuboidal basal cells were present and positive for basal cell markers (KRT14 and TRP63). Nulliparous bred Fgfr2 mutants contained normal numbers of blastocysts on Day 3.5 postmating, but the number of embryo implantation sites was substantially reduced on Day 5.5 postmating. These results support the idea that loss of FGFR2 in the uterus after birth alters its development, resulting in LE stratification and peri-implantation pregnancy loss.


Asunto(s)
Células Epiteliales/fisiología , Fertilidad/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Útero/citología , Útero/fisiología , Animales , Implantación del Embrión/genética , Pérdida del Embrión/genética , Femenino , Genotipo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Útero/crecimiento & desarrollo
5.
J Virol ; 87(10): 5586-92, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23468505

RESUMEN

Cache Valley virus (CVV)-induced malformations have been previously reproduced in ovine fetuses. To evaluate the development of the antiviral response by the early, infected fetus, before the development of immunocompetency, ovine fetuses at 35 days of gestation were inoculated in utero with CVV and euthanized at 7, 10, 14, 21, and 28 days postinfection. The antiviral immune response in immature fetuses infected with CVV was evaluated. Gene expression associated with an innate, immune response was quantified by real-time quantitative PCR. The upregulated genes in infected fetuses included ISG15, Mx1, Mx2, IL-1, IL-6, TNF-α, TLR-7, and TLR-8. The amount of Mx1 protein, an interferon-stimulated GTPase capable of restricting growth of bunyaviruses, was elevated in the allantoic and amniotic fluid in infected fetuses. ISG15 protein expression was significantly increased in target tissues of infected animals. B lymphocytes and immunoglobulin-positive cells were detected in lymphoid tissues and in the meninges of infected animals. These results demonstrated that the infected ovine fetus is able to initiate an innate and adaptive immune response much earlier than previously known, which presumably contributes to viral clearance in infected animals.


Asunto(s)
Virus Bunyamwera/inmunología , Infecciones por Bunyaviridae/inmunología , Enfermedades Fetales/inmunología , Enfermedades de las Cabras/inmunología , Animales , Virus Bunyamwera/patogenicidad , Infecciones por Bunyaviridae/virología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Enfermedades de las Cabras/virología , Cabras , Inmunidad Innata , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Biol Reprod ; 89(4): 86, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23946541

RESUMEN

All mammalian uteri have luminal (LE) and glandular epithelia (GE) in their endometrium. The LE mediates uterine receptivity and blastocyst attachment for implantation, and the GE synthesize and secrete or transport bioactive substances involved in blastocyst implantation, uterine receptivity, and stromal cell decidualization. However, the mechanisms governing uterine epithelial development after birth and their function in the adult are not fully understood. Here, comprehensive microarray analysis was conducted on LE and GE isolated by laser capture microdissection from uteri on Postnatal Day 10 (PD 10) and day of pseudopregnancy (DOPP) 2.5 and 3.5. This data was integrated with analysis of uteri from gland-containing control and aglandular progesterone-induced uterine gland knockout mice from PD 10 and DOPP 3.5. Many genes were expressed in both epithelia, but there was greater expression of genes in the LE than in the GE. In the neonate, GE-expressed genes were enriched for morphogenesis, development, migration, and retinoic acid signaling. In the adult, LE-expressed genes were enriched for metabolic processes and steroid biosynthesis, whereas retinoid signaling, tight junction, extracellular matrix, and regulation of kinase activity were enriched in the GE. The transcriptome differences in the epithelia support the idea that each cell type has a distinct and complementary function in the uterus. The candidate genes and regulatory networks identified here provide a framework to discover new mechanisms regulating development of epithelia in the postnatal uterus and their functions in early pregnancy.


Asunto(s)
Envejecimiento , Endometrio/citología , Endometrio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transcripción Genética , Transcriptoma , Animales , Animales Recién Nacidos , Decidua/citología , Decidua/efectos de los fármacos , Decidua/crecimiento & desarrollo , Decidua/metabolismo , Endometrio/efectos de los fármacos , Endometrio/crecimiento & desarrollo , Femenino , Fármacos para la Fertilidad Femenina/farmacología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Captura por Microdisección con Láser , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Progesterona/farmacología , Seudoembarazo/inducido químicamente , Seudoembarazo/metabolismo , Transcripción Genética/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Útero/citología , Útero/efectos de los fármacos , Útero/crecimiento & desarrollo , Útero/metabolismo
7.
Biol Reprod ; 88(4): 93, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23407384

RESUMEN

Uterine glands and their secretions are hypothesized to be essential for blastocyst implantation and decidualization in the uterus of rodents and humans. One factor solely expressed by uterine glands in mice is leukemia inhibitory factor (LIF), and Lif null mice are infertile because of defective blastocyst attachment to the uterine luminal epithelium (LE). Progesterone treatment of neonatal mice permanently ablates differentiation of uterine glands, resulting in an aglandular uterus in the adult. Progesterone-induced uterine gland knockout (PUGKO) mice were used to investigate the biological role of uterine glands in blastocyst implantation and stromal cell decidualization. As compared to controls, PUGKO mice cycled normally but were infertile. Histological assessment of PUGKO uteri on Days 5.5 and 8.5 postmating found a hatched blastocyst apposed to an intact LE without evidence of implantation or stromal cell decidualization. Expression of several implantation-related factors, including Lif and PTGS2, were altered in the PUGKO uterus, whereas expression of steroid hormone receptors and their regulated genes was not different. Artificial decidualization was observed in the uteri of control but not PUGKO mice. Further, intrauterine administration of LIF failed to promote artificial decidualization in the uterus of PUGKO mice. Thus, uterine glands and their secretions have important biological roles in blastocyst implantation and stromal cell decidualization in the uterus.


Asunto(s)
Decidua/fisiología , Implantación del Embrión/fisiología , Endometrio/fisiología , Glándulas Exocrinas/fisiología , Útero/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Endometrio/ultraestructura , Glándulas Exocrinas/efectos de los fármacos , Glándulas Exocrinas/metabolismo , Femenino , Factor Inhibidor de Leucemia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Progesterona/administración & dosificación , Útero/efectos de los fármacos , Útero/metabolismo , Útero/ultraestructura
8.
Biol Reprod ; 88(3): 72, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23390164

RESUMEN

Introduction of semen into the female reproductive tract may induce molecular and cellular changes facilitating conception and pregnancy. Because prostaglandins (PGs) and appropriate vascularization of the endometrium are crucial for pregnancy success, the effect of seminal plasma (SP) on PG synthesis and angiogenesis was investigated. Gilts at estrus received an infusion of 100 ml of either SP or PBS (control). Uterine horns were collected on Days 1, 3, 5, and 10 after each treatment. Concentrations of PGE2, PGF2alpha , and PGFM were measured in the uterine lumen and endometrial tissue. Expression of PG synthesis pathway enzymes and angiogenic factors, infiltration of immune cells, and vascular bed development were assessed. One day after SP infusion, the PGE2:PGF2alpha ratio in the uterine lumen was lower than in controls. In endometrial tissue, however, PGE2 levels and the PGE2:PGF2alpha ratio were elevated on Day 10. PG-endoperoxide synthase expression in the endometrium was up-regulated on Day 1 and decreased on Day 5 after SP treatment compared to that in controls. PGF2alpha synthase levels were decreased on Day 5 and 10 in SP-treated animals when compared to controls. SP-induced vascular bed development was apparent shortly before the time corresponding to maternal recognition of pregnancy in the pig. Together, these data indicate that the porcine uterus can be sensitized shortly after SP exposure to evoke prolonged effects on PG synthesis and angiogenesis in the endometrium, persisting over the course of the prereceptive phase. Thus, SP can affect uterine receptivity and embryo-maternal interactions in pigs through locally direct and/or indirect mechanisms.


Asunto(s)
Endometrio/fisiología , Neovascularización Fisiológica , Prostaglandinas/biosíntesis , Semen/fisiología , Animales , Biomarcadores/metabolismo , Células Endoteliales/metabolismo , Femenino , Leucocitos/fisiología , Masculino , ARN Mensajero/metabolismo , Porcinos , Factores de Crecimiento Endotelial Vascular/metabolismo , Factor de von Willebrand/metabolismo
9.
Biol Reprod ; 86(5): 146, 1-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22238285

RESUMEN

Uterine glands and their secretions are required for conceptus (embryo/fetus and associated placenta) survival and development. In most mammals, uterine gland morphogenesis or adenogenesis is a uniquely postnatal event; however, little is known about the mechanisms governing the developmental event. In sheep, progestin treatment of neonatal ewes permanently ablated differentiation of the endometrial glands. Similarly, progesterone (P4) inhibits adenogenesis in neonatal mouse uterus. Thus, P4 can be used as a tool to discover mechanisms regulating endometrial adenogenesis. Female pups were treated with sesame vehicle alone as a control or P4 from Postnatal Day 2 (PD 2) to PD 10, and reproductive tracts were examined on PD 5, 10, or 20. Endometrial glands were fully developed in control mice by PD 20 but not in P4-treated mice. All other uterine cell types appeared normal. Treatment with P4 stimulated proliferation of the stroma but suppressed proliferation of the luminal epithelium. Microarray analysis revealed that expression of genes were reduced (Car2, Fgf7, Fgfr2, Foxa2, Fzd10, Met, Mmp7, Msx1, Msx2, Wnt4, Wnt7a, Wnt16) and increased (Hgf, Ihh, Wnt11) by P4 in the neonatal uterus. These results support the idea that P4 inhibits endometrial adenogenesis in the developing neonatal uterus by altering expression of morphoregulatory genes and consequently disrupting normal patterns of cell proliferation and development.


Asunto(s)
Progesterona/farmacología , Útero/efectos de los fármacos , Útero/crecimiento & desarrollo , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Ratones , Morfogénesis/efectos de los fármacos
10.
Biol Reprod ; 85(2): 386-96, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21508348

RESUMEN

The success of postnatal uterine morphogenesis dictates, in part, the embryotrophic potential and functional capacity of the adult uterus. The definitive role of Wnt7a in postnatal uterine development and adult function requires a conditional knockout, because global deletion disrupts müllerian duct patterning, specification, and cell fate in the fetus. The Wnt7a-null uterus appears to be posteriorized because of developmental defects in the embryo, as evidenced by the stratified luminal epithelium that is normally found in the vagina and the presence of short and uncoiled oviducts. To understand the biological role of WNT7A after birth and allow tissue-selective deletion of Wnt7a, we generated loxP-flanked exon 2 mice and conditionally deleted Wnt7a after birth in the uterus by crossing them with Pgr(Cre) mice. Morphological examination revealed no obvious differences in the vagina, cervix, oviduct, or ovary. The uteri of Wnt7a mutant mice contained no endometrial glands, whereas all other uterine cell types appeared to be normal. Postnatal differentiation of endometrial glands was observed in control mice, but not in mutant mice, between Postnatal Days 3 and 12. Expression of morphoregulatory genes, particularly Foxa2, Hoxa10, Hoxa11, Msx1, and Wnt16, was disrupted in the Wnt7a mutant uteri. Conditional Wnt7a mutant mice were not fertile. Although embryos were present in uteri of mutant mice on Day 3.5 of pregnancy, blastocyst implantation was not observed on Day 5.5. Furthermore, expression of several genes (Foxa2, Lif, Msx1, and Wnt16) was reduced or absent in adult Wnt7a-deleted uteri on Day 3.5 postmating. These results indicate that WNT7A plays a critical role in postnatal uterine gland morphogenesis and function, which are important for blastocyst implantation and fertility in the adult uterus.


Asunto(s)
Fertilidad/fisiología , Eliminación de Gen , Útero/crecimiento & desarrollo , Proteínas Wnt/metabolismo , Animales , Proliferación Celular , Implantación del Embrión/fisiología , Femenino , Fertilidad/genética , Regulación de la Expresión Génica/fisiología , Ratones , Útero/citología , Útero/metabolismo , Proteínas Wnt/genética
12.
Methods Mol Biol ; 1740: 43-57, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29388135

RESUMEN

Extracellular RNAs are initiating increased interest due to their potentials in serving as novel biomarkers, mediators of intercellular communication, and therapeutic applications. As a newly emerging field, one of the main obstacles is the lack of standardized protocols for RNA isolations. Here we describe protocols for commercially available kits that have been modified to yield consistent results for isolation of extracellular RNA from both whole serum/plasma and extracellular vesicle-enriched serum/plasma samples.


Asunto(s)
Vesículas Extracelulares/metabolismo , ARN/sangre , ARN/aislamiento & purificación , Animales , Espacio Extracelular/metabolismo , Humanos , Biología Molecular/métodos , Plasma/metabolismo , Ultracentrifugación/métodos
13.
Oncotarget ; 8(12): 20145-20164, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28423620

RESUMEN

Exosomes have emerged as important mediators of diverse biological functions including tumor suppression, tumor progression, invasion, immune escape and cell-to-cell communication, through the release of molecules such as mRNAs, miRNAs, and proteins. Here, we identified differentially expressed exosomal miRNAs between normal epithelial ovarian cell line and both resistant and sensitive ovarian cancer (OC) cell lines. We found miR-940 as abundant in exosomes from SKOV3-IP1, HeyA8, and HeyA8-MDR cells. The high expression of miR-940 is associated with better survival in patients with ovarian serous cystadenocarcinoma. Ectopic expression of miR-940 inhibited proliferation, colony formation, invasion, and migration and triggered G0/G1 cell cycle arrest and apoptosis in OC cells. Overexpression of miR-940 also inhibited tumor cell growth in vivo. We showed that proto-oncogene tyrosine-protein kinase (SRC) is directly targeted by miR-940 and that miR-940 inhibited SRC expression at mRNA and protein levels. Following this inhibition, the expression of proteins downstream of SRC, such as FAK, paxillin and Akt was also reduced. Collectively, our results suggest that OC cells secrete the tumor-suppressive miR-940 into the extracellular environment via exosomes, to maintain their invasiveness and tumorigenic phenotype.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Exosomas/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias Ováricas/patología , Familia-src Quinasas/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Femenino , Genes Supresores de Tumor , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Proto-Oncogenes Mas , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/genética
14.
J Natl Cancer Inst ; 109(7)2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28376174

RESUMEN

Background: The PI3K/AKT/P70S6K pathway is an attractive therapeutic target in ovarian and uterine malignancies because of its high rate of deregulation and key roles in tumor growth. Here, we examined the biological effects of MSC2363318A, which is a novel inhibitor of AKT1, AKT3, and P70S6K. Methods: Orthotopic murine models of ovarian and uterine cancer were utilized to study the effect of MSC2363318A on survival and regression. For each cell line, 10 mice were treated in each of the experimental arms tested. Moreover, in vitro experiments in 21 cell lines (MTT, immunoblot analysis, plasmid transfection, reverse phase protein array [RPPA]) were carried out to characterize underlying mechanisms and potential biomarkers of response. All statistical tests were two-sided. Results: MSC2363318A decreased tumor growth and metastases in multiple murine orthotopic models of ovarian (SKOV3ip1, HeyA8, and Igrov1) and uterine (Hec1a) cancer by reducing proliferation and angiogenesis and increasing cell death. Statistically significant prolonged overall survival was achieved with combination MSC2363318A and paclitaxel in the SKUT2 (endometrioid) uterine cancer mouse model ( P < .001). Mice treated with combination MSC2363318A and paclitaxel had the longest overall survival (mean = 104.2 days, 95% confidence interval [CI] = 97.0 to 111.4) compared with those treated with vehicle (mean = 61.9 days, 95% CI = 46.3 to 77.5), MSC2363318A alone (mean = 89.7 days, 95% CI = 83.0 to 96.4), and paclitaxel alone (mean = 73.6 days, 95% CI = 53.4 to 93.8). Regression and stabilization of established tumors in the Ishikawa (endometrioid) uterine cancer model was observed in mice treated with combination MSC2363318A and paclitaxel. Synergy between MSC2363318A and paclitaxel was observed in vitro in cell lines that had an IC50 of 5 µM or greater. RPPA results identified YAP1 as a candidate marker to predict cell lines that were most sensitive to MSC2363318A (R = 0.54, P = .02). After establishment of a murine ovarian cancer model of adaptive anti-angiogenic resistance (SKOV3ip1-luciferase), we demonstrate that resensitization to bevacizumab occurs with the addition of MSC2363318A, resulting in improved overall survival ( P = .01) using the Kaplan-Meier method. Mice treated with bevacizumab induction followed by MSC2363318A had the longest overall survival (mean = 66.0 days, 95% CI = 53.9 to 78.1) compared with mice treated with control (mean = 42.0 days, 95% CI = 31.4 to 52.6) and bevacizumab-sensitive mice (mean = 47.2 days; 95% CI = 37.5 to 56.9). Conclusions: MSC2363318A has therapeutic efficacy in multiple preclinical models of ovarian and uterine cancer. These findings support clinical development of a dual AKT/P70S6K inhibitor.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Ováricas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Neoplasias Uterinas/metabolismo , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Bevacizumab/administración & dosificación , Bevacizumab/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Concentración 50 Inhibidora , Estimación de Kaplan-Meier , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Factores de Transcripción , Carga Tumoral/efectos de los fármacos , Neoplasias Uterinas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
15.
J Biomed Nanotechnol ; 12(6): 1159-73, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27319211

RESUMEN

The bottleneck in current vector-based cancer therapy is the targeted and controlled release of therapeutics in tumors. Exosomes are submicron-sized vesicles that are secreted by all cell types and are involved in communication and transportation of materials between cells. Analogous in size and function to synthetic nanoparticles, exosomes offer many advantages, rendering them the most promising candidates for targeted drug or gene delivery vehicles. Patient-specific customized therapeutic strategies can be engineered using exosomes derived from the patient's own healthy cells. Therefore, exosome-based cancer therapy has the potential to become an important part of personalized medicine. Interest in exosomes as carrier organelles is relatively recent. Knowledge about exosomal biology and its applications remains limited. The present review is an attempt to describe the current status of the application of exosomes to cancer therapy and the potential challenges associated with their use.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Exosomas , Terapia Genética , Inmunoterapia , Nanopartículas , Neoplasias/terapia , Línea Celular , Humanos
16.
Mol Cancer Ther ; 15(12): 2894-2904, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27638860

RESUMEN

To determine the efficacy of a novel and safer (for gastrointestinal tract) aspirin (aspirin-PC) in preclinical models of ovarian cancer, in vitro dose-response studies were performed to compare the growth-inhibitory effect of aspirin-PC versus aspirin on three human (A2780, SKOV3ip1, and HeyA8) and a mouse (ID8) ovarian cancer cell line over an 8-day culture period. In the in vivo studies, the aspirin test drugs were studied alone and in the presence of a VEGF-A inhibitor (bevacizumab or B20), due to an emerging role for platelets in tumor growth following antiangiogenic therapy, and we examined their underlying mechanisms. Aspirin-PC was more potent (vs. aspirin) in blocking the growth of both human and mouse ovarian cancer cells in monolayer culture. Using in vivo model systems of ovarian cancer, we found that aspirin-PC significantly reduced ovarian cancer growth by 50% to 90% (depending on the ovarian cell line). The efficacy was further enhanced in combination with Bevacizumab or B20. The growth-inhibitory effect on ovarian tumor mass and number of tumor nodules was evident, but less pronounced for aspirin and the VEGF inhibitors alone. There was no detectable gastrointestinal toxicity. Both aspirin and aspirin-PC also inhibited cell proliferation, angiogenesis, and increased apoptosis of ovarian cancer cells. In conclusion, PC-associated aspirin markedly inhibits the growth of ovarian cancer cells, which exceeds that of the parent drug, in both cell culture and in mouse model systems. We also found that both aspirin-PC and aspirin have robust antineoplastic action in the presence of VEGF-blocking drugs. Mol Cancer Ther; 15(12); 2894-904. ©2016 AACR.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Aspirina/farmacología , Neovascularización Patológica , Neoplasias Ováricas/patología , Fosfatidilcolinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Tromboxanos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 76(24): 7194-7207, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27742688

RESUMEN

Cancer cells actively promote their tumorigenic behavior by reprogramming gene expression. Loading intraluminal vesicles with specific miRNAs and releasing them into the tumor microenvironment as exosomes is one mechanism of reprogramming whose regulation remains to be elucidated. Here, we report that miR-6126 is ubiquitously released in high abundance from both chemosensitive and chemoresistant ovarian cancer cells via exosomes. Overexpression of miR-6126 was confirmed in healthy ovarian tissue compared with ovarian cancer patient samples and correlated with better overall survival in patients with high-grade serous ovarian cancer. miR-6126 acted as a tumor suppressor by directly targeting integrin-ß1, a key regulator of cancer cell metastasis. miR-6126 mimic treatment of cancer cells resulted in increased miR-6126 and decreased integrin-ß1 mRNA levels in the exosome. Functional analysis showed that treatment of endothelial cells with miR-6126 mimic significantly reduced tube formation as well as invasion and migration capacities of ovarian cancer cells in vitro Administration of miR-6126 mimic in an orthotopic mouse model of ovarian cancer elicited a relative reduction in tumor growth, proliferating cells, and microvessel density. miR-6126 inhibition promoted oncogenic behavior by leading ovarian cancer cells to release more exosomes. Our findings provide new insights into the role of exosomal miRNA-mediated tumor progression and suggest a new therapeutic approach to disrupt oncogenic phenotypes in tumors. Cancer Res; 76(24); 7194-207. ©2016 AACR.


Asunto(s)
Exosomas/metabolismo , MicroARNs/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Animales , Western Blotting , Femenino , Genes Supresores de Tumor , Xenoinjertos , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Ratones , Ratones Desnudos , Reacción en Cadena de la Polimerasa , Transcriptoma , Transfección
18.
Nat Commun ; 7: 11169, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-27041221

RESUMEN

A deeper mechanistic understanding of tumour angiogenesis regulation is needed to improve current anti-angiogenic therapies. Here we present evidence from systems-based miRNA analyses of large-scale patient data sets along with in vitro and in vivo experiments that miR-192 is a key regulator of angiogenesis. The potent anti-angiogenic effect of miR-192 stems from its ability to globally downregulate angiogenic pathways in cancer cells through regulation of EGR1 and HOXB9. Low miR-192 expression in human tumours is predictive of poor clinical outcome in several cancer types. Using 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) nanoliposomes, we show that miR-192 delivery leads to inhibition of tumour angiogenesis in multiple ovarian and renal tumour models, resulting in tumour regression and growth inhibition. This anti-angiogenic and anti-tumour effect is more robust than that observed with an anti-VEGF antibody. Collectively, these data identify miR-192 as a central node in tumour angiogenesis and support the use of miR-192 in an anti-angiogenesis therapy.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Redes Reguladoras de Genes , Proteínas de Homeodominio/fisiología , Neoplasias Renales/genética , MicroARNs/fisiología , Neovascularización Patológica/genética , Neoplasias Ováricas/genética , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Terapia Genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Renales/irrigación sanguínea , Neoplasias Renales/terapia , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/terapia , Fosfatidilcolinas , Carga Tumoral
19.
Curr Gene Ther ; 15(2): 182-92, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25537774

RESUMEN

Exosomes are 30-100 nm bodies secreted from almost all types of cells into the extracellular spaces. They enclose in their lumen active genetic information in the form of messenger RNA (mRNA), micro RNA (miRNA), DNA and active peptides that are representative of the parental cell and can be isolated from different body fluids. Exosomes can participate in inter-cellular communication by trafficking molecules to their target cells. Because they can stably carry cargo including miRNA, mRNA, and proteins and can pass through stringent biological barriers (e.g., blood brain barrier) without eliciting an immune response, they are considered as an ideal acellular vehicle for drug delivery. In this review, we describe the structure and biogenesis of exosomes and new directions related to their role in diagnosis and treatment of diseases, especially for cancer. We also discuss potential challenges associated with exosomes that should be addressed before exosome-based therapy can be applied to clinical settings.


Asunto(s)
Sistemas de Liberación de Medicamentos , Exosomas/genética , Terapia Genética , Neoplasias/terapia , Barrera Hematoencefálica , Comunicación Celular/genética , Humanos , MicroARNs/genética , MicroARNs/uso terapéutico , Neoplasias/genética , ARN Mensajero/genética , ARN Mensajero/uso terapéutico
20.
Noncoding RNA ; 1(1): 53-68, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-29861415

RESUMEN

Non-coding RNAs, such as microRNAs and long non-coding RNAs, are important regulatory molecules which are corrupted in cancer, often in a tissue and stage specific manner. Accumulated data suggests that these promising biomarkers, may also form the basis of novel targeted therapeutic strategies. The role of exosomes in cancer development and metastasis pathways is also increasingly well described. These endosome derived extracellular vesicles which are trafficked horizontally between tumor cells, and vertically between tumor cells and the surrounding microenvironment, carry bioactive cargos, which can reprogram the phenotype of recipient cells with important oncogenic consequences. Exosomes are enriched with non-coding RNA content. Within exosomes, non-coding RNAs are secreted into the peripheral circulation and other bodily fluids where they are protected from enzymatic degradation by the surrounding phospholipid membrane. Exosomes are therefore a highly promising source of diagnostic and prognostic material in cancer. Furthermore, as exosomes are natural ncRNA carriers, they may be adapted for the purpose of drug delivery by the introduction of exogenous ncRNAs or by manipulating their endogenous ncRNA content. In the current review, we will explore these highly clinically relevant themes by examining the roles of exosomal ncRNAs in cancer diagnostics, prognostics and therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA