Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci ; 295: 120394, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35157910

RESUMO

AIMS: Medulloblastoma (MB) is one of the most common malignant central nervous system tumors of childhood. Despite intensive treatments that often leads to severe neurological sequelae, the risk for resistant relapses remains significant. In this study we have evaluated the effects of the ω3-long chain polyunsaturated fatty acids (ω3-LCPUFA) docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on MB cell lines and in a MB xenograft model. MAIN METHODS: Effects of ω3-LCPUFA treatment of MB cells were assessed using the following: WST-1 assay, cell death probes, clonogenic assay, ELISA and western blot. MB cells were implanted into nude mice and the mice were randomized to DHA, or a combination of DHA and EPA treatment, or to control group. Treatment effects in tumor tissues were evaluated with: LC-MS/MS, RNA-sequencing and immunohistochemistry, and tumors, erythrocytes and brain tissues were analyzed with gas chromatography. KEY FINDINGS: ω3-LCPUFA decreased prostaglandin E2 (PGE2) secretion from MB cells, and impaired MB cell viability and colony forming ability and increased apoptosis in a dose-dependent manner. DHA reduced tumor growth in vivo, and both PGE2 and prostacyclin were significantly decreased in tumor tissue from treated mice compared to control animals. All ω3-LCPUFA and dihomo-γ-linolenic acid increased in tumors from treated mice. RNA-sequencing revealed 10 downregulated genes in common among ω3-LCPUFA treated tumors. CRYAB was the most significantly altered gene and the downregulation was confirmed by immunohistochemistry. SIGNIFICANCE: Our findings suggest that addition of DHA and EPA to the standard MB treatment regimen might be a novel approach to target inflammation in the tumor microenvironment.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Meduloblastoma/tratamento farmacológico , Meduloblastoma/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinogênese , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida/métodos , Dinoprostona/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Ácidos Graxos/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Prostaglandinas/metabolismo , Espectrometria de Massas em Tandem/métodos , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Cadeia B de alfa-Cristalina/efeitos dos fármacos , Cadeia B de alfa-Cristalina/metabolismo
2.
Acta Anaesthesiol Scand ; 65(3): 360-363, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33165936

RESUMO

BACKGROUND: The management of COVID-19 ARDS is debated. Although current evidence does not suggest an atypical acute respiratory distress syndrome (ARDS), the physiological response to prone positioning is not fully understood and it is unclear which patients benefit. We aimed to determine whether proning increases oxygenation and to evaluate responders. METHODS: This case series from a single, tertiary university hospital includes all mechanically ventilated patients with COVID-19 and proning between 17 March 2020 and 19 May 2020. The primary measure was change in PaO2 :FiO2 . RESULTS: Forty-four patients, 32 males/12 females, were treated with proning for a total of 138 sessions, with median (range) two (1-8) sessions. Median (IQR) time for the five sessions was 14 (12-17) hours. In the first session, median (IQR) PaO2 :FiO2 increased from 104 (86-122) to 161 (127-207) mm Hg (P < .001). 36/44 patients (82%) improved in PaO2 :FiO2 , with a significant increase in PaO2 :FiO2 in the first three sessions. Median (IQR) FiO2 decreased from 0.7 (0.6-0.8) to 0.5 (0.35-0.6) (<0.001). A significant decrease occurred in the first three sessions. PaO2 , tidal volumes, PEEP, mean arterial pressure (MAP), and norepinephrine infusion did not differ. Primarily, patients with PaO2 :FiO2 approximately < 120 mm Hg before treatment responded to proning. Age, sex, BMI, or SAPS 3 did not predict success in increasing PaO2 :FiO2 . CONCLUSION: Proning increased PaO2 :FiO2 , primarily in patients with PaO2 :FiO2 approximately < 120 mm Hg, with a consistency over three sessions. No characteristic was associated with non-responding, why proning may be considered in most patients. Further study is required to evaluate mortality.


Assuntos
COVID-19/complicações , COVID-19/terapia , Posicionamento do Paciente/métodos , Respiração Artificial/métodos , Síndrome do Desconforto Respiratório/complicações , Síndrome do Desconforto Respiratório/terapia , Idoso , COVID-19/fisiopatologia , Feminino , Humanos , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Decúbito Ventral , Estudos Prospectivos , Síndrome do Desconforto Respiratório/fisiopatologia , SARS-CoV-2 , Volume de Ventilação Pulmonar/fisiologia , Resultado do Tratamento
3.
Int J Cancer ; 128(7): 1703-11, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20499314

RESUMO

Epidemiological and preclinical studies have revealed that omega-3 fatty acids have anticancer properties. We have previously shown that the omega-3 fatty acid docosahexaenoic acid (DHA) induces apoptosis of neuroblastoma cells in vitro by mechanisms involving intracellular peroxidation of DHA by means of 15-lipoxygenase or autoxidation. In our study, the effects of DHA supplementation on neuroblastoma tumor growth in vivo were investigated using two complementary approaches. For the purpose of prevention, DHA as a dietary supplement was fed to athymic rats before the rats were xenografted with human neuroblastoma cells. For therapeutic purposes, athymic rats with established neuroblastoma xenografts were given DHA daily by gavage and tumor growth was monitored. DHA levels in plasma and tumor tissue were analyzed by gas liquid chromatography. DHA delayed neuroblastoma xenograft development and inhibited the growth of established neuroblastoma xenografts in athymic rats. A revised version of the Pediatric Preclinical Testing Program evaluation scheme used as a measurement of treatment response showed that untreated control animals developed progressive disease, whereas treatment with DHA resulted in stable disease or partial response, depending on the DHA concentration. In conclusion, prophylactic treatment with DHA delayed neuroblastoma development, suggesting that DHA could be a potential agent in the treatment of minimal residual disease and should be considered for prevention in selected cases. Treatment results on established aggressive neuroblastoma tumors suggest further studies aiming at a clinical application in children with high-risk neuroblastoma.


Assuntos
Ácidos Graxos Ômega-3/metabolismo , Neuroblastoma/metabolismo , Administração Oral , Animais , Linhagem Celular Tumoral , Cromatografia Gasosa , Suplementos Nutricionais , Progressão da Doença , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Cromatografia Gasosa-Espectrometria de Massas/métodos , Humanos , Cinética , Transplante de Neoplasias , Ratos , Ratos Nus
4.
Exp Cell Res ; 316(8): 1365-73, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20211172

RESUMO

Omega-3 fatty acids have been implicated in cancer prevention and treatment. Conventional chemotherapeutics are considered "double-edged swords", as they kill the cancer cells but also strike the healthy cells causing severe morbidity and sometimes also mortality. Could omega-3 fatty acids in this setting work as a "sword and shield" instead, by being cytotoxic to cancer cells, but at the same time protect healthy cells from these deleterious effects? In addition, may our current diet with decreased omega-3/omega-6 ratio contribute to the increased cancer incidence, and could an omega-3 enriched diet be used as a preventive measure against cancer? Here, we seek answers to these questions by reviewing the effects of omega-3 fatty acids, particularly DHA, on various cancers with emphasis on a cancer of neural origin, neuroblastoma. Results from preventive and therapeutic animal as well as human studies together with mechanisms behind the observed toxicity are summarized.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Neoplasias/prevenção & controle , Animais , Humanos
5.
FASEB J ; 24(3): 906-15, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19890019

RESUMO

Docosahexaenoic acid (DHA) protects neural cells from stress-induced apoptosis. On the contrary, DHA exerts anticancer effects, and we have shown that DHA induces apoptosis in neuroblastoma, an embryonal tumor of the sympathetic nervous system. We now investigate the DHA metabolome in neuroblastoma using a targeted lipidomic approach in order to elucidate the mechanisms behind the DHA-induced cytotoxicity. LC-MS/MS analysis was used to identify DHA-derived lipid mediators in neuroblastoma cells. Presence of the 15-lipoxygenase enzyme was investigated using immunoblotting, and cytotoxic potency of DHA and DHA-derived compounds was compared using the MTT cell viability assay. Neuroblastoma cells metabolized DHA to 17-hydroxydocosahexaenoic acid (17-HDHA) via 17-hydroperoxydocosahexaenoic acid (17-HpDHA) through 15-lipoxygenase and autoxidation. In contrast to normal neural cells, neuroblastoma cells did not produce the anti-inflammatory and protective lipid mediators, resolvins and protectins. 17-HpDHA had significant cytotoxic potency, with an IC(50) of 3-6 microM at 72 h, compared to 12-15 microM for DHA. alpha-Tocopherol protected cells from 17-HpDHA-induced cytotoxicity. DHA inhibited secretion of prostaglandin-E(2) and augmented the cytotoxic potency of the cyclooxygenase-2-inhibitor celecoxib. The cytotoxic effect of DHA in neuroblastoma is mediated through production of hydroperoxy fatty acids that accumulate to toxic intracellular levels with restricted production of its products, resolvins and protectins.-Gleissman, H., Yang, R., Martinod, K., Lindskog, M., Serhan, C. N., Johnsen, J. I., Kogner, P. Docosahexaenoic acid metabolome in neural tumors: identification of cytotoxic intermediates.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Neuroblastoma/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida , Ácidos Docosa-Hexaenoicos/toxicidade , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Citometria de Fluxo , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Immunoblotting , Inibidores de Lipoxigenase , Extração em Fase Sólida , Espectrometria de Massas em Tandem
6.
Int J Cancer ; 124(5): 1227-34, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19058199

RESUMO

Imatinib is currently in early clinical trials as targeted therapy for relapsed neuroblastomas and other childhood solid tumors expressing platelet-derived growth factor receptors (PDGFR) or c-Kit. Short-term treatment with imatinib in clinically achievable concentrations is ineffective in neuroblastoma in vitro. However, clinically, imatinib is administered daily over long time periods. The effects of combining imatinib with chemotherapy in neuroblastoma are unknown. Here, a panel of neuroblastoma cell lines (n = 5) were studied, representing tumors with different biological (MYCN-amplification +/-) and clinical (drug resistance) features. Using a protracted low-dose treatment schedule (1-3 weeks; 0.5-5microM) imatinib dose-dependently inhibited proliferation and clonogenic survival for all tested cell lines with IC50 <2.5microM. In contrast, short-term treatment (<96 hrs) was ineffective. Low-dose imatinib was synergistic in combination with doxorubicin and caused increased G2/M- and S-phase arrest and apoptosis as evidenced by enhanced caspase-3 activation and sub-G1 DNA accumulation. A significant but less pronounced effect was observed when imatinib was combined with etoposide or vincristine, as opposed to cisplatin, melphalan, or irinotecan. All cell lines expressed PDGFRbeta, whereas no protein expression of PDGFRalpha was detected in MYCN amplified cell lines. PDGF-BB caused PDGFRbeta phosphorylation and partially rescued neuroblastoma cells from doxorubicin-induced apoptosis, in an imatinib-sensitive manner. In vivo, treatment with imatinib in combination with doxorubicin induced a significant growth inhibition of established neuroblastoma xenografts. These findings suggest clinical testing of imatinib in combination with selected chemotherapeutic drugs, in particular doxorubicin, in children with high-risk neuroblastoma.


Assuntos
Antineoplásicos/administração & dosagem , Neuroblastoma/tratamento farmacológico , Piperazinas/administração & dosagem , Pirimidinas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Becaplermina , Benzamidas , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Esquema de Medicação , Humanos , Mesilato de Imatinib , Masculino , Neuroblastoma/patologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia
7.
Clin Cancer Res ; 13(3): 1036-44, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17289900

RESUMO

PURPOSE: Neuroblastoma is the most common and deadly solid tumor of childhood. Cyclooxygenase-2 is expressed in clinical neuroblastoma tumors and cell lines and inhibitors of this enzyme induce apoptosis in human neuroblastoma cells in vitro and in neuroblastoma xenografts in vivo. We hypothesized that the cyclooxygenase-2-specific inhibitor celecoxib could enhance the cytotoxic effect of chemotherapeutic drugs currently used in neuroblastoma treatment. Furthermore, we investigated if prophylactic treatment with celecoxib could prevent neuroblastoma tumor development in vivo. EXPERIMENTAL DESIGN: Neuroblastoma cell cytotoxicity of chemotherapeutic drugs in combination with celecoxib was examined. In vivo, athymic rats carrying established SH-SY5Y xenografts were treated with celecoxib in combination with irinotecan, doxorubicin or etoposide, or with either drug alone. For prevention studies, rats received celecoxib in the diet, 250 to 2,500 ppm, from the time of tumor cell injection. RESULTS: Celecoxib induced a synergistic or an additive cytotoxic effect in combination with doxorubicin, etoposide, irinotecan or vincristine in vitro. In vivo, treatment with celecoxib in combination with irinotecan or doxorubicin induced a significant growth inhibition of established neuroblastoma tumors. Rats receiving celecoxib in the diet showed a distinct dose-dependent delay in tumor development compared with untreated rats. Plasma levels of celecoxib were comparable with levels obtainable in humans. CONCLUSIONS: Celecoxib potentiates the antitumor effect of chemotherapeutic drugs currently used in neuroblastoma treatment, which argues for clinical trials combining these drugs. Celecoxib could also be a potential drug for treatment of minimal residual disease.


Assuntos
Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclo-Oxigenase 2/biossíntese , Neuroblastoma/tratamento farmacológico , Pirazóis/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Apoptose , Celecoxib , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase/farmacologia , Humanos , Técnicas In Vitro , Antígeno Ki-67/biossíntese , Masculino , Transplante de Neoplasias , Neuroblastoma/metabolismo , Ratos , Ratos Nus
8.
Int J Cancer ; 118(10): 2584-93, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16353135

RESUMO

Docosahexaenoic acid (DHA) is an omega-3 polyunsaturated fatty acid vital for the developing nervous system and significantly decreased in neuroblastoma cells compared to nontransformed nervous tissue. We investigated whether supplementation of DHA affects the susceptibility of neuroblastoma cells to oxidative stress generated endogenously and in response to cytotoxic therapy. DHA, but not the monounsaturated oleic acid (OA), induced dose- and time-dependent neuroblastoma cell death. DHA supplementation was associated with depolarization of the mitochondrial membrane potential, production of reactive oxygen species (ROS) and accumulation of DNA in sub-G1 phase of the cell cycle. The antioxidant, vitamin E, inhibited mitochondrial depolarization and subsequent cell death induced by DHA, whereas, the mitochondrial pore inhibitor, cyclosporin A, partly inhibited DHA-induced neuroblastoma cell death. Depletion of glutathione by L-buthionine-sulfoximine significantly enhanced the cytotoxic effects of DHA. Nontransformed fibroblasts were not substantially affected by DHA. DHA, but not OA, significantly enhanced the cytotoxicity of cisplatin, doxorubicin and irinotecan both in chemosensitive and in multidrug-resistant neuroblastoma cells. DHA potently sensitized neuroblastoma cells to a clinically relevant concentration (1 microM) of arsenic trioxide (As2O3) and enhanced the effect of the nonsteroidal antiinflammatory drug (NSAID), diclofenac. These findings provide experimental evidence that the omega-3 fatty acid, DHA, is cytotoxic to drug-resistant neuroblastoma. The potent action of DHA with arsenic trioxide, NSAID and chemotherapeutic agents suggests clinical testing of this therapeutic concept in children with neuroblastoma.


Assuntos
Arsênio/toxicidade , Ácidos Docosa-Hexaenoicos/farmacologia , Resistência a Múltiplos Medicamentos , Neuroblastoma/patologia , Estresse Oxidativo , Antioxidantes/farmacologia , Apoptose , Relação Dose-Resposta a Droga , Fase G1 , Humanos , Células Tumorais Cultivadas , Vitamina E/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...