Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Blood Cancer ; 67(7): e28284, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32333633

RESUMEN

BACKGROUND: VTP-50469 is a potent inhibitor of the menin-MLL1 interaction and is implicated in signaling downstream of EWSR1-FLI1. PROCEDURE: VTP-50469 was evaluated against seven Ewing sarcoma (EwS) xenograft models and in vitro against EwS cell lines. RESULTS: VTP-50469 showed limited antitumor activity, statistically significantly slowing tumor progression in four tumor models but with no evidence of tumor regression. In vitro, the IC50 concentration was 10 nM for the mixed lineage leukemia (MLL)-rearranged leukemia cell line MV4;11, but > 3 µM for EwS cell lines. CONCLUSIONS: In contrast to its high level of activity against MLL1-rearranged leukemia xenografts, VTP-50469 shows little activity against EwS models.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , N-Metiltransferasa de Histona-Lisina/efectos de los fármacos , Proteína de la Leucemia Mieloide-Linfoide/efectos de los fármacos , Proteínas Proto-Oncogénicas/efectos de los fármacos , Sarcoma de Ewing/tratamiento farmacológico , Animales , Antineoplásicos/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Pediatría , Proteínas Proto-Oncogénicas/metabolismo , Sarcoma de Ewing/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Pediatr Blood Cancer ; 66(8): e27820, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31099166

RESUMEN

BACKGROUND: Entinostat, a selective class I histone deacetylase inhibitor, has been reported to enhance the activity of cytotoxic agents and suppress expression of PAX3-FOXO1 in alveolar rhabdomyosarcoma (ARMS). PROCEDURES: Entinostat was tested against three rhabdomyosarcoma cell lines using 96-hour drug exposure. Entinostat alone or in binary combination with vincristine, actinomycin D or cyclophosphamide was tested in ARMS and two embryonal rhabdomyosarcoma (ERMS) xenograft models. Tumor growth was measured at weekly intervals. Drug-induced changes in acetylated histone H3(K9) and entinostat pharmacokinetics were determined. RESULTS: In vitro, the IC50 concentration of entinostat ranged from 280 to 1300 nM. In vivo, entinostat significantly inhibited the growth of only Rh10 xenografts. For most studies, entinostat did not potentiate the activity of the cytotoxic agent. Exceptions included the vincristine and entinostat combination for Rh10 and the entinostat and actinomycin D combination for Rh10 and Rh18, although the effects were modest. For Rh18, the combination of entinostat with vincristine showed evidence of an antagonistic interaction compared with single-agent vincristine. Pharmacokinetic studies showed the average Cmax was 569.4 ng/mL (1.51 µM) with Tmax at 15 minutes, and total exposure (AUC0-12 h ) was 435.6 h × ng/mL. Entinostat treatment increased acetylated histone H3. CONCLUSIONS: Entinostat demonstrated modest antitumor activity in only one of four models at dose and shedule that gave drug exposures relevant to human treatment. The addition of entinostat to standard-of-care cytotoxic agents was in most instances no more effective than the cytotoxic agents used alone. Entinostat demonstrated target inhibition with increased histone 2A acetylation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Nivel de Atención , Animales , Benzamidas/administración & dosificación , Neoplasias de la Mama/patología , Ciclofosfamida/administración & dosificación , Dactinomicina/administración & dosificación , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Piridinas/administración & dosificación , Células Tumorales Cultivadas , Vincristina/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Pediatr Blood Cancer ; 65(2)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29080385

RESUMEN

BACKGROUND: Integrating molecularly targeted agents with cytotoxic drugs used in curative treatment of pediatric cancers is complex. An evaluation was undertaken with the ERBB3/Her3-specific antibody patritumab (P) either alone or with the ERBB1/epidermal growth factor receptor inhibitor erlotinib (E) in combination with standard cytotoxic agents, cisplatin, vincristine, and cyclophosphamide, in pediatric sarcoma xenograft models that express receptors and ligands targeted by these agents. PROCEDURES: Tumor models were selected based upon ERBB3 expression and phosphorylation, and ligand (heregulin) expression. Patritumab, E, or these agents combined was evaluated without or with concomitant cytotoxic agents using procedures developed by the Pediatric Preclinical Testing Program. RESULTS: Full doses of cytotoxic agents were tolerated when combined with P, whereas dose reductions of 25% (vincristine, cisplatin) or 50% (cyclophosphamide) were required when combined with P + E. Patritumab, E alone, or in combination did not significantly inhibit growth of any tumor model, except for Rh18 xenografts (E alone). Patritumab had no single-agent activity and marginally enhanced the activity of vincristine and cisplatin only in Ewing sarcoma ES-4. P + E did not increase the antitumor activity of vincristine or cisplatin, whereas dose-reduced cyclophosphamide was significantly less active than cyclophosphamide administered at its maximum tolerated dose when combined with P + E. CONCLUSIONS: P had no single-agent activity, although it marginally potentiated the activity of vincristine and cisplatin in one of three models studied. However, the addition of E necessitated dose reduction of each cytotoxic agent, abrogating the enhancement observed with P alone.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Óseas/tratamiento farmacológico , Clorhidrato de Erlotinib/farmacología , Sarcoma de Ewing/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados , Neoplasias Óseas/metabolismo , Anticuerpos ampliamente neutralizantes , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones SCID , Receptor ErbB-3/antagonistas & inhibidores , Receptor ErbB-3/metabolismo , Sarcoma de Ewing/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancers (Basel) ; 16(11)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38893160

RESUMEN

Malignant rhabdoid tumors (MRTs) are among the most aggressive and treatment-resistant malignancies affecting infants, originating in the kidney, brain, liver, and soft tissues. The 5-year event-free survival rate for these cancers is a mere 20%. In nearly all cases of MRT, the SMARCB1 gene (occasionally SMARCA4)-a pivotal component of the SWI/SNF chromatin remodeling complex-is homozygously deleted, although the precise etiology of these tumors remains unknown. While young patients with localized MRT generally show improved outcomes, especially those who are older and have early-stage disease, the overall prognosis remains poor despite optimal standard treatments. This highlights the urgent need for more effective treatment strategies. We investigated the antitumor activity of a PARP1 inhibitor (talazoparib, TLZ) combined with a DNA alkylating agent (temozolomide, TMZ) in MRT xenograft models. PARP1 is a widely targeted molecule in cancer treatment and, beyond its role in DNA repair, it participates in transcriptional regulation by recruiting chromatin remodeling complexes to modulate DNA accessibility for RNA polymerases. To widen the therapeutic window of the drug combination, we employed PEGylated TLZ (PEG~TLZ), which has been reported to reduce systemic toxicity through slow drug release. Remarkably, our findings indicate that five out of six MRT xenografts exhibited an objective response to PEG~TLZ+TMZ therapy. Significantly, the loss of SMARCB1 was found to confer a protective effect, correlating with higher expression levels of DNA damage and repair proteins in SMARCB1-deficient MRT cells. Additionally, we identified MGMT as a potential biomarker indicative of in vivo MRT response to PEG~TLZ+TMZ therapy. Moreover, our analysis revealed alterations in signaling pathways associated with the observed antitumor efficacy. This study presents a novel and efficacious therapeutic approach for MRT, along with a promising candidate biomarker for predicting tumor response.

5.
Elife ; 122023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37266578

RESUMEN

In embryonal rhabdomyosarcoma (ERMS) and generally in sarcomas, the role of wild-type and loss- or gain-of-function TP53 mutations remains largely undefined. Eliminating mutant or restoring wild-type p53 is challenging; nevertheless, understanding p53 variant effects on tumorigenesis remains central to realizing better treatment outcomes. In ERMS, >70% of patients retain wild-type TP53, yet mutations when present are associated with worse prognosis. Employing a kRASG12D-driven ERMS tumor model and tp53 null (tp53-/-) zebrafish, we define wild-type and patient-specific TP53 mutant effects on tumorigenesis. We demonstrate that tp53 is a major suppressor of tumorigenesis, where tp53 loss expands tumor initiation from <35% to >97% of animals. Characterizing three patient-specific alleles reveals that TP53C176F partially retains wild-type p53 apoptotic activity that can be exploited, whereas TP53P153Δ and TP53Y220C encode two structurally related proteins with gain-of-function effects that predispose to head musculature ERMS. TP53P153Δ unexpectedly also predisposes to hedgehog-expressing medulloblastomas in the kRASG12D-driven ERMS-model.


Asunto(s)
Neoplasias Cerebelosas , Rabdomiosarcoma Embrionario , Animales , Carcinogénesis , Mutación , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Rabdomiosarcoma Embrionario/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
6.
iScience ; 26(11): 108171, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37915590

RESUMEN

Patient-derived xenografts (PDX) remain valuable models for understanding the biology and for developing novel therapeutics. To expand current PDX models of childhood leukemia, we have developed new PDX models from Hispanic patients, a subgroup with a poorer overall outcome. Of 117 primary leukemia samples obtained, successful engraftment and serial passage in mice were achieved in 82 samples (70%). Hispanic patient samples engrafted at a rate (51/73, 70%) that was similar to non-Hispanic patient samples (31/45, 70%). With a new algorithm to remove mouse contamination in multi-omics datasets including methylation data, we found PDX models faithfully reflected somatic mutations, copy-number alterations, RNA expression, gene fusions, whole-genome methylation patterns, and immunophenotypes found in primary tumor (PT) samples in the first 50 reported here. This cohort of characterized PDX childhood leukemias represents a valuable resource in that germline DNA sequencing has allowed the unambiguous determination of somatic mutations in both PT and PDX.

7.
Nat Commun ; 14(1): 7600, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37990009

RESUMEN

Subcutaneous patient-derived xenografts (PDXs) are an important tool for childhood cancer research. Here, we describe a resource of 68 early passage PDXs established from 65 pediatric solid tumor patients. Through genomic profiling of paired PDXs and patient tumors (PTs), we observe low mutational similarity in about 30% of the PT/PDX pairs. Clonal analysis in these pairs show an aggressive PT minor subclone seeds the major clone in the PDX. We show evidence that this subclone is more immunogenic and is likely suppressed by immune responses in the PT. These results suggest interplay between intratumoral heterogeneity and antitumor immunity may underlie the genetic disparity between PTs and PDXs. We further show that PDXs generally recapitulate PTs in copy number and transcriptomic profiles. Finally, we report a gene fusion LRPAP1-PDGFRA. In summary, we report a childhood cancer PDX resource and our study highlights the role of immune constraints on tumor evolution.


Asunto(s)
Neoplasias , Animales , Niño , Humanos , Xenoinjertos , Neoplasias/genética , Neoplasias/patología , Transcriptoma/genética , Mutación , Modelos Animales de Enfermedad , Genómica/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Cell Biol ; 27(1): 324-39, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17074815

RESUMEN

SnoN is an important negative regulator of transforming growth factor beta signaling through its ability to interact with and repress the activity of Smad proteins. It was originally identified as an oncoprotein based on its ability to induce anchorage-independent growth in chicken embryo fibroblasts. However, the roles of SnoN in mammalian epithelial carcinogenesis have not been well defined. Here we show for the first time that SnoN plays an important but complex role in human cancer. SnoN expression is highly elevated in many human cancer cell lines, and this high level of SnoN promotes mitogenic transformation of breast and lung cancer cell lines in vitro and tumor growth in vivo, consistent with its proposed pro-oncogenic role. However, this high level of SnoN expression also inhibits epithelial-to-mesenchymal transdifferentiation. Breast and lung cancer cells expressing the shRNA for SnoN exhibited an increase in cell motility, actin stress fiber formation, metalloprotease activity, and extracellular matrix production as well as a reduction in adherens junction proteins. Supporting this observation, in an in vivo breast cancer metastasis model, reducing SnoN expression was found to moderately enhance metastasis of human breast cancer cells to bone and lung. Thus, SnoN plays both pro-tumorigenic and antitumorigenic roles at different stages of mammalian malignant progression. The growth-promoting activity of SnoN appears to require its ability to bind to and repress the Smad proteins, while the antitumorigenic activity can be mediated by both Smad-dependent and Smad-independent pathways and requires the activity of small GTPase RhoA. Our study has established the importance of SnoN in mammalian epithelial carcinogenesis and revealed a novel aspect of SnoN function in malignant progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias Glandulares y Epiteliales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína de Unión al GTP cdc42/fisiología , Proteína de Unión al GTP rhoA
9.
Clin Cancer Res ; 26(12): 3012-3023, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32184294

RESUMEN

PURPOSE: Vincristine combined with camptothecin derivatives showed synergy in preclinical pediatric cancer models, and the combinations are effective in treatment of childhood solid tumors. We determined whether the synergy between vincristine and irinotecan extends to eribulin, another microtubule inhibitor. EXPERIMENTAL DESIGN: Vincristine or eribulin, alone or combined with irinotecan, was studied in 12 xenograft models. Tumor regression and time to event were used to assess antitumor activity. Pharmacodynamic studies and RNA sequencing (RNA-seq) were conducted 24 and 144 hours after single-agent or combination treatment. Effects on vascular development were studied in Matrigel plugs implanted in mice. The interaction between binary combinations was examined in vitro. RESULTS: Eribulin combined with irinotecan was more effective than vincristine-irinotecan in 6 of 12 models. Pharmacodynamic markers induced by eribulin (phospho-histone H3) and irinotecan (γ-H2A.X) were abrogated in combination-treated tumors. The predominant RNA-seq signature in combination-treated tumors was activation of the TP53 pathway with increased nuclear TP53. Massive apoptosis was observed 24 hours only after treatment with the eribulin combination. In vitro, neither combination showed interaction using combination index analysis. Eribulin alone and the combination caused alterations in developing vasculature. CONCLUSIONS: The eribulin combination is very active in these xenograft models, but not synergistic in vitro. The combination reduced pharmacodynamic markers indicative of single-agent mechanisms but in tumors, dramatically activated the TP53 pathway. Although a mechanism for in vivo synergy requires further study, it is possible that eribulin-induced inhibition of microtubule dynamics enhances irinotecan-induced nuclear accumulation of TP53, leading to rapid cell death.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Renales/tratamiento farmacológico , Rabdomiosarcoma/tratamiento farmacológico , Tumor de Wilms/tratamiento farmacológico , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Femenino , Furanos/administración & dosificación , Perfilación de la Expresión Génica , Humanos , Irinotecán/administración & dosificación , Cetonas/administración & dosificación , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Ratones , Ratones SCID , Pronóstico , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Células Tumorales Cultivadas , Vincristina/administración & dosificación , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Tumor de Wilms/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Res ; 67(12): 5737-46, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575140

RESUMEN

Increasing evidence points to an active stromal involvement in cancer initiation and progression. Cytokines derived from tumor cells are believed to modulate stromal cells to produce growth and angiogenic factors, which in turn provide the tumor with the necessary microenvironment for expansion and invasion. Transforming growth factor beta (TGFbeta) has been implicated as a candidate cytokine to mediate this communication. However, how its signaling in stromal cells regulates tumorigenesis and tumor progression remains unresolved. We show that normal, presenescent fibroblasts or prostate stromal cells cotransplanted with prostate carcinoma cells s.c. into nude mice reduced tumor latency and accelerated tumor growth. When their TGFbeta signaling was blocked, the fibroblasts and stromal cells still stimulated tumor initiation but no longer supported tumor growth as control cells did. The loss of the tumor growth-promoting activity of the stromal cells with attenuated TGFbeta signaling was not associated with altered cellular senescence or tumor angiogenicity. TGFbeta and the medium conditioned by the prostate carcinoma cells stimulated myofibroblast differentiation of the intact stromal cells, but not the stromal cells with attenuated TGFbeta signaling. Gene microarray and quantitative reverse transcription-PCR analyses showed that TGFbeta up-regulated a host of genes in stromal cells that are involved in tissue remodeling and wound healing. Thus, our study provides evidence for TGFbeta as a supporting agent in tumor progression through the induction of a perpetual wound healing process in the tumor microenvironment.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Próstata/metabolismo , Transducción de Señal/fisiología , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/fisiología , Animales , Western Blotting , Comunicación Celular/fisiología , Proliferación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Próstata/citología , Próstata/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
11.
Cancer Res ; 66(13): 6714-21, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16818646

RESUMEN

Transforming growth factor-beta (TGF-beta) signaling has been shown to promote invasion and metastasis in various models of human cancers. In this study, we investigated the efficacy of a TGF-beta type I receptor kinase inhibitor (TbetaRI-I) to limit early systemic metastases in an orthotopic xenograft model of lung metastasis and in an intracardiac injection model of experimental bone and lung metastasis using human breast carcinoma MDA-MB-435-F-L cells, a highly metastatic variant of human breast cancer MDA-MB-435 cells, expressing the enhanced green fluorescent protein (EGFP). Treatment of the cells with the TbetaRI-I had no effect on their growth but blocked TGF-beta-stimulated expression of integrin alpha(v)beta(3) and cell migration in vitro. Systemic administration of the TbetaRI-I via i.p. injection effectively reduced the number and size of the lung metastasis in both orthotopic xenograft and experimental metastasis models with no effects on primary tumor growth rate compared with controls. TbetaRI-I treatment also reduced the incidence of widespread early skeletal metastases in the femur, tibia, mandible, and spine detected by whole-body EGFP fluorescence imaging. Tumor burden in femora and tibiae was also reduced after TbetaRI-I treatment as detected by histomorphometry analysis compared with the placebo controls. Our results indicate for the first time that abrogation of TGF-beta signaling by systemic administration of the TbetaRI-I can inhibit both early lung and bone metastasis in animal model systems and suggest antimetastatic therapeutic potential of the TbetaRI-I.


Asunto(s)
Receptores de Activinas Tipo I/antagonistas & inhibidores , Neoplasias Óseas/prevención & control , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias Pulmonares/prevención & control , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , Neoplasias Óseas/enzimología , Neoplasias Óseas/secundario , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/secundario , Ratones , Proteínas Serina-Treonina Quinasas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Res ; 62(16): 4690-5, 2002 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12183427

RESUMEN

We have demonstrated previously that ectopic expression of a soluble betaglycan, also known as transforming growth factor (TGF) beta type III receptor, can suppress the malignant properties of human carcinoma cells by antagonizing the tumor-promoting activity of TGF-beta (A. Bandyopadhyay et al., Cancer Res., 59: 5041-5046, 1999). In the current study, we investigated the potential therapeutic utility of a recombinant preparation of human and rat soluble betaglycan (sBG). Purified recombinant human sBG showed similar properties to its rat counterpart (M. M. Vilchis-Landeros et al., Biochem J., 355: 215-222, 2001). It bound TGF-beta with high affinity and isoform selectivity and neutralized the activity of TGF-beta(1) in two bioassays. Peritumoral (50 micro g/tumor, twice a week) or i.p. (100 micro g/animal, every alternate day) injection of sBG into human breast carcinoma MDA-MB-231 xenograft-bearing athymic nude mice significantly inhibited the tumor growth. The administration of sBG also reduced metastatic incidence and colonies in the lungs. The tumor-inhibitory activity of sBG was found to be associated with the inhibition of angiogenesis. Systemic sBG treatment significantly reduced tumor microvessel density detected with histological analyses and CD-31 immunostainings, as well as tumor blood volume measured with hemoglobin content. In an in vitro angiogenesis assay, treatment with the recombinant sBG significantly reduced the ability of human dermal microvascular endothelial cells to form a capillary tube-like structure on Matrigel. These findings support the conclusion that sBG treatment suppresses tumor growth and metastasis, at least in part by inhibiting angiogenesis. As such, it could be a useful therapeutic agent to antagonize the tumor-promoting activity of TGF-beta.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteoglicanos/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , División Celular/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Inhibidores de Crecimiento/farmacología , Humanos , Ratones , Ratones Desnudos , Visón , Ratas , Receptores de Factores de Crecimiento Transformadores beta , Proteínas Recombinantes/farmacología , Solubilidad , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oncotarget ; 7(26): 39097-39107, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27259243

RESUMEN

Exposure to estrogen has long been associated with an increased risk of developing breast cancer. However, how estrogen signaling promotes breast carcinogenesis remains elusive. Senescence is known as an important protective response to oncogenic events. We aimed to elucidate the role of estrogen receptor alpha (ERα) on senescence in transformed human mammary epithelial cells and breast cancer cells. Our results show that ectopic expression of oncoprotein H-ras-V12 in immortalized human mammary epithelial cells (HMEC) significantly inhibited the phosphorylation of the retinoblastoma protein (Rb) and increased the activity of the senescence-associated beta-galactosidase (SA-ß-Gal). These senescence-like phenotypes were reversed by ectopic expression of ERα. Similar inhibition of the H-ras-V12-induced SA-ß-Gal activity by ERα was also observed in the human mammary epithelial MCF-10A cells. Co-expression of ERα and H-ras-V12 resulted in HMEC anchorage-independent growth in vitro and tumor formation in vivo. Furthermore, inhibition of ERα expression induced senescence-like phenotypes in ERα positive human breast cancer cells such as increased activity of SA-ß-Gal, decreased phosphorylation of RB, and loss of mitogenic activity. Thus, the suppression of cellular senescence induced by oncogenic signals may be a major mechanism by which ERα promotes breast carcinogenesis.


Asunto(s)
Mama/citología , Transformación Celular Neoplásica , Senescencia Celular , Células Epiteliales/citología , Receptor alfa de Estrógeno/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular , ADN/metabolismo , Estrógenos/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Oncogenes , Fenotipo , Fosforilación , Transfección
14.
Springerplus ; 5: 140, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26933638

RESUMEN

Murine mammary stem/progenitor cell isolation has been routinely used in many laboratories, yet direct comparison among different methods is lacking. In this study, we compared two frequently used digestion methods and three sets of frequently used surface markers for their efficiency in enriching mammary stem and progenitor cells in two commonly used mouse strains, C57BL/6J and FVB. Our findings revealed that the slow overnight digestion method using gentle collagenase/hyaluronidase could be easily adopted and yielded reliable and consistent results in different batches of animals. In contrast, the different fast digestion protocols, as described in published studies, yielded high percent of non-epithelial cells with very few basal epithelial cells liberated in our hands. The three sets of markers tested in our hands reveal rather equally efficiency in separating luminal and basal cells if same fluorochrome conjugations were used. However, the tendency of non-epithelial cell inclusion in the basal cell gate was highest in samples profiled by CD24/CD29 and lowest in samples profiled by CD49f/EpCAM, this is especially true in mammary cells isolated from C57BL/6J mice. This finding will have significant implication when sorted basal cells are used for subsequent gene expression analysis.

15.
Sci Rep ; 6: 32190, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27558284

RESUMEN

Age is the number one risk factor for breast cancer, yet the underlying mechanisms are unexplored. Age-associated mammary stem cell (MaSC) dysfunction is thought to play an important role in breast cancer carcinogenesis. Non-human primates with their close phylogenetic relationship to humans provide a powerful model system to study the effects of aging on human MaSC. In particular, the common marmoset monkey (Callithrix jacchus) with a relatively short life span is an ideal model for aging research. In the present study, we characterized for the first time the mammary epithelial stem/progenitor cells in the common marmoset. The MaSC-enriched cells formed four major types of morphologically distinct colonies when cultured on plates pre-seeded with irradiated NIH3T3 fibroblasts, and were also capable of forming mammospheres in suspension culture and subsequent formation of 3D organoids in Matrigel culture. Most importantly, these 3D organoids were found to contain stem/progenitor cells that can undergo self-renewal and multi-lineage differentiation both in vitro and in vivo. We also observed a significant decrease of luminal-restricted progenitors with age. Our findings demonstrate that common marmoset mammary stem/progenitor cells can be isolated and quantified with established in vitro and in vivo assays used for mouse and human studies.


Asunto(s)
Envejecimiento/fisiología , Callithrix/fisiología , Glándulas Mamarias Animales/citología , Células Madre/fisiología , Animales , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Células Epiteliales/fisiología , Femenino , Cadenas alfa de Integrinas/metabolismo , Glándulas Mamarias Animales/fisiología , Ratones SCID , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Células Madre/citología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Aging (Albany NY) ; 8(11): 2754-2776, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27852980

RESUMEN

Breast cancer incidence increases during aging, yet the mechanism of age-associated mammary tumorigenesis is unclear. Mammary stem cells are believed to play an important role in breast tumorigenesis, but how their function changes with age is unknown. We compared mammary epithelial cells isolated from young and old mammary glands of different cohorts of C57BL6/J and BALB/c mice, and our findings revealed that old mammary glands were characterized by increased basal cell pool comprised of mostly CD49fhi cells, altered luminal-to-basal cell ratio, and irregular ductal morphology. More interestingly, basal stem cells in old mice were increased in frequency, but showed a functional decline of differentiation and increased neoplastic transformation potential. Gene signature enrichment analysis revealed a significant enrichment of a luminal cell gene expression signature in the basal stem cell-enriched population from old mice, suggesting some luminal cells were expressing basal markers. Immunofluorescence staining confirmed the presence of luminal cells with high CD49f expression in hyperplastic lesions implicating these cells as undergoing luminal to basal phenotypic changes during aging. Whole transcriptome analysis showed elevated immune and inflammatory responses in old basal stem cells and stromal cells, which may be the underlying cause for increased CD49fhi basal-like cells in aged glands.


Asunto(s)
Envejecimiento/patología , Transformación Celular Neoplásica/patología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/patología , Células Madre/patología , Factores de Edad , Envejecimiento/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Femenino , Perfilación de la Expresión Génica , Inflamación/metabolismo , Inflamación/patología , Integrina alfa6/metabolismo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos BALB C , Células Madre/metabolismo
17.
Oncogene ; 21(49): 7514-23, 2002 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-12386814

RESUMEN

Using a cell model system established by ectopic expression of a soluble TGFbeta type III receptor (sRIII) containing the whole extracellular domain of the type III receptor in human breast cancer MDA-MB-231 cells, we observed that the expression of sRIII antagonized TGFbeta activity and inhibited both anchorage-dependent and anchorage-independent cell growth. Further studies revealed that sRIII expression induced apoptosis both in vitro and in vivo. Treatment with TGFbeta neutralizing antibodies or a recombinant human sRIII also induced apoptosis in the MDA-MB-231 parental cells, suggesting that the increased apoptosis after sRIII expression was specifically due to antagonization of autocrine TGFbeta signaling. Western blotting showed that sRIII clones had a higher PTEN expression level than the control cells did. Treatment with TGFbeta(1) decreased PTEN and inhibited apoptosis in sRIII cells to a level similar to that in the control cells. sRIII clones also showed a lower level of phosphorylated-Akt than the control cells, consistent with the inhibitory activity of PTEN on Akt activation. Treatment with LY294002, a specific inhibitor of Akt activator, phosphatidylinositol 3-kinase, also induced apoptosis in a dose dependent manner in the control cells. Our results suggest that autocrine TGFbeta signaling is necessary for the growth and survival of MDA-MB-231 cells.


Asunto(s)
Neoplasias de la Mama/patología , División Celular/fisiología , Supervivencia Celular/fisiología , Proteínas Serina-Treonina Quinasas , Factor de Crecimiento Transformador beta/fisiología , Apoptosis/fisiología , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Humanos , Proteoglicanos/genética , Proteoglicanos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
18.
Oncogene ; 21(22): 3541-51, 2002 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-12032856

RESUMEN

TGFbeta overexpression in human cancer cells has been shown to promote tumor progression. In the present study, we sought to determine whether sequestration of endogenous TGFbeta by the expression of a soluble TGFbeta type III receptor (sRIII), can reduce malignancy in human carcinoma cells and whether the tumor-suppressive activity of sRIII is associated with the inhibition of angiogenesis. Ectopic expression of sRIII significantly inhibited the growth of tumors formed by human colon carcinoma HCT116 and breast carcinoma MDA-MB-435 cells in nude mice. It also reduced the metastatic potential of the MDA-MB-435 cells. Thus, endogenous TGFbeta appears to be necessary for the progression of these two carcinomas. Furthermore, when the tumor cells were mixed with Matrigel and embedded subcutaneously in nude mice, the blood volume in Matrigel plugs containing sRIII-expressing cells as indicated by hemoglobin levels was significantly lower than that in Matrigel plugs containing the respective control cells. Blood vessel counts in paraffin sections of the Matrigel plugs containing sRIII-expressing cells were also significantly lower than those in paraffin sections of the Matrigel plugs containing control cells. Treatment of human endothelial cells with a recombinant sRIII significantly inhibited their ability to form a capillary web structure on Matrigel. These results for the first time indicate that the sRIII-induced tumor suppression appears to be in part due to the inhibition of angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/genética , Carcinoma/terapia , Neovascularización Patológica/terapia , Proteoglicanos/química , Proteoglicanos/genética , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/genética , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Carcinoma/irrigación sanguínea , Carcinoma/patología , División Celular , Línea Celular , Células Cultivadas , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Cinética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Estructura Terciaria de Proteína , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transfección , Células Tumorales Cultivadas
19.
Cancer Biol Ther ; 4(2): 168-74, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15655343

RESUMEN

We have developed a model system of late stage metastatic progression by isolating a highly malignant variant of human breast cancer cells from the parental MDA-MB-435 cell line. These cells, isolated from early lung metastasis, displayed increased anchorage independent growth in vitro and when transplanted ortho-topically into nude mice showed accelerated tumor growth rate and early lung spontaneous metastasis when compared to its parental counterpart. These cells, designated as MDA-MB-435-F-L, also showed intense wide spread early skeletal metastasis in vertebrae, mandible, femur, tibia and skull as detected by fluorescence imaging in an experimental bone metastasis model. Gene expression profiles from cDNA microarray showed up or downregulation of the expression of several significant genes regulating angiogenesis, apoptosis, ECM remodeling and metastasis in the MDA-MB-435-F-L cells in comparison to the parental cells. Among the up or downregulated genes, some have also been implicated in the survival of breast cancer patients. As such, the candidate genes selected in this breast cancer progression model system may serve as biomarkers of metastatic progression and also as potential tumor targets for breast cancer therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/genética , Neoplasias de la Mama/genética , Perfilación de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Animales , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Femenino , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Neoplasias Experimentales , Análisis de Secuencia por Matrices de Oligonucleótidos , Trasplante Heterólogo , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
20.
PLoS One ; 10(4): e0121136, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25837259

RESUMEN

In postmenopausal women, local estrogen produced by adipose stromal cells in the breast is believed to support estrogen receptor alpha (ERα) positive breast cancer cell survival and growth. This raises the question of how the ERα positive metastatic breast cancer cells survive after they enter blood and lymph circulation, where estrogen level is very low in postmenopausal women. In this study, we show that the aromatase expression increased when ERα positive breast cancer cells were cultured in suspension. Furthermore, treatment with the aromatase substrate, testosterone, inhibited suspension culture-induced apoptosis whereas an aromatase inhibitor attenuated the effect of testosterone suggesting that suspended circulating ERα positive breast cancer cells may up-regulate intracrine estrogen activity for survival. Consistent with this notion, a moderate level of ectopic aromatase expression rendered a non-tumorigenic ERα positive breast cancer cell line not only tumorigenic but also metastatic in female nude mice without exogenous estrogen supplementation. The increased malignant phenotype was confirmed to be due to aromatase expression as the growth of orthotopic tumors regressed with systemic administration of an aromatase inhibitor. Thus, our study provides experimental evidence that aromatase plays an important role in the survival of metastatic ERα breast cancer cells by suppressing anoikis.


Asunto(s)
Aromatasa/genética , Neoplasias Óseas/genética , Neoplasias de la Mama/genética , Carcinoma Ductal/genética , Receptor alfa de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , Animales , Anoicis/genética , Aromatasa/metabolismo , Inhibidores de la Aromatasa/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/enzimología , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Carcinoma Ductal/tratamiento farmacológico , Carcinoma Ductal/enzimología , Carcinoma Ductal/secundario , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Estrógenos/biosíntesis , Femenino , Humanos , Letrozol , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Nitrilos/farmacología , Transducción de Señal , Testosterona/metabolismo , Testosterona/farmacología , Triazoles/farmacología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA