Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Med ; 23(6): 2487-2502, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36764998

RESUMEN

Rhabdomyosarcoma (RMS) is an aggressive rare neoplasm that derives from mesenchymal cells, which frequently develops resistance to the current therapies and the formation of metastases. Thus, new therapies are needed. The alteration of iron metabolism in cancer cells was effective in reducing the progression of many tumors but not yet investigated in RMS. Here we investigated the effect of iron modulation in RMS both in vitro and in vivo. We first characterized the most used RMS cell lines representing the most common subtypes, embryonal (ERMS, RD cells) and alveolar (ARMS, RH30 cells), for their iron metabolism, in basal condition and in response to its modulation. Then we investigated the effects of both iron overload and chelation strategies in vitro and in vivo. RMS cell lines expressed iron-related proteins, even if at lower levels compared to hepatic cell lines and they are correctly modulated in response to iron increase and deprivation. Interestingly, the treatment with different doses of ferric ammonium citrate (FAC, as iron source) and with deferiprone (DFP, as iron chelator), significantly affected the cell viability of RD and RH30. Moreover, iron supplementation (in the form of iron dextran) or iron chelation (in the form of DFP) were also effective in vivo in inhibiting the tumor mass growth both derived from RD and RH30 with iron chelation treatment the most effective one. All the data suggest that the iron modulation could be a promising approach to overcome the RMS tumor growth. The mechanism of action seems to involve the apoptotic cell death for both iron supplementation and chelation with the concomitant induction of ferroptosis in the case of iron supplementation.


Asunto(s)
Rabdomiosarcoma , Humanos , Línea Celular Tumoral , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Apoptosis , Hierro , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico
2.
BMC Cancer ; 22(1): 725, 2022 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-35780096

RESUMEN

BACKGROUND: Metastatic soft tissue sarcoma (STS) are a heterogeneous group of malignancies which are not curable with chemotherapy alone. Therefore, understanding the molecular mechanisms of sarcomagenesis and therapy resistance remains a critical clinical need. ASPP2 is a tumor suppressor, that functions through both p53-dependent and p53-independent mechanisms. We recently described a dominant-negative ASPP2 isoform (ASPP2κ), that is overexpressed in human leukemias to promote therapy resistance. However, ASPP2κ  has never been studied in STS.  MATERIALS AND METHODS: Expression of ASPP2κ was quantified in human rhabdomyosarcoma tumors using immunohistochemistry and qRT-PCR from formalin-fixed paraffin-embedded (FFPE) and snap-frozen tissue. To study the functional role of ASPP2κ in rhabdomyosarcoma, isogenic cell lines were generated by lentiviral transduction with short RNA hairpins to silence ASPP2κ expression. These engineered cell lines were used to assess the consequences of ASPP2κ silencing on cellular proliferation, migration and sensitivity to damage-induced apoptosis. Statistical analyses were performed using Student's t-test and 2-way ANOVA. RESULTS: We found elevated ASPP2κ mRNA in different soft tissue sarcoma cell lines, representing five different sarcoma sub-entities. We found that ASSP2κ mRNA expression levels were induced in these cell lines by cell-stress. Importantly, we found that the median ASPP2κ expression level was higher in human rhabdomyosarcoma in comparison to a pool of tumor-free tissue. Moreover, ASPP2κ levels were elevated in patient tumor samples versus adjacent tumor-free tissue within individual patients. Using isogenic cell line models with silenced ASPP2κ expression, we found that suppression of ASPP2κ enhanced chemotherapy-induced apoptosis and attenuated cellular proliferation. CONCLUSION: Detection of oncogenic ASPP2κ in human sarcoma provides new insights into sarcoma tumor biology. Our data supports the notion that ASPP2κ promotes sarcomagenesis and resistance to therapy. These observations provide the rationale for further evaluation of ASPP2κ as an oncogenic driver as well as a prognostic tool and potential therapeutic target in STS.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Carcinogénesis , Rabdomiosarcoma , Sarcoma , Neoplasias de los Tejidos Blandos , Empalme Alternativo , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/fisiología , Humanos , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Sarcoma/genética , Sarcoma/metabolismo , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
BMC Complement Med Ther ; 21(1): 136, 2021 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-33947373

RESUMEN

BACKGROUND: Prevention of metastatic invasion is one of the main challenges in the treatment of alveolar rhabdomyosarcoma. Still the therapeutic options are limited. Therefore, an anti-tumor screening was initiated focusing on the anti-metastatic and anti-invasion properties of selected medicinal plant extracts and phytoestrogens, already known to be effective in the prevention and treatment of different cancer entities. METHODS: Treatment effects were first evaluated by cell viability, migration, invasion, and colony forming assays on the alveolar rhabdomyosarcoma cell line RH-30 in comparison with healthy primary cells. RESULTS: Initial anti-tumor screenings of all substances analyzed in this study, identified the plant extract of Vincetoxicum arnottianum (VSM) as the most promising candidate, harboring the highest anti-metastatic potential. Those significant anti-motility properties were proven by a reduced ability for migration (60%), invasion (99%) and colony formation (61%) under 48 h exposure to 25 µg/ml VSM. The restricted motility features were due to an induction of the stabilization of the cytoskeleton - actin fibers were 2.5-fold longer and were spanning the entire cell. Decreased proliferation (PCNA, AMT, GCSH) and altered metastasis (e. g. SGPL1, CXCR4, stathmin) marker expression on transcript and protein level confirmed the significant lowered tumorigenicity under VSM treatment. Finally, significant alterations in the cell metabolism were detected for 25 metabolites, with levels of uracil, N-acetyl serine and propanoyl phosphate harboring the greatest alterations. Compared to the conventional therapy with cisplatin, VSM treated cells demonstrated a similar metabolic shutdown of the primary cell metabolism. Primary control cells were not affected by the VSM treatment. CONCLUSIONS: This study revealed the VSM root extract as a potential, new migrastatic drug candidate for the putative treatment of pediatric alveolar rhabdomyosarcoma with actin filament stabilizing properties and accompanied by a marginal effect on the vitality of primary cells.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Antineoplásicos/farmacología , Extractos Vegetales/farmacología , Rabdomiosarcoma/metabolismo , Vincetoxicum , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Niño , Humanos
4.
Mol Oncol ; 15(8): 2156-2171, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33523558

RESUMEN

Rhabdomyosarcoma (RMS) is an aggressive pediatric soft tissue sarcoma. There are two main subtypes of RMS, alveolar rhabdomyosarcoma (ARMS) and embryonal rhabdomyosarcoma. ARMS typically encompasses fusion-positive rhabdomyosarcoma, which expresses either PAX3-FOXO1 or PAX7-FOXO1 fusion proteins. There are no targeted therapies for ARMS; however, recent studies have begun to illustrate the cooperation between epigenetic proteins and the PAX3-FOXO1 fusion, indicating that epigenetic proteins may serve as targets in ARMS. Here, we investigate the contribution of BMI1, given the established role of this epigenetic regulator in sustaining aggression in cancer. We determined that BMI1 is expressed across ARMS tumors, patient-derived xenografts, and cell lines. We depleted BMI1 using RNAi and inhibitors (PTC-209 and PTC-028) and found that this leads to a decrease in cell growth/increase in apoptosis in vitro, and delays tumor growth in vivo. Our data suggest that BMI1 inhibition activates the Hippo pathway via phosphorylation of LATS1/2 and subsequent reduction in YAP levels and YAP/TAZ target genes. These results identify BMI1 as a potential therapeutic vulnerability in ARMS and warrant further investigation of BMI1 in ARMS and other sarcomas.


Asunto(s)
Proliferación Celular/fisiología , Epigénesis Genética/fisiología , Complejo Represivo Polycomb 1/fisiología , Rabdomiosarcoma/patología , Apoptosis/fisiología , Línea Celular Tumoral , Xenoinjertos , Vía de Señalización Hippo , Humanos , Fosforilación , Complejo Represivo Polycomb 1/genética , Interferencia de ARN , Rabdomiosarcoma/metabolismo
5.
Cancer Res ; 80(14): 3046-3056, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32354737

RESUMEN

Rhabdomyosarcoma is the most common childhood soft-tissue sarcoma, yet patients with metastatic or recurrent disease continue to do poorly, indicating a need for new treatments. The SRC family tyrosine kinase YES1 is upregulated in rhabdomyosarcoma and is necessary for growth, but clinical trials using single agent dasatinib, a SRC family kinase inhibitor, have failed in sarcomas. YAP1 (YES-associated protein) is highly expressed in rhabdomyosarcoma, driving growth and survival when the upstream Hippo tumor suppressor pathway is silenced, but efforts to pharmacologically inhibit YAP1 have been unsuccessful. Here we demonstrate that treatment of rhabdomyosarcoma with DNA methyltransferase inhibitor (DNMTi) upregulates Hippo activators RASSF1 and RASSF5 by promoter demethylation, activating canonical Hippo signaling and increasing inactivation of YAP1 by phosphorylation. Treatment with DNMTi decreased rhabdomyosarcoma cell growth and increased apoptosis and differentiation, an effect partially rescued by expression of constitutively active YAP (S127A), suggesting the effects of DNMTi treatment are, in part, due to Hippo-dependent inhibition of YAP1. In addition, YES1 and YAP1 interacted in the nucleus of rhabdomyosarcoma cells, and genetic or pharmacologic suppression of YES1 resulted in cytoplasmic retention of YAP1 and decreased YAP1 target gene expression, suggesting YES1 regulates YAP1 in a Hippo-independent manner. Combined treatment with DNMTi and dasatinib targeted both Hippo-dependent and Hippo-independent regulation of YAP1, ablating rhabdomyosarcoma cell growth in vitro and trending toward decreased tumor growth in vivo. These results show that the mechanisms regulating YAP1 in rhabdomyosarcoma can be inhibited by combinatorial therapy of DNMTi and dasatinib, laying the groundwork for future clinical investigations. SIGNIFICANCE: This study elucidates the signaling pathways that regulate the oncogenic protein YAP1 and identifies a combination therapy to target these pathways in the childhood tumor rhabdomyosarcoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Azacitidina/análogos & derivados , Terapia Molecular Dirigida , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Rabdomiosarcoma/tratamiento farmacológico , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis , Azacitidina/farmacología , Proliferación Celular , Niño , Femenino , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , Ratones , Ratones SCID , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
6.
Contrast Media Mol Imaging ; 2019: 2645928, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30956626

RESUMEN

In solid tumors, rapid local intravascular release of anticancer agents, e.g., doxorubicin (DOX), from thermosensitive liposomes (TSLs) can be an option to overcome poor extravasation of drug nanocarriers. The driving force of DOX penetration is the drug concentration gradient between the vascular compartment and the tumor interstitium. In this feasibility study, we used fibered confocal fluorescence microscopy (FCFM) to monitor in real-time DOX penetration in the interstitium of a subcutaneous tumor after its intravascular release from TSLs, Thermodox®. Cell uptake kinetics of the released DOX was quantified, along with an in-depth assessment of released-DOX penetration using an evolution model. A subcutaneous rat R1 rhabdomyosarcoma xenograft was used. The rodent was positioned in a setup including a water bath, and FCFM identification of functional vessels in the tumor tissue was applied based on AngioSense. The tumor-bearing leg was immersed in the 43°C water for preheating, and TSLs were injected intravenously. Real-time monitoring of intratumoral (i.t.) DOX penetration could be performed, and it showed the progressing DOX wave front via its native fluorescence, labeling successively all cell nuclei. Cell uptake rates (1/k) of 3 minutes were found (n=241 cells), and a released-DOX penetration in the range of 2500 µm2·s-1 was found in the tumor extravascular space. This study also showed that not all vessels, identified as functional based on AngioSense, gave rise to local DOX penetration.


Asunto(s)
Doxorrubicina/farmacocinética , Hipertermia Inducida , Liposomas/metabolismo , Animales , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Cinética , Microscopía Confocal , Ratas , Rabdomiosarcoma/metabolismo
7.
J Cancer Res Clin Oncol ; 144(9): 1717-1730, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29971532

RESUMEN

PURPOSE: Ferroptosis is a programmed form of iron-dependent cell death caused by lipid hydroperoxide accumulation, which can be prevented by glutathione peroxidase 4 (GPx4) activity. Here we investigated the effects of ferroptosis inducers called erastin and RSL3, which act by glutathione depletion and GPx4 inactivation, respectively, on muscle-derived cell lines of embryonal and alveolar rhabdomyosarcoma (RMS), and mouse normal skeletal C2C12 myoblasts. METHODS: Myogenic lines were exposed to stepwise increasing concentrations of ferroptosis inducers either alone or in combination with iron supplementation, iron chelating agents (bathophenanthrolinedisulfonic acid, BPS), antioxidant molecules (glutathione, N-acetylcysteine), lipid peroxidation inhibitors (ferrostatin-1), and chemotherapeutic agents (doxorubicin and actinomycin D). Drug susceptibility was quantified by measuring cell viability, proliferation and differentiation via neutral red assay, crystal violet assay and Giemsa staining, respectively. The detection of lipid hydroperoxide and protein levels was performed by immunofluorescence and Western blot analysis, respectively. RESULTS: Erastin and RSL3 increased lipid hydroperoxide levels preferentially in the embryonal U57810 and myoblast C2C12 lines, leading to ferroptosis that was accentuated by iron supplementation or prevented by co-treatment with BPS, glutathione, N-acetylcysteine and ferrostatin-1. The inhibition of extracellular regulated kinases (ERK) pathway prevented ferroptosis in U57810 and C2C12 cells, whereas its increased activation in the embryonal RD cells mediated by caveolin-1 (Cav-1) overexpression led to augmented ferroptosis susceptibility. Finally, we observed the combination of erastin or RSL3 with chemotherapeutic doxorubicin and actinomycin D agents to be effective in increasing cell death in all RMS lines. CONCLUSIONS: Erastin and RSL3 trigger ferroptosis in highly proliferating myogenic lines through a ERK pathway-dependent fashion.


Asunto(s)
Muerte Celular/fisiología , Proliferación Celular/fisiología , Mioblastos/patología , Rabdomiosarcoma/patología , Animales , Carbolinas/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclohexilaminas/farmacología , Dactinomicina/farmacología , Doxorrubicina/farmacología , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Ratones , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Fenilendiaminas/farmacología , Piperazinas/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo
8.
Mol Cancer Ther ; 17(7): 1365-1380, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29967215

RESUMEN

Targeted therapies have revolutionized cancer treatment; however, progress lags behind in alveolar (ARMS) and embryonal rhabdomyosarcoma (ERMS), a soft-tissue sarcoma mainly occurring at pediatric and young adult age. Insulin-like growth factor 1 receptor (IGF1R)-directed targeted therapy is one of the few single-agent treatments with clinical activity in these diseases. However, clinical effects only occur in a small subset of patients and are often of short duration due to treatment resistance. Rational selection of combination treatments of either multiple targeted therapies or targeted therapies with chemotherapy could hypothetically circumvent treatment resistance mechanisms and enhance clinical efficacy. Simultaneous targeting of distinct mechanisms might be of particular interest in this regard, as this affects multiple hallmarks of cancer at once. To determine the most promising and clinically relevant targeted therapy-based combination treatments for ARMS and ERMS, we provide an extensive overview of preclinical and (early) clinical data concerning a variety of targeted therapy-based combination treatments. We concentrated on the most common classes of targeted therapies investigated in rhabdomyosarcoma to date, including those directed against receptor tyrosine kinases and associated downstream signaling pathways, the Hedgehog signaling pathway, apoptosis pathway, DNA damage response, cell-cycle regulators, oncogenic fusion proteins, and epigenetic modifiers. Mol Cancer Ther; 17(7); 1365-80. ©2018 AACR.


Asunto(s)
Terapia Molecular Dirigida , Rabdomiosarcoma/terapia , Animales , Biomarcadores de Tumor , Ensayos Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Rabdomiosarcoma/etiología , Rabdomiosarcoma/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Proc Natl Acad Sci U S A ; 114(24): E4802-E4811, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28566498

RESUMEN

Several thermal-therapy strategies such as thermal ablation, hyperthermia-triggered drug delivery from temperature-sensitive liposomes (TSLs), and combinations of the above were investigated in a rhabdomyosarcoma rat tumor model (n = 113). Magnetic resonance-guided high-intensity focused ultrasound (MR-HIFU) was used as a noninvasive heating device with precise temperature control for image-guided drug delivery. For the latter, TSLs were prepared, coencapsulating doxorubicin (dox) and [Gd(HPDO3A)(H2O)], and injected in tumor-bearing rats before MR-HIFU treatment. Four treatment groups were defined: hyperthermia, ablation, hyperthermia followed by ablation, or no HIFU. The intratumoral TSL and dox distribution were analyzed by single-photon emission computed tomography (SPECT)/computed tomography (CT), autoradiography, and fluorescence microscopy. Dox biodistribution was quantified and compared with that of nonliposomal dox. Finally, the treatment efficacy of all heating strategies plus additional control groups (saline, free dox, and Caelyx) was assessed by tumor growth measurements. All HIFU heating strategies combined with TSLs resulted in cellular uptake of dox deep into the interstitial space and a significant increase of tumor drug concentrations compared with a treatment with free dox. Ablation after TSL injection showed [Gd(HPDO3A)(H2O)] and dox release along the tumor rim, mirroring the TSL distribution pattern. Hyperthermia either as standalone treatment or before ablation ensured homogeneous TSL, [Gd(HPDO3A)(H2O)], and dox delivery across the tumor. The combination of hyperthermia-triggered drug delivery followed by ablation showed the best therapeutic outcome compared with all other treatment groups due to direct induction of thermal necrosis in the tumor core and efficient drug delivery to the tumor rim.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Hipertermia Inducida/métodos , Imagen por Resonancia Magnética/métodos , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacocinética , Línea Celular Tumoral , Terapia Combinada , Doxorrubicina/administración & dosificación , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacocinética , Femenino , Radioisótopos de Indio , Liposomas , Polietilenglicoles/administración & dosificación , Polietilenglicoles/farmacocinética , Ratas , Rabdomiosarcoma/diagnóstico por imagen , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/terapia , Temperatura , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único
10.
Oncotarget ; 7(8): 8797-808, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26848777

RESUMEN

There is no effective drug to treat EV71 infection yet. Traditional Chinese herbs are great resources for novel antiviral compounds. Here we showed that Oblongifolin M (OM), an active compound isolated from Garcinia oblongifolia, potently inhibited EV71 infection in a dose dependent manner. To identify its potential effectors in the host cells, we successfully identified 18 proteins from 52 differentially expressed spots by comparative proteomics studies. Further studies showed that knockdown of ERp57 inhibited viral replication through downregulating viral IRES (internal ribosome entry site) activities, whereas ectopic expression of ERp57 increased IRES activity and partly rescued the inhibitory effects of OM on viral replication. We demonstrated that OM is an effective antiviral agent; and that ERp57 is one of its cellular effectors against EV71 infection.


Asunto(s)
Infecciones por Enterovirus/prevención & control , Garcinia/química , Extractos Vegetales/farmacología , Proteína Disulfuro Isomerasas/metabolismo , Rabdomiosarcoma/prevención & control , Terpenos/farmacología , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Western Blotting , Electroforesis en Gel Bidimensional , Enterovirus Humano A/patogenicidad , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Genoma Viral , Humanos , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética , Proteómica , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Rabdomiosarcoma/virología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Células Tumorales Cultivadas , Proteínas Virales/metabolismo
11.
Tumour Biol ; 37(1): 1025-33, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26269112

RESUMEN

Rhabdomyosarcoma (RMS) is a highly malignant and metastatic pediatric cancer that arises from the skeletal muscle. Recent studies have identified an important role of AKT signaling in RMS progression. In the current study, we investigated the activity of perifosine, an oral alkylphospholipid AKT inhibitor, against human RMS cells (RD and Rh-30 lines) both in vivo and in vitro, and studied the underlying mechanisms. We showed that perifosine significantly inhibited RMS cell growth in concentration- and time-dependent manners. Meanwhile, perifosine induced dramatic apoptosis in RMS cells. At the signaling level, perifosine blocked AKT activation, while inducing reactive oxygen species (ROS) production as well as JNK and P38 phosphorylations in RMS cells. Restoring AKT activation by introducing a constitutively active-AKT (CA-AKT) only alleviated (not abolished) perifosine-induced cytotoxicity in RD cells. Yet, the ROS scavenger N-acetyl cysteine (NAC) as well as pharmacological inhibitors against JNK (SP-600125) or P38 (SB-203580) suppressed perifosine-induced cytotoxicity in RMS cells. Thus, perifosine induces growth inhibition and apoptosis in RMS cells through mechanisms more than just blocking AKT. In vivo, oral administration of perifosine significantly inhibited growth of Rh-30 xenografts in severe combined immunodeficient (SCID) mice. Our data indicate that perifosine might be further investigated as a promising anti-RMS agent.


Asunto(s)
Antineoplásicos/farmacología , Fosforilcolina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones SCID , Fosforilcolina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Chemother Pharmacol ; 64(6): 1059-69, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19277661

RESUMEN

PURPOSE: Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children. Current chemotherapy regimes include the topoisomerase II poison etoposide and the transcription inhibitor actinomycin D. Poor clinical response necessitate identification of new agents to improve patient outcomes. METHODS: We assessed the in vitro cytotoxicity (MTT assay) of DNA intercalating agents in five established human RMS cell lines. These include novel classes of transcription inhibitors and topoisomerase poisons, previously shown to have potential as anti-cancer agents. RESULTS: Amongst the former agents, bisintercalating bis(9-aminoacridine-4-carboxamides) linked through the 9-position, and bis(phenazine-1-carboxamides) linked via their side chains, are compared with established transcription inhibitors. Amongst the latter, monofunctional acridine-4-carboxamides related to N-[2-(dimethylamino)ethyl]acridine-4-carboxamide, DACA, are compared with established topoisomerase poisons. CONCLUSIONS: Our findings specifically highlight the topoisomerase poison 9-amino-DACA, its 5-methylsulphone derivative, AS-DACA, and the bis(phenazine-1-carboxamide) transcription inhibitor MLN944/XR5944, currently in phase I trial, as candidates for further research into new agents for the treatment of RMS.


Asunto(s)
Evaluación Preclínica de Medicamentos , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Rabdomiosarcoma Alveolar/tratamiento farmacológico , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Rabdomiosarcoma/tratamiento farmacológico , Inhibidores de Topoisomerasa , Acridinas/farmacología , Aminoacridinas/farmacología , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Fenazinas/farmacología , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma Alveolar/enzimología , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/metabolismo , Rabdomiosarcoma Embrionario/enzimología , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/metabolismo
13.
Cancer Cell ; 14(5): 369-81, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18977326

RESUMEN

Studies support the importance of microRNAs in physiological and pathological processes. Here we describe the regulation and function of miR-29 in myogenesis and rhabdomyosarcoma (RMS). Results demonstrate that in myoblasts, miR-29 is repressed by NF-kappaB acting through YY1 and the Polycomb group. During myogenesis, NF-kappaB and YY1 downregulation causes derepression of miR-29, which in turn accelerates differentiation by targeting its repressor YY1. However, in RMS cells and primary tumors that possess impaired differentiation, miR-29 is epigenetically silenced by an activated NF-kappaB-YY1 pathway. Reconstitution of miR-29 in RMS in mice inhibits tumor growth and stimulates differentiation, suggesting that miR-29 acts as a tumor suppressor through its promyogenic function. Together, these results identify a NF-kappaB-YY1-miR-29 regulatory circuit whose disruption may contribute to RMS.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Desarrollo de Músculos/fisiología , Mioblastos Esqueléticos/citología , FN-kappa B/metabolismo , Rabdomiosarcoma/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Western Blotting , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Biología Computacional , Regulación hacia Abajo , Retroalimentación Fisiológica , Fibroblastos , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Mioblastos Esqueléticos/metabolismo , FN-kappa B/genética , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Rabdomiosarcoma/genética , Rabdomiosarcoma/prevención & control , Transducción de Señal , Factor de Transcripción YY1/genética
14.
Oncol Rep ; 20(2): 365-72, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18636199

RESUMEN

The efficacy of novel thermosensitive liposomes (40 degrees C) containing doxorubicin (Dox-Lip) together with local hyperthermia (HT) was studied on solid growing rat rhabdomyosarcomas. Tumor response and systemic toxicity were evaluated by comparing to free doxorubicin (Free Dox) with or without hyperthermia. Tumors were heated with infrared-A-radiation and drugs were infused intravenously after preheating the tumors followed by a further 60 min of heating at 42.5 degrees C. Recorded temperatures at various locations in the tumors indicated that all intratumoral temperatures, especially at the back rim, were definitely >40 degrees C. After single doses, tumor growth was further inhibited by Dox-Lip+HT compared to Free Dox+HT or Free Dox alone. Repeated treatments with Dox-Lip+HT (2x2.5 mg/kg+HT/2 weeks) resulted in a statistically significant tumor growth delay and was associated with a much lower systemic toxicity. Uptake studies of drugs in blood, tumor and normal tissues showed that Dox-liposomes (40 degrees C) are long circulating liposomes in the blood. However, the enhanced tumor response did not correlate with an increased uptake of Dox-Lip+HT in the tumor. The findings suggest that repeated applications of thermosensitive liposomal doxorubicin (40 degrees C) and local hyperthermia can control primary rat rhabdomyosarcomas while reducing the systemic toxicity of free doxorubicin.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Hipertermia Inducida , Rabdomiosarcoma/terapia , Neoplasias de los Tejidos Blandos/terapia , Animales , Peso Corporal/efectos de los fármacos , Terapia Combinada , Modelos Animales de Enfermedad , Portadores de Fármacos , Liposomas , Masculino , Ratas , Ratas Endogámicas , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología , Tasa de Supervivencia
15.
J Biomol Screen ; 12(5): 705-14, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17537987

RESUMEN

The no-wash calcium assay kits developed by Molecular Devices Corporation have greatly enhanced the throughput of cell-based calcium mobilization high-throughput screening (HTS) assays and enabled screening using nonadherent cells. The fluorescent imaging plate reader (FLIPR) Calcium 3 Assay Kit, optimal for targets that have proteins or peptides as agonists, has 2 potential drawbacks: 1) a significant downward spike in fluorescence signal upon liquid transfer that can be the same magnitude as the agonist response, making data analysis difficult; and 2) medium removal is required for some targets, which essentially reintroduces a wash step. Several no-wash products were introduced in 2005. The authors compare the Fluo-4 NW Calcium Assay Kit and the BD Calcium Assay Kit with the FLIPR Calcium 3 Assay Kit using human native rhabdomyosarcoma cells expressing the urotensin-II receptor (UT). The BDtrade mark Calcium Assay Kit gives the best performance in the true no-wash mode, in which both agonist and antagonist activity are easily quantified. Although these new products provide additional options for measuring calcium mobilization, the different results observed with each kit, using the UT receptor as an example, suggest that one should characterize all dyes against each target in a systematic way prior to choosing one for HTS.


Asunto(s)
Calcio/análisis , Juego de Reactivos para Diagnóstico , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Compuestos de Anilina , Calcio/metabolismo , Células Cultivadas , Células Clonales , Evaluación Preclínica de Medicamentos , Colorantes Fluorescentes , Fluorometría/instrumentación , Fluorometría/métodos , Humanos , Concentración 50 Inhibidora , Receptores Acoplados a Proteínas G/metabolismo , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Urotensinas/metabolismo , Urotensinas/farmacología , Xantenos
16.
Nucl Med Biol ; 33(3): 399-407, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16631089

RESUMEN

PURPOSE: An inverse correlation has been established between tumor levels of the DNA repair protein alkylguanine-DNA alkyltransferase (AGT) and a positive outcome after alkylator chemotherapy. Quantitative imaging of AGT could provide important information for patient-specific cancer treatment. Several radiolabeled analogues of O6-benzylguanine (BG), a potent AGT inactivator, have been developed and shown to be capable of labeling pure AGT protein. Herein, two of these analogues--O6-3-[*I]iodobenzylguanine ([*I]IBG) and O6-3-[*I]iodobenzyl-2'-deoxyguanosine ([*I]IBdG)--were further evaluated in two murine xenograft models. (AcO)2-[131I]IBdG, a peracetylated derivative of IBdG, also was investigated as an alternative agent. METHODS: Several biodistribution studies of radioiodinated IBG and IBdG were performed in TE-671 human rhabdomyosarcoma and DAOY human medulloblastoma murine xenograft models. Mice were treated with BG or its nucleoside analogue dBG to deplete the tumor AGT content. The effect of unlabeled IBG and that of 7,8-benzoflavone (BF), an inhibitor of the cytochrome P-450 isozyme CYP1A2, on the tumor uptake of the tracers was determined. The uptake of (AcO)2-[131I]IBdG along with that of [125I]IBdG in DAOY cells in vitro was determined in the presence and absence of a nucleoside transporter inhibitor, dipyridamole. RESULTS: Pretreatment of mice either with BG or dBG failed to reduce tumor levels of [*I]IBG or [*I]IBdG even though such treatments completely depleted tumor AGT content. Treatment of mice with BF increased tumor uptake of [125I]IBG by 56%; however, differentiation of tumors with and without AGT still was not possible. (AcO)2-[131I]IBdG, a peracetylated derivative of IBdG, had a higher uptake in vitro in DAOY tumor cells. However, its uptake, like that of [125I]IBdG, was blocked by dipyridamole. CONCLUSIONS: Taken together, these results suggest that labeled agents that are more specific for cellular AGT and that are more metabolically stable are needed.


Asunto(s)
Guanina/análogos & derivados , Radioisótopos de Yodo/farmacocinética , Meduloblastoma/diagnóstico por imagen , Rabdomiosarcoma/diagnóstico por imagen , Animales , Benzoflavonas/farmacología , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , Guanina/farmacocinética , Humanos , Masculino , Meduloblastoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Cintigrafía , Rabdomiosarcoma/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
17.
Int J Cancer ; 119(4): 757-64, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16550606

RESUMEN

Curcumin (diferuloylmethane), a polyphenol natural product of the plant Curcuma longa, is undergoing early clinical trials as a novel anticancer agent. However, the anticancer mechanism of curcumin remains to be elucidated. Here we show that curcumin inhibited growth of rhabdomyosarcoma cells (Rh1 and Rh30) (IC50 = 2-5 microM) and arrested cells in G1 phase of the cell cycle. Curcumin also induced apoptosis and inhibited the basal or type I insulin-like growth factor-induced motility of the cells. At physiological concentrations (2.5 microM), curcumin rapidly inhibited phosphorylation of the mammalian target of rapamycin (mTOR) and its downstream effector molecules, p70 S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E (eIF4E) binding protein 1 (4E-BP1), in a panel of cell lines (Rh1, Rh30, DU145, MCF-7 and Hela). Curcumin also inhibited phosphorylation of Akt in the cells, but only at high concentrations (>40 microM). The data suggest that curcumin may execute its anticancer activity primarily by blocking mTOR-mediated signaling pathways in the tumor cells.


Asunto(s)
Curcumina/farmacología , Proteínas Quinasas/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fase G1/efectos de los fármacos , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Fitoterapia , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Rabdomiosarcoma/patología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR , Proteína p53 Supresora de Tumor/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-16317957

RESUMEN

BACKGROUND: HBOC-201 is an ultra purified bovine hemoglobin solution. It has already been used in clinical phase II/III trials for emergency treatments. Animal experiments have shown that HBOC-201 is highly effective in tissue oxygenation. The study was performed in order to assess the potential of low dose HBOC-201 to improve tumor oxygenation. METHODS: 30 rats with a subcutaneously growing rhabdomyosarcoma R1H tumor were randomly assigned either to be ventilated with carbogen (n = 10), or to receive an IV injection of 0.3 g/kg HBOC-201 (n = 10) or a combination of 0.3 g/kg HBOC-201 and carbogen breathing (n = 10). Under general anesthesia the effects of the respective treatment on the tissue oxygen tension (tpO2) of the tumor were determined using a flexible stationary probe at baseline (b) and 15 and 60 min after application of the respective medication. RESULTS: HBOC-201 alone failed to improve tumor tpO2 (b: 1.3 +/- 1.2mmHg; 15min: 1.4 +/- 1 mmHg; 60min: 1 +/- 1 mmHg). In combination with carbogen the mean tpO2 of the tumor raised in comparison to baseline values (b: 3.1 +/- 4.6 mmHg; mmHg; 15min: 8.5 +/- 11*mmHg; 60min: 4.8 +/- 5mmHg; *p < 0.05 vs. b), but this effect was less pronounced than the increase in tpO2 by carbogen alone (b: 3.4 +/- 3.4mmHg; 15min: 9 +/- 10* mmHg; 60 min: 13 +/- 19* mmHg; *p < 0.05 vs. b). CONCLUSION: The application of low dose hemoglobin solution HBOC-201 does not result in improvement of tissue oxygenation in the rat rhabdomyosarcoma R1H.


Asunto(s)
Hemoglobinas/farmacología , Oxígeno/metabolismo , Rabdomiosarcoma/metabolismo , Animales , Sustitutos Sanguíneos/administración & dosificación , Sustitutos Sanguíneos/farmacología , Dióxido de Carbono/administración & dosificación , Dióxido de Carbono/farmacología , Evaluación Preclínica de Medicamentos , Hemoglobinas/administración & dosificación , Hipoxia/tratamiento farmacológico , Oxígeno/administración & dosificación , Oxígeno/farmacología , Ratas , Rabdomiosarcoma/patología , Distribución Tisular/efectos de los fármacos
19.
J Biol Chem ; 277(7): 5548-55, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11719506

RESUMEN

The zinc finger transcription factor GLI1, which mediates Sonic hedgehog signaling during development, is expressed in several human cancers, including basal cell carcinoma, medulloblastoma, and sarcomas. We identified 147 genes whose levels of expression were significantly altered in RNA obtained from cells demonstrating a transformed phenotype with stable GLI1 expression or stable Ha-ras expression. Comparison of expression profiles from GLI1- and Ha-ras-expressing cells established a set of genes unique to GLI1-induced cell transformation. Thirty genes were altered by stable GLI1 expression, and 124 genes were changed by stable Ha-ras expression. Seven genes had altered expression levels in both GLI1- and Ha-ras-expressing cells. Genes whose expression was altered by GLI1 included cell cycle genes, cell adhesion genes, signal transduction genes, and genes regulating apoptosis. GLI1 consensus DNA-binding sequences were identified in the 5' regions of cyclin D2, IGFBP-6, osteopontin, and plakoglobin, suggesting that these genes represent immediate downstream targets. Gel shift analysis confirmed the ability of the GLI1 protein to bind these sequences. Up-regulation of cyclin D2 and down-regulation of plakoglobin were demonstrated in GLI1-amplified compared with non-amplified human rhabdomyosarcoma cells. Many of the GLI1 targets with known function identified in this study increase cell proliferation, indicating that GLI1-induced cell transformation occurs through multiple downstream pathways.


Asunto(s)
Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Secuencias de Aminoácidos , Animales , Apoptosis , Secuencia de Bases , Biotinilación , Northern Blotting , Adhesión Celular , Línea Celular Transformada , Ciclina D2 , Ciclinas/biosíntesis , Proteínas del Citoesqueleto/biosíntesis , ADN Complementario/metabolismo , Desmoplaquinas , Regulación hacia Abajo , Humanos , Hibridación in Situ , Proteína 6 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Modelos Biológicos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteopontina , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , ARN/metabolismo , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Rabdomiosarcoma/metabolismo , Sialoglicoproteínas/biosíntesis , Transducción de Señal , Transactivadores , Células Tumorales Cultivadas , Regulación hacia Arriba , Proteína con Dedos de Zinc GLI1 , gamma Catenina , Proteínas ras/biosíntesis
20.
Int J Radiat Oncol Biol Phys ; 50(3): 783-92, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11395248

RESUMEN

PURPOSE: To establish dose-effect relationships for tumor acidification induced by heat and glucose as a basis for testing the value of adding glucose administration to combined heat and x-ray treatment at clinically achievable glucose and temperature levels. METHODS AND MATERIALS: Rhabdomyosarcoma BA1112 was grown s.c. in the upper leg of 16-20-week-old Wag/Rij rats. Animals were given 2 consecutive 100-min periods of saline (S) or glucose (G) infusion, while keeping tumor temperature at 37 degrees, 42 degrees, or 43 degrees C for 1 or 2 periods, in various combinations, each involving 6 animals. Glucose was infused i.v. as a 20% solution at 2.4-3 g/kg/h. Tumors were heated using 2,450-MHz electromagnetic radiation, and tumor pH was measured using a 0.7 mm fiberoptic probe. RESULTS: Mean overall baseline pH was 7.00 (SD 0.10). The change induced by G37G43 (i.e., glucose infusion for a full 200 min, first 100 min at 37 degrees C, final 100 min at 43 degrees C) was -0.48 +/- 0.03 (SEM) pH units, and -0.17 +/- 0.03 for S37S43. The effect of G37G42 was -0.37 +/- 0.03 pH units, compared with -0.08 +/- 0.02 for S37S42 and -0.28 +/- 0.04 for glucose alone (G37G37). Glucose was less effective when given after or fully parallel to heating: -0.21 +/- 0.02 pH units for S43G37 and -0.37 +/- 0.02 for G43G43. CONCLUSION: The glucose-induced tumor pH drop is much more pronounced than that induced by heat, both of which are dose dependent. The effects of glucose and heat seem additive if heating is started when glucose-induced acidification has reached its plateau level, but the overall effect is diminished if administration is fully simultaneous or in reversed order. Schedule G37G43 is optimal with respect to tumor acidification. Its predicted superiority in thermoradiotherapy as compared with S37S42, S37S43, and G37G42 treatment regimens was confirmed in a subsequent experimental tumor control study.


Asunto(s)
Glucemia/metabolismo , Glucosa/farmacología , Hipertermia Inducida , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/terapia , Animales , Terapia Combinada , Relación Dosis-Respuesta a Droga , Femenino , Concentración de Iones de Hidrógeno , Ratas , Rabdomiosarcoma/radioterapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA