Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cell Stem Cell ; 19(5): 599-612, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27570067

RESUMEN

Age-related human hematopoietic stem cell (HSC) exhaustion and myeloid-lineage skewing promote oncogenic transformation of hematopoietic progenitor cells into therapy-resistant leukemia stem cells (LSCs) in secondary acute myeloid leukemia (AML). While acquisition of clonal DNA mutations has been linked to increased rates of secondary AML for individuals older than 60 years, the contribution of RNA processing alterations to human hematopoietic stem and progenitor aging and LSC generation remains unclear. Comprehensive RNA sequencing and splice-isoform-specific PCR uncovered characteristic RNA splice isoform expression patterns that distinguished normal young and aged human stem and progenitor cells (HSPCs) from malignant myelodysplastic syndrome (MDS) and AML progenitors. In splicing reporter assays and pre-clinical patient-derived AML models, treatment with a pharmacologic splicing modulator, 17S-FD-895, reversed pro-survival splice isoform switching and significantly impaired LSC maintenance. Therapeutic splicing modulation, together with monitoring splice isoform biomarkers of healthy HSPC aging versus LSC generation, may be employed safely and effectively to prevent relapse, the leading cause of leukemia-related mortality.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Empalme del ARN/genética , Animales , Supervivencia Celular/genética , Senescencia Celular/genética , Técnicas de Cocultivo , Células HEK293 , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Síndromes Mielodisplásicos/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Empalmosomas/metabolismo , Células del Estroma/metabolismo
2.
Cell Stem Cell ; 19(2): 177-191, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27292188

RESUMEN

Post-transcriptional adenosine-to-inosine RNA editing mediated by adenosine deaminase acting on RNA1 (ADAR1) promotes cancer progression and therapeutic resistance. However, ADAR1 editase-dependent mechanisms governing leukemia stem cell (LSC) generation have not been elucidated. In blast crisis chronic myeloid leukemia (BC CML), we show that increased JAK2 signaling and BCR-ABL1 amplification activate ADAR1. In a humanized BC CML mouse model, combined JAK2 and BCR-ABL1 inhibition prevents LSC self-renewal commensurate with ADAR1 downregulation. Lentiviral ADAR1 wild-type, but not an editing-defective ADAR1(E912A) mutant, induces self-renewal gene expression and impairs biogenesis of stem cell regulatory let-7 microRNAs. Combined RNA sequencing, qRT-PCR, CLIP-ADAR1, and pri-let-7 mutagenesis data suggest that ADAR1 promotes LSC generation via let-7 pri-microRNA editing and LIN28B upregulation. A small-molecule tool compound antagonizes ADAR1's effect on LSC self-renewal in stromal co-cultures and restores let-7 biogenesis. Thus, ADAR1 activation represents a unique therapeutic vulnerability in LSCs with active JAK2 signaling.


Asunto(s)
Adenosina Desaminasa/metabolismo , Autorrenovación de las Células , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/genética , Animales , Secuencia de Bases , Autorrenovación de las Células/genética , Proteínas de Fusión bcr-abl/metabolismo , Regulación Leucémica de la Expresión Génica , Janus Quinasa 2/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Edición de ARN/genética , Proteínas de Unión al ARN/genética , Transducción de Señal/genética
3.
Proc Natl Acad Sci U S A ; 112(50): 15444-9, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621726

RESUMEN

Formative research suggests that a human embryonic stem cell-specific alternative splicing gene regulatory network, which is repressed by Muscleblind-like (MBNL) RNA binding proteins, is involved in cell reprogramming. In this study, RNA sequencing, splice isoform-specific quantitative RT-PCR, lentiviral transduction, and in vivo humanized mouse model studies demonstrated that malignant reprogramming of progenitors into self-renewing blast crisis chronic myeloid leukemia stem cells (BC LSCs) was partially driven by decreased MBNL3. Lentiviral knockdown of MBNL3 resulted in reversion to an embryonic alternative splice isoform program typified by overexpression of CD44 transcript variant 3, containing variant exons 8-10, and BC LSC proliferation. Although isoform-specific lentiviral CD44v3 overexpression enhanced chronic phase chronic myeloid leukemia (CML) progenitor replating capacity, lentiviral shRNA knockdown abrogated these effects. Combined treatment with a humanized pan-CD44 monoclonal antibody and a breakpoint cluster region - ABL proto-oncogene 1, nonreceptor tyrosine kinase (BCR-ABL1) antagonist inhibited LSC maintenance in a niche-dependent manner. In summary, MBNL3 down-regulation-related reversion to an embryonic alternative splicing program, typified by CD44v3 overexpression, represents a previously unidentified mechanism governing malignant progenitor reprogramming in malignant microenvironments and provides a pivotal opportunity for selective BC LSC detection and therapeutic elimination.


Asunto(s)
Empalme Alternativo/genética , Autorrenovación de las Células/genética , Células Madre Embrionarias Humanas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Adulto , Animales , Apoptosis/genética , Crisis Blástica/genética , Crisis Blástica/patología , Médula Ósea/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Reprogramación Celular/genética , Femenino , Proteínas de Fusión bcr-abl/metabolismo , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hematopoyesis , Humanos , Receptores de Hialuranos/metabolismo , Ligandos , Masculino , Ratones , Persona de Mediana Edad , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Pluripotentes/citología , Proto-Oncogenes Mas
4.
Clin Lymphoma Myeloma Leuk ; 15 Suppl: S167-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26297272

RESUMEN

Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncoembryonic antigen. Because of its expression on the cell surface of leukemia cells from patients with chronic lymphocytic leukemia (CLL), but not on normal B-cells or other postpartum tissues, ROR1 is an attractive candidate for targeted therapies. UC-961 is a first-in-class humanized monoclonal antibody that binds the extracellular domain of ROR1. In this article we outline some of the preclinical studies leading to an investigational new drug designation, enabling clinical studies in patients with CLL.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/inmunología , Apoptosis , Línea Celular Tumoral , Humanos
5.
J Transl Med ; 13: 98, 2015 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-25889765

RESUMEN

BACKGROUND: Dormant leukemia stem cells (LSC) promote therapeutic resistance and leukemic progression as a result of unbridled activation of stem cell gene expression programs. Thus, we hypothesized that 1) deregulation of the hedgehog (Hh) stem cell self-renewal and cell cycle regulatory pathway would promote dormant human LSC generation and 2) that PF-04449913, a clinical antagonist of the GLI2 transcriptional activator, smoothened (SMO), would enhance dormant human LSC eradication. METHODS: To test these postulates, whole transcriptome RNA sequencing (RNA-seq), microarray, qRT-PCR, stromal co-culture, confocal fluorescence microscopic, nanoproteomic, serial transplantation and cell cycle analyses were performed on FACS purified normal, chronic phase (CP) chronic myeloid leukemia (CML), blast crisis (BC) phase CML progenitors with or without PF-04449913 treatment. RESULTS: Notably, RNA-seq analyses revealed that Hh pathway and cell cycle regulatory gene overexpression correlated with leukemic progression. While lentivirally enforced GLI2 expression enhanced leukemic progenitor dormancy in stromal co-cultures, this was not observed with a mutant GLI2 lacking a transactivation domain, suggesting that GLI2 expression prevented cell cycle transit. Selective SMO inhibition with PF-04449913 in humanized stromal co-cultures and LSC xenografts reduced downstream GLI2 protein and cell cycle regulatory gene expression. Moreover, SMO inhibition enhanced cell cycle transit and sensitized BC LSC to tyrosine kinase inhibition in vivo at doses that spare normal HSC. CONCLUSION: In summary, while GLI2, forms part of a core HH pathway transcriptional regulatory network that promotes human myeloid leukemic progression and dormant LSC generation, selective inhibition with PF-04449913 reduces the dormant LSC burden thereby providing a strong rationale for clinical trials predicated on SMO inhibition in combination with TKIs or chemotherapeutic agents with the ultimate aim of obviating leukemic therapeutic resistance, persistence and progression.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Leucemia/patología , Células Madre Neoplásicas/patología , Proteínas Nucleares/antagonistas & inhibidores , Animales , Secuencia de Bases , Técnicas de Cocultivo , Cartilla de ADN , Sangre Fetal/citología , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma , Proteína Gli2 con Dedos de Zinc
6.
Reprod Sci ; 21(9): 1161-70, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24784716

RESUMEN

Uterine fibroids are the most common solid tumors found in women of reproductive age. It has been reported that deregulation of the mammalian target of rapamycin (mTOR) pathway plays an important role in the etiology of leiomyoma. Here, we investigated the effect of rapamycin, an inhibitor of mTORC1, on the growth of primary fibroid smooth muscle cells (fSMCs) and human telomerase reverse transcriptase (hTERT)-transduced and immortalized fSMCs. With the primary fSMCs, a 24-hour treatment with rapamycin was sufficient to trigger a growth arrest that was not reversible upon drug removal. By contrast, the growth inhibitory effect of rapamycin on the hTERT-transduced fSMCs was readily reversible, as these cells resumed proliferation upon the withdrawal of the drug. These results suggest that rapamycin-induced irreversible growth arrest of fSMCs is dependent on the senescence barrier that is abrogated by the ectopic expression of telomerase.


Asunto(s)
Inhibidores de Crecimiento/farmacología , Leiomioma/enzimología , Miocitos del Músculo Liso/enzimología , Sirolimus/farmacología , Telomerasa/biosíntesis , Neoplasias Uterinas/enzimología , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/fisiología , Línea Celular Transformada , Células Cultivadas , Femenino , Inhibidores de Crecimiento/uso terapéutico , Humanos , Leiomioma/tratamiento farmacológico , Leiomioma/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Sirolimus/antagonistas & inhibidores , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/patología
7.
Cell Stem Cell ; 12(3): 316-28, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23333150

RESUMEN

Leukemia stem cells (LSCs) play a pivotal role in the resistance of chronic myeloid leukemia (CML) to tyrosine kinase inhibitors (TKIs) and its progression to blast crisis (BC), in part, through the alternative splicing of self-renewal and survival genes. To elucidate splice-isoform regulators of human BC LSC maintenance, we performed whole-transcriptome RNA sequencing, splice-isoform-specific quantitative RT-PCR (qRT-PCR), nanoproteomics, stromal coculture, and BC LSC xenotransplantation analyses. Cumulatively, these studies show that the alternative splicing of multiple prosurvival BCL2 family genes promotes malignant transformation of myeloid progenitors into BC LSCS that are quiescent in the marrow niche and that contribute to therapeutic resistance. Notably, sabutoclax, a pan-BCL2 inhibitor, renders marrow-niche-resident BC LSCs sensitive to TKIs at doses that spare normal progenitors. These findings underscore the importance of alternative BCL2 family splice-isoform expression in BC LSC maintenance and suggest that the combinatorial inhibition of prosurvival BCL2 family proteins and BCR-ABL may eliminate dormant LSCs and obviate resistance.


Asunto(s)
Leucemia/patología , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Crisis Blástica/metabolismo , Crisis Blástica/patología , Gosipol/análogos & derivados , Gosipol/farmacología , Humanos , Leucemia/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Mol Cancer Res ; 11(3): 207-18, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23339186

RESUMEN

Tumor necrosis factor (TNF)-α promotes tumor development under chronic inflammation. Because TNF also activates caspase-8, selective inhibition of TNF-induced extrinsic apoptosis would be required for inflammation-associated tumor growth. In a mouse model of inflammation-associated colon carcinogenesis, we found nuclear expression of ß-catenin in tumors of wild-type, but not mutant, mice that were made resistant to TNF-induced apoptosis by a germline mutation blocking caspase cleavage of the retinoblastoma (RB) protein, despite similar frequencies of ß-catenin exon-3 mutations in these two genetic backgrounds. TNF-induced apoptosis was also attenuated in human colon cancer cell lines with genetically activated ß-catenin. However, we found that HCT116 cells, which contain an activated allele of ß-catenin but do not express nuclear ß-catenin, were sensitive to TNF-induced apoptosis. In HCT116 cells, TNF stimulated efficient RB cleavage that preceded chromatin condensation. In contrast, TNF did not induce RB cleavage in colon cancer cells expressing nuclear ß-catenin and these cells could be sensitized to basal and/or TNF-induced apoptosis by the knockdown of ß-catenin or RB. In the apoptosis-resistant colon cancer cells, knockdown of ß-catenin led to a reduction in the RB protein without affecting RB mRNA. Furthermore, ectopic expression of the caspase-resistant, but not the wild-type, RB re-established resistance to TNF-induced caspase activation in colon cancer cells without ß-catenin. Together, these results suggest that nuclear ß-catenin-dependent RB stabilization suppresses TNF-induced apoptosis in caspase-8-positive colon cancer cells.


Asunto(s)
Apoptosis/genética , Neoplasias del Colon/genética , Proteína de Retinoblastoma/genética , beta Catenina/genética , Animales , Caspasa 8/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Ratones , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , beta Catenina/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(3): 1041-6, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23275297

RESUMEN

The molecular etiology of human progenitor reprogramming into self-renewing leukemia stem cells (LSC) has remained elusive. Although DNA sequencing has uncovered spliceosome gene mutations that promote alternative splicing and portend leukemic transformation, isoform diversity also may be generated by RNA editing mediated by adenosine deaminase acting on RNA (ADAR) enzymes that regulate stem cell maintenance. In this study, whole-transcriptome sequencing of normal, chronic phase, and serially transplantable blast crisis chronic myeloid leukemia (CML) progenitors revealed increased IFN-γ pathway gene expression in concert with BCR-ABL amplification, enhanced expression of the IFN-responsive ADAR1 p150 isoform, and a propensity for increased adenosine-to-inosine RNA editing during CML progression. Lentiviral overexpression experiments demonstrate that ADAR1 p150 promotes expression of the myeloid transcription factor PU.1 and induces malignant reprogramming of myeloid progenitors. Moreover, enforced ADAR1 p150 expression was associated with production of a misspliced form of GSK3ß implicated in LSC self-renewal. Finally, functional serial transplantation and shRNA studies demonstrate that ADAR1 knockdown impaired in vivo self-renewal capacity of blast crisis CML progenitors. Together these data provide a compelling rationale for developing ADAR1-based LSC detection and eradication strategies.


Asunto(s)
Adenosina Desaminasa/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Adenosina Desaminasa/genética , Empalme Alternativo , Animales , Crisis Blástica/etiología , Crisis Blástica/genética , Crisis Blástica/metabolismo , Crisis Blástica/patología , Transformación Celular Neoplásica , Progresión de la Enfermedad , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Mediadores de Inflamación/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mieloide de Fase Crónica/genética , Leucemia Mieloide de Fase Crónica/metabolismo , Leucemia Mieloide de Fase Crónica/patología , Ratones , Edición de ARN , Proteínas de Unión al ARN , Transcriptoma , Trasplante Heterólogo , Ensayo de Tumor de Célula Madre
10.
PLoS One ; 7(6): e39725, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768113

RESUMEN

BACKGROUND: Leukemia initiating cells (LIC) contribute to therapeutic resistance through acquisition of mutations in signaling pathways, such as NOTCH1, that promote self-renewal and survival within supportive niches. Activating mutations in NOTCH1 occur commonly in T cell acute lymphoblastic leukemia (T-ALL) and have been implicated in therapeutic resistance. However, the cell type and context specific consequences of NOTCH1 activation, its role in human LIC regeneration, and sensitivity to NOTCH1 inhibition in hematopoietic microenvironments had not been elucidated. METHODOLOGY AND PRINCIPAL FINDINGS: We established humanized bioluminescent T-ALL LIC mouse models transplanted with pediatric T-ALL samples that were sequenced for NOTCH1 and other common T-ALL mutations. In this study, CD34(+) cells from NOTCH1(Mutated) T-ALL samples had higher leukemic engraftment and serial transplantation capacity than NOTCH1(Wild-type) CD34(+) cells in hematopoietic niches, suggesting that self-renewing LIC were enriched within the NOTCH1(Mutated) CD34(+) fraction. Humanized NOTCH1 monoclonal antibody treatment reduced LIC survival and self-renewal in NOTCH1(Mutated) T-ALL LIC-engrafted mice and resulted in depletion of CD34(+)CD2(+)CD7(+) cells that harbor serial transplantation capacity. CONCLUSIONS: These results reveal a functional hierarchy within the LIC population based on NOTCH1 activation, which renders LIC susceptible to targeted NOTCH1 inhibition and highlights the utility of NOTCH1 antibody targeting as a key component of malignant stem cell eradication strategies.


Asunto(s)
Células Madre Neoplásicas/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptor Notch1/metabolismo , Regeneración , Transducción de Señal , Nicho de Células Madre , Adolescente , Animales , Anticuerpos Monoclonales/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Niño , Preescolar , Humanos , Ratones , Mutación/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/trasplante , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Adulto Joven
11.
Syst Biol Reprod Med ; 56(4): 318-26, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20462350

RESUMEN

The aim of this study is to compare the expression patterns of tyrosine kinases and their ligand activators between matched myometrium and leiomyoma tissues. Total RNA extracted from 42 pairs of matched leiomyomal and adjacent myometrial tissues were hybridized to a set of 840 customized oligonucleotide microarrays to compare the expression profiles of 244 selected human genes including 90 tyrosine kinases and 103 ligands. Among the 244 genes surveyed, 38 were found to exhibit differential expression between pairs of myometrium and leiomyoma tissues. Clustering analysis of the expression ratios of these 38 genes from 42 pairs of samples led to the subdivision of fibroid samples into three groups, based in principle on the expression ratios of two peptide ligands, CYR61 and EFNA4. Real-time quantitative RT-PCR measurements of an additional 32 pairs of samples further confirmed the three subgroups. The altered expression of ligand activators between myometrium and leiomyoma suggest that tyrosine kinases regulated by CYR61 and EFNA4 may be exploited as therapeutic targets to develop non-surgical treatments of symptomatic leiomyomas.


Asunto(s)
Perfilación de la Expresión Génica , Leiomioma/enzimología , Proteínas Tirosina Quinasas/genética , Neoplasias Uterinas/enzimología , Adulto , Secuencia de Bases , Cartilla de ADN , Femenino , Humanos , Persona de Mediana Edad , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Reprod Sci ; 16(9): 827-42, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19516078

RESUMEN

Uterine fibroids are the most prevalent benign tumors in women of reproductive age. The current knowledge on the fibroid disease mechanism has derived from studies of the Eker rat model where a unique germ line defect in the tuberous sclerosis 2 (Tsc2) tumor suppressor gene leads to the development of leiomyosarcoma, leiomyoma, and renal cancer. To study fibroids of human origin, we sought to establish fibroid xenografts in immune-compromised mice. We determined that lentiviral-mediated transduction of a green fluorescence protein (GFP)-luciferase (LUC) fusion gene and bioluminescence-based whole animal imaging allowed for the monitoring of transplanted fibroid cells in mice. We used this in vivo imaging approach to test a series of transplantation protocols and found that only freshly dissociated fibroid cells, but not the fibroid-derived smooth muscle cells grown in ex vivo cultures, can generate stable xenografts in subcutaneous Matrigel implants. Formation of the fibroid-xenografts requires the implantation of 17betaestradiol-releasing pellets in the recipient mice. Furthermore, freshly dissociated myometrial cells do not form xenografts under the experimental conditions. The xenograft protocol developed from this study provides an avenue for investigating the pathogenesis and drug responses of human fibroids.


Asunto(s)
Modelos Animales de Enfermedad , Leiomioma/patología , Trasplante de Neoplasias/métodos , Trasplante Heterólogo/métodos , Neoplasias Uterinas/patología , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inyecciones Subcutáneas , Riñón/citología , Lentivirus/genética , Luciferasas/genética , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/trasplante , Transfección
13.
Reprod Sci ; 15(9): 878-94, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19050321

RESUMEN

OBJECTIVE: The estrogen metabolite 2-methoxyestradiol has shown antitumorigenic action in some epithelial tumors. In the present work we investigate its effects in ovarian cancer used alone or in combination with other apoptotic-inducing reagents such as tumor necrosis factor-related apoptosis-inducing ligand. METHODS: To assess the effect of 2-methoxyestradiol, dose response and time courses in ovarian cancer and normal cells were conducted. Apoptosis was confirmed through DNA laddering, by flow cytometry, and Western blotting of proteins involved in the apoptotic cascade. RESULTS: 2-Methoxyestradiol induced apoptosis in ovarian cancer cells but not in normal counterparts. 2-Methoxyestradiol activates both the intrinsic and extrinsic apoptotic pathways. 2-Methoxyestradiol-mediated apoptosis involves reactive oxygen species generation and caspase-dependent and caspase-independent mechanisms. We also demonstrate that 2-methoxyestradiol selectively induces an additive/synergistic apoptotic response in ovarian cancer cells when used in combination with tumor necrosis factor-related apoptosis-inducing ligand. CONCLUSIONS: 2-Methoxyestradiol, alone or in combination with tumor necrosis factor-related apoptosis-inducing ligand, should be considered as a potential treatment for ovarian cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Estradiol/análogos & derivados , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , 2-Metoxiestradiol , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/fisiología , Línea Celular Tumoral , Sinergismo Farmacológico , Estradiol/administración & dosificación , Estradiol/farmacología , Femenino , Humanos , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
14.
Endocr Relat Cancer ; 14(2): 351-68, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17639050

RESUMEN

Cancers of the reproductive tract account for 12% of all malignancies in women. As previous studies have shown that oestrogen metabolites can cause apoptosis, we characterised the effect of oestrogen and oestrogen metabolites on non-cancerous and cancerous human endometrial cells. Herein, we demonstrate that 2-methoxyoestradiol (2ME), but not 17beta-oestradiol, induces apoptosis in cancer cell lines and primary cultured tumours of endometrial origin. In contrast, 2ME had no effect on cell viability of corresponding normal tissue. This ability of 2ME to induce apoptosis does not require oestrogen receptor activation, but is associated with increased entry into the G2/M phases of the cell cycle and the activation of both the intrinsic and the extrinsic apoptotic pathways. The selective behaviour of 2ME on cancerous as opposed to normal tissue may be due to a reduction in 17beta-hydroxysteroid dehydrogenase type II levels in cancer cells and to a differential down-regulation of superoxide dismutase. Furthermore, we demonstrate that pre-treatment with 2ME enhances the sensitivity of reproductive tract cancer cells to the apoptotic drug tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), without the loss in cell viability to normal cells incurred by currently chemotherapeutic drugs. In conclusion, 2ME, alone or in combination with TRAIL, may be an effective treatment for cancers of uterine origin with minimal toxicity to corresponding healthy female reproductive tissue.


Asunto(s)
Apoptosis , Neoplasias Endometriales/enzimología , Estriol/análogos & derivados , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Antineoplásicos/farmacología , Caspasas/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Endometrio/efectos de los fármacos , Endometrio/enzimología , Estriol/farmacología , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
15.
Apoptosis ; 12(1): 73-85, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17136491

RESUMEN

Cancer of the reproductive tract encompasses malignancies of the uterine corpus, cervix, ovary, Fallopian tube, among others and accounts for 15% of female cancer mortalities. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) mediates apoptosis by binding to death receptors and offers a promising cancer treatment. The goal of this study was to investigate and characterize the effect of TRAIL in endometrial cancer cell lines and normal (non-cancerous) epithelial cells of endometrial origin. We also examined the effect of TRAIL in other primary cultured cancers and normal cells of the human female reproductive tract and evaluated if TRAIL mediated apoptosis correlated with death receptors and decoy receptors 1 and 2.Herein, we demonstrate that TRAIL at concentrations which kill cancerous cells, does not mediate apoptosis or alter cell viability in normal human endometrium, ovary, cervix or Fallopian tube. The partial inhibition by a caspase 9 inhibitor and the total inhibition by a caspase 8 inhibitor demonstrates the dependency on the extrinsic apoptotic pathway. The selective mortality does not correlate with the presence of death or decoy receptors. These results suggest that TRAIL may be an effective treatment for endometrial cancer and other female reproductive cancers, with minimal secondary effects on healthy tissue.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Neoplasias de los Genitales Femeninos/patología , Genitales Femeninos/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis/genética , Apoptosis/fisiología , Secuencia de Bases , Línea Celular Tumoral , Células Cultivadas , ADN Complementario/genética , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Neoplasias Endometriales/fisiopatología , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/fisiología , Femenino , Neoplasias de los Genitales Femeninos/genética , Neoplasias de los Genitales Femeninos/fisiopatología , Genitales Femeninos/citología , Genitales Femeninos/fisiología , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Proteínas Recombinantes/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Células Tumorales Cultivadas
16.
Biol Res ; 38(2-3): 245-58, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16238103

RESUMEN

Estrogen and progestin combination in hormone replacement therapy (HRT) increases the incidence of breast cancer, but decreases the endometrial cancer risk of unopposed estrogen. Therefore, a SERM such as Tibolone, that delivers the beneficial, but not the adverse side effects, of steroid hormones would be clinically advantageous. However, data from the Million Women Study suggests that Tibolone increases the risk of both breast and endometrial cancer. Herein, we assessed the estrogenic and progestagenic actions of Tibolone using transvaginal sonography studies and an in vitro model of breast (ZR-75, MCF7) and endometrial cancer (Ishikawa). The known cancer associated proteins (ER, EGFR, STATS, tissue factor and Bcl-xL) were selected for study. Transvaginal sonography demonstrated that postmenopausal women treated with Tibolone displayed a thinner endometrium than in the late proliferative phase, but had a phenotype characteristic of the secretory phase, thus demonstrating the estrogenic and progestagenic actions of this SERM. In vitro, Tibolone acted as an estrogen in downregulating ER and upregulating Bcl-xL, yet as progesterone, increasing STAT5 and tissue factor in breast cancer cells. The increase in tissue factor by Tibolone correlated with its coagulative potential. Interestingly, EGFR was up-regulated by progesterone in the breast and by estrogen in endometrial cells, while Tibolone increased protein levels in both cell types. In conclusion, this study further demonstrates the estrogenic and progestagenic nature of Tibolone. The pattern of regulation of known oncogenes in cells of breast and endometrial origin dictates caution and vigilance in the prescription of Tibolone and subsequent patient monitoring.


Asunto(s)
Biomarcadores de Tumor/análisis , Endometrio/efectos de los fármacos , Moduladores de los Receptores de Estrógeno/farmacología , Estrógenos/metabolismo , Norpregnenos/farmacología , Progestinas/metabolismo , Western Blotting , Neoplasias de la Mama/química , Neoplasias de la Mama/genética , Línea Celular Tumoral , Neoplasias Endometriales/química , Neoplasias Endometriales/genética , Endometrio/diagnóstico por imagen , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Factores de Tiempo , Ultrasonografía
17.
J Clin Endocrinol Metab ; 90(2): 1181-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15562024

RESUMEN

Progesterone in hormonal preparations increases the incidence of breast cancer. Tissue factor (TF), the initiator of the extrinsic coagulation pathway, is associated with metastasis in a wide variety of cancers. We demonstrate herein that TF mRNA and protein are up-regulated by progesterone in the breast cancer cell line ZR-75. Epidermal growth factor, also associated with increased breast cancer risk, did not regulate TF. The increase in TF is both rapid and transient; increasing after 6 h, reaching a maximum at 24 h, before decreasing to basal levels at 72 h. Sucrose gradient experiments demonstrated that TF is located in the heavy fraction of the plasma membrane, although caveolin-1 is not expressed in ZR-75. To understand the physiological implications of an increase in TF, we performed coagulation and invasion assays. An increase in TF corresponded to an increase in procoagulant activity. Furthermore, progesterone increased the invasion of ZR-75 cells through a matrigel, an effect that was blocked by an antibody against TF. Because TF expression is associated with an enhanced risk of metastasis, we postulate that the progesterone-dependent up-regulation of TF provides a survival advantage to burgeoning breast cancer cells and may contribute to the increased risk of cancer associated with combined hormone replacement therapy.


Asunto(s)
Neoplasias de la Mama/genética , Progesterona/farmacología , Tromboplastina/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular , Neoplasias Endometriales , Factor de Crecimiento Epidérmico/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Invasividad Neoplásica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sacarosa , Tromboplastina/aislamiento & purificación
18.
Biol. Res ; 38(2/3): 245-258, 2005. ilus, graf
Artículo en Inglés | LILACS | ID: lil-424728

RESUMEN

Estrogen and progestin combination in hormone replacement therapy (HRT) increases the incidence of breast cancer, but decreases the endometrial cancer risk of unopposed estrogen. Therefore, a SERM such as Tibolone, that delivers the beneficial, but not the adverse side effects, of steroid hormones would be clinically advantageous. However, data from the Million Women Study suggests that Tibolone increases the risk of both breast and endometrial cancer. Herein, we assessed the estrogenic and progestagenic actions of Tibolone using transvaginal sonography studies and an in vitro model of breast (ZR-75, MCF7) and endometrial cancer (Ishikawa). The known cancer associated proteins (ER, EGFR, STAT5, tissue factor and Bcl-xL) were selected for study. Transvaginal sonography demonstrated that postmenopausal women treated with Tibolone displayed a thinner endometrium than in the late proliferative phase, but had a phenotype characteristic of the secretory phase, thus demonstrating the estrogenic and progestagenic actions of this SERM. In vitro, Tibolone acted as an estrogen in downregulating ER and upregulating Bcl-xL, yet as progesterone, increasing STAT5 and tissue factor in breast cancer cells. The increase in tissue factor by Tibolone correlated with its coagulative potential. Interestingly, EGFR was up-regulated by progesterone in the breast and by estrogen in endometrial cells, while Tibolone increased protein levels in both cell types. In conclusion, this study further demonstrates the estrogenic and progestagenic nature of Tibolone. The pattern of regulation of known oncogenes in cells of breast and endometrial origin dictates caution and vigilance in the prescription of Tibolone and subsequent patient monitoring.


Asunto(s)
Humanos , Animales , Femenino , Persona de Mediana Edad , Ratas , Técnicas In Vitro , Menopausia , Terapia de Reemplazo de Estrógeno/efectos adversos , Terapia de Reemplazo de Estrógeno , Terapia de Reemplazo de Hormonas/efectos adversos , Terapia de Reemplazo de Hormonas , Neoplasias Endometriales/inducido químicamente , Neoplasias Endometriales/prevención & control , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/prevención & control , Neoplasias de la Mama/terapia , Progestinas , Western Blotting
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...