Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 8(13)2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37279073

RESUMO

Osteosarcoma (OS) is the most common primary bone tumor of childhood. Approximately 20%-30% of OSs carry amplification of chromosome 8q24, which harbors the oncogene c-MYC and correlates with a poor prognosis. To understand the mechanisms that underlie the ability of MYC to alter both the tumor and its surrounding tumor microenvironment (TME), we generated and molecularly characterized an osteoblast-specific Cre-Lox-Stop-Lox-c-MycT58A p53fl/+ knockin genetically engineered mouse model (GEMM). Phenotypically, the Myc-knockin GEMM had rapid tumor development with a high incidence of metastasis. MYC-dependent gene signatures in our murine model demonstrated significant homology to the human hyperactivated MYC OS. We established that hyperactivation of MYC led to an immune-depleted TME in OS demonstrated by the reduced number of leukocytes, particularly macrophages. MYC hyperactivation led to the downregulation of macrophage colony-stimulating factor 1, through increased microRNA 17/20a expression, causing a reduction of macrophage population in the TME of OS. Furthermore, we developed cell lines from the GEMM tumors, including a degradation tag-MYC model system, which validated our MYC-dependent findings both in vitro and in vivo. Our studies utilized innovative and clinically relevant models to identify a potentially novel molecular mechanism through which MYC regulates the profile and function of the OS immune landscape.


Assuntos
Neoplasias Ósseas , MicroRNAs , Osteossarcoma , Humanos , Camundongos , Animais , Macrófagos Associados a Tumor/patologia , Fator Estimulador de Colônias de Macrófagos/genética , Osteossarcoma/genética , Osteossarcoma/patologia , Neoplasias Ósseas/patologia , MicroRNAs/genética , Microambiente Tumoral/genética
2.
Curr Protoc ; 3(2): e670, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36799651

RESUMO

Ewing Sarcoma (EwS) is the second most common malignant bone tumor in adolescents and young adults. The single-most powerful predictor of outcome in EwS is presence of metastatic burden at the time of diagnosis. Patients with metastatic Ewing Sarcoma have an abysmal 5-year survival rate of 10%-25%, which has not changed over the past 30-40 years. Thus, unraveling underlying mechanisms of EwS metastasis are imperative for developing effective therapeutic measures. Investigations towards this goal are limited by the lack of reliable genetically engineered mouse models and specialized metastatic models. Using two established cell lines, A673 and TC71, we generated lung specific metastatic cell lines by serial orthotopic intra-tibial injection followed by isolation of cells from lung metastases. The lung metastatic lines generated exhibit distinct differential molecular signatures from the parental cells when analyzed using a multi-omics approach. These signatures overlapped with EwS patient primary bone and metastatic lung specimens supporting the clinical relevance of these preclinical models of EwS. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Intra-Tibial injection in NSG mice Basic Protocol 2: Development and characterization of lung metastatic cell line.


Assuntos
Neoplasias Ósseas , Neoplasias Pulmonares , Tumores Neuroectodérmicos Primitivos Periféricos , Sarcoma de Ewing , Animais , Camundongos , Sarcoma de Ewing/tratamento farmacológico , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Pulmonares/secundário
3.
Cancer Res ; 81(1): 199-212, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33168646

RESUMO

Rhabdomyosarcoma (RMS) is the most prevalent pediatric soft-tissue sarcoma. Multimodal treatment, including surgery and traditional chemotherapy with radiotherapy, has contributed to improvements in overall survival rates. However, patients with recurrent or metastatic disease have 5-year survival rates of less than 30%. One reason for the lack of therapeutic advancement is identification and targeting of critical signaling nodes. p21-activated kinases (PAK) are a family of serine/threonine kinases downstream of multiple critical tumorigenic receptor tyrosine kinase receptors and oncogenic regulators, including IGFR and RAS signaling, that significantly contribute to aggressive malignant phenotypes. Here, we report that RMS cell lines and tumors exhibit enhanced PAK4 expression levels and activity, which are further activated by growth factors involved in RMS development. Molecular perturbation of PAK4 in multiple RMS models in vitro and in vivo resulted in inhibition of RMS development and progression. Fusion-positive and -negative RMS models were sensitive to two PAK4 small-molecule inhibitors, PF-3758309 and KPT-9274, which elicited significant antitumor and antimetastatic potential in several primary and metastatic in vivo models, including a relapsed RMS patient-derived xenograft model. Transcriptomic analysis of PAK4-targeted tumors revealed inhibition of the RAS-GTPase, Hedgehog, and Notch pathways, along with evidence of activation of antitumor immune response signatures. This PAK4-targeting gene signature showed prognostic significance for patients with sarcoma. Overall, our results show for the first time that PAK4 is a novel and viable therapeutic target for the treatment of high-risk RMS. SIGNIFICANCE: These data demonstrate a novel oncogenic role for PAK4 in rhabdomyosarcoma and show that targeting PAK4 activity is a promising viable therapeutic option for advanced rhabdomyosarcoma.


Assuntos
Acrilamidas/farmacologia , Aminopiridinas/farmacologia , Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Pirazóis/farmacologia , Pirróis/farmacologia , Rabdomiossarcoma/patologia , Quinases Ativadas por p21/antagonistas & inibidores , Proteínas ras/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Criança , Humanos , Masculino , Camundongos , Rabdomiossarcoma/genética , Rabdomiossarcoma/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Proteínas ras/genética
4.
Int J Radiat Oncol Biol Phys ; 101(1): 118-127, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29534895

RESUMO

PURPOSE: Radiation therapy (RT) is a viable therapeutic option for Ewing sarcoma (ES) patients. However, little progress has been made to elucidate the mechanisms of radioresistance. This study establishes a novel ES irradiation-adapted model designed to assess molecular and 18F fluorodeoxyglucose (FDG) positron emission tomography (PET) alterations secondary to RT. METHODS AND MATERIALS: Radiation-adapted cell lines (RACLs) were created in vitro by exposing ES human cell lines to fractionated doses of radiation. Assays to assess migration or invasion potential and RNA expression were performed on the RACLs. Orthotopic intratibial in vivo investigations were performed with irradiation-sensitive and irradiation-adapted ES cells to generate tumors. Transplanted mice were imaged using 18F-FDG PET followed by fractionated RT directed at the primary tumor. Mice were monitored for tumor regression and change in metabolic activity using 18F-FDG PET imaging. Protein expression analyses were performed on the RACLs and orthotopic tumors. RESULTS: Exposure to fractionated doses of radiation caused a significant increase in migratory and invasive properties in the RACLs when compared with nonirradiated wild-type ES cells. RACL transcriptomic and proteomic analysis suggests enhanced activation of the mammalian target of rapamycin-AKT pathway when compared with wild-type ES cells. Irradiation-adapted tumors demonstrated significantly less tumor regression (P = .03) than wild-type tumors. Wild-type tumors also had decreased expression of lactate dehydrogenase A protein and significantly lower metabolic activity after RT compared with irradiation-adapted tumors (P = .03). CONCLUSIONS: We developed novel in vitro and in vivo irradiation-adapted ES models. In vitro investigations revealed increased migratory and invasive phenotypes in the RACLs. In vivo investigations demonstrated increased metabolic activity and significantly decreased sensitivity to RT in the irradiation-adapted tumors as demonstrated by growth response curves and 18F-FDG PET activity. Investigations of the RACLs identified possible radiosensitizing-dependent targets in lactate dehydrogenase A and the mammalian target of rapamycin-AKT pathway.


Assuntos
Neoplasias Ósseas/radioterapia , Tolerância a Radiação , Sarcoma de Ewing/radioterapia , Adaptação Fisiológica , Animais , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos da radiação , Fluordesoxiglucose F18 , Humanos , L-Lactato Desidrogenase/metabolismo , Camundongos , Invasividade Neoplásica , Tomografia por Emissão de Pósitrons , Proteínas Proto-Oncogênicas c-akt/metabolismo , Hipofracionamento da Dose de Radiação , Compostos Radiofarmacêuticos , Sarcoma de Ewing/diagnóstico por imagem , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Serina-Treonina Quinases TOR/metabolismo
5.
Oncotarget ; 8(44): 77292-77308, 2017 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-29100387

RESUMO

Ewing sarcoma (EWS) is a highly aggressive and metabolically active malignant tumor. Metabolic activity can broadly be characterized by features of glycolytic activity and oxidative phosphorylation. We have further characterized metabolic features of EWS cells to identify potential therapeutic targets. EWS cells had significantly more glycolytic activity compared to their non-malignant counterparts. Thus, metabolic inhibitors of glycolysis such as 2-deoxy-D-glucose (2DG) and of the mitochondrial respiratory pathway, such as metformin, were evaluated as potential therapeutic agents against a panel of EWS cell lines in vitro. Results indicate that 2DG alone or in combination with metformin was effective at inducing cell death in EWS cell lines. The predominant mechanism of cell death appears to be through stimulating apoptosis leading into necrosis with concomitant activation of AMPK-α. Furthermore, we demonstrate that the use of metabolic modulators can target putative EWS stem cells, both in vitro and in vivo, and potentially overcome chemotherapeutic resistance in EWS. Based on these data, clinical strategies using drugs targeting tumor cell metabolism present a viable therapeutic modality against EWS.

6.
Int J Mol Sci ; 18(1)2016 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-28025559

RESUMO

Apoptosis, and the more recently discovered necroptosis, are two avenues of programmed cell death. Cancer cells survive by evading these two programs, driven by oncogenes and tumor suppressor genes. While traditional therapy using small molecular inhibitors and chemotherapy are continuously being utilized, a new and exciting approach is actively underway by identifying and using synergistic relationship between driver and rescue genes in a cancer cell. Through these synthetic lethal relationships, we are gaining tremendous insights into tumor vulnerabilities and specific molecular avenues for induction of programmed cell death. In this review, we briefly discuss the two cell death processes and cite examples of such synergistic manipulations for therapeutic purposes.


Assuntos
Apoptose , Neoplasias/metabolismo , Animais , Antineoplásicos/classificação , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Necrose , Neoplasias/tratamento farmacológico , Neoplasias/genética
7.
Breast Cancer Res ; 17: 88, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26111812

RESUMO

Metformin, a diabetes drug with well-established side effect and safety profiles, has been widely studied for its anti-tumor activities in a number of cancers, including breast cancer. But its mechanism of action in the clinical arena remains elusive. In a window of opportunity trial of metformin in non-diabetic breast cancer patients, Dowling and colleagues examined both the direct actions of the drug on cancer cells (as mediated by AMP kinase), as well as its indirect actions (as mediated by circulating insulin). The data suggest that short-term administration of metformin in this setting has anti-tumor effects significantly involving the indirect, insulin-dependent pathway. The role of the direct pathway remains to be determined. This study represents an important step forward in establishing one of several possible mechanisms for metformin, information that will be useful in determining candidate biomarkers to evaluate in large clinical trials of metformin, such as the ongoing NCIC CTG MA.32 trial of adjuvant metformin. The potential significance of these data for metformin in the treatment of breast cancer is discussed here.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Metformina/farmacologia , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Feminino , Humanos
8.
J Neurooncol ; 107(3): 487-501, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22249692

RESUMO

Glioma stem-like cells (GSCs) may be the initiating cells in glioblastoma (GBM) and contribute to the resistance of these tumors to conventional therapies. Development of novel chemotherapeutic agents and treatment approaches against GBM, especially those specifically targeting GSCs are thus necessary. In the present study, we found that a novel Janus kinase 2/Signal Transducer and Activator of Transcription 3 (JAK2/STAT3) pathway inhibitor (WP1193) significantly decreased the proliferation of established glioma cell lines in vitro and inhibit the growth of glioma in vivo. To test the efficacy of WP1193 against GSCs, we then administrated WP1193 to GSCs isolated and expanded from multiple human GBM tumors. We revealed that WP1193 suppressed phosphorylation of JAK2 and STAT3 with high potency and demonstrated a dose-dependent inhibition of proliferation and neurosphere formation of GSCs. These effects were at least due in part to G1 arrest associated with down-regulation of cyclin D1 and up-regulation of p21( Cip1/Waf-1 ). Furthermore, WP1193 exposure decreased expression of stem cell markers including CD133 and c-myc, and induced cell death in GSCs through apoptosis. Taken together, our data indicate that WP1193 is a potent small molecule inhibitor of the JAK2/STAT3 pathway that shows promise as a therapeutic agent against GBM by targeting GSCs.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Cianoacrilatos/farmacologia , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Piridinas/farmacologia , Animais , Western Blotting , Citometria de Fluxo , Glioblastoma/metabolismo , Humanos , Janus Quinase 2/metabolismo , Masculino , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Eur J Cancer ; 45(4): 677-84, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19121577

RESUMO

Previously we reported that Stat3 is persistently activated in GBM tumours and derived cell lines. Hypoxia, necrosis and neo-angiogenesis are hallmarks of GBM. To unfold the contribution of activated Stat3 to the growth of GBM, we generated human GBM cell line (U87)-derived stable clones expressing a dominant negative mutant (DN)-Stat3 in a hypoxia-inducible manner, and examined their tumour-forming potentials in immune-compromised mice. We found that the parental and vector control cell-derived tumours grew steadily, whereas DN-Stat3-expressing clone-derived tumours failed to grow beyond 2mm of thickness in mouse flanks. This blockade of tumour growth was associated with induction of tumour cell apoptosis and suppression of tumour angiogenesis. Consistent with this, mice bearing orthotopically implanted DN-Stat3-expressing clones survived significantly longer than the control mice. These data suggest that activated Stat3 is required for the growth of GBM, and that targeting Stat3 may intervene with the growth of GBM.


Assuntos
Glioblastoma/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose , Hipóxia Celular , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Vetores Genéticos , Glioblastoma/irrigação sanguínea , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Hospedeiro Imunocomprometido , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Transplante de Neoplasias , Neovascularização Patológica/prevenção & controle , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/fisiologia , Análise de Sobrevida , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...