Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Biochem Biophys Res Commun ; 589: 1-8, 2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-34883284

RESUMO

BNIP3 is found to eliminate cancer cells via causing mitochondrial damage and endoplasmic reticulum stress, but it remains elusive of its role in regulating DNA double strand breaks (DSBs). In this study, we find that silibinin triggers DNA DSBs, ROS accumulation and expressional upregulation of BNIP3 in glioma cells. Mitigation of ROS with antioxidant GSH significantly inhibits silibinin-induced DNA DSBs and glioma cell death. Then, we find knockdown of BNIP3 with SiRNA obviously prevents silibinin-induced DNA DSBs and ROS accumulation. Mechanistically, BNIP3 knockdown not only reverses silibinin-triggered depletion of cysteine and GSH via maintaining xCT level, but also abrogates catalase decrease. Notably, silibinin-induced dephosphorylation of mTOR is also prevented when BNIP3 is knocked down. Given that activated mTOR could promote xCT expression and inhibit autophagic degradation of catalase, our data suggest that BNIP3 contributes to silibinin-induced DNA DSBs via improving intracellular ROS by inhibition of mTOR.


Assuntos
Quebras de DNA de Cadeia Dupla , Glioma/metabolismo , Glioma/patologia , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Silibina/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Sistema y+ de Transporte de Aminoácidos/metabolismo , Catalase/metabolismo , Linhagem Celular Tumoral , Cisteína/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
Acta Pharmacol Sin ; 42(8): 1324-1337, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33879840

RESUMO

FOXO3a (forkhead box transcription factor 3a) is involved in regulating multiple biological processes in cancer cells. BNIP3 (Bcl-2/adenovirus E1B 19-kDa-interacting protein 3) is a receptor accounting for priming damaged mitochondria for autophagic removal. In this study we investigated the role of FOXO3a in regulating the sensitivity of glioma cells to temozolomide (TMZ) and its relationship with BNIP3-mediated mitophagy. We showed that TMZ dosage-dependently inhibited the viability of human U87, U251, T98G, LN18 and rat C6 glioma cells with IC50 values of 135.75, 128.26, 142.65, 155.73 and 111.60 µM, respectively. In U87 and U251 cells, TMZ (200 µM) induced DNA double strand breaks (DSBs) and nuclear translocation of apoptosis inducing factor (AIF), which was accompanied by BNIP3-mediated mitophagy and FOXO3a accumulation in nucleus. TMZ treatment induced intracellular ROS accumulation in U87 and U251 cells via enhancing mitochondrial superoxide, which not only contributed to DNA DSBs and exacerbated mitochondrial dysfunction, but also upregulated FOXO3a expression. Knockdown of FOXO3a aggravated TMZ-induced DNA DSBs and mitochondrial damage, as well as glioma cell death. TMZ treatment not only upregulated BNIP3 and activated autophagy, but also triggered mitophagy by prompting BNIP3 translocation to mitochondria and reinforcing BNIP3 interaction with LC3BII. Inhibition of mitophagy by knocking down BNIP3 with SiRNA or blocking autophagy with 3MA or bafilomycin A1 exacerbated mitochondrial superoxide and intracellular ROS accumulation. Moreover, FOXO3a knockdown inhibited TMZ-induced BNIP3 upregulation and autophagy activation. In addition, we showed that treatment with TMZ (100 mg·kg-1·d-1, ip) for 12 days in C6 cell xenograft mice markedly inhibited tumor growth accompanied by inducing FOXO3a upregulation, oxidative stress and BNIP3-mediated mitophagy in tumor tissues. These results demonstrate that FOXO3a attenuates temozolomide-induced DNA double strand breaks in human glioma cells via promoting BNIP3-mediated mitophagy.


Assuntos
Antineoplásicos/uso terapêutico , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Proteína Forkhead Box O3/metabolismo , Glioma/metabolismo , Mitofagia/efeitos dos fármacos , Temozolomida/uso terapêutico , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Glioma/tratamento farmacológico , Humanos , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Regulação para Cima/efeitos dos fármacos
3.
Acta Pharmacol Sin ; 42(10): 1690-1702, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34112960

RESUMO

Ferroptotic cell death is characterized by iron-dependent lipid peroxidation that is initiated by ferrous iron and H2O2 via Fenton reaction, in which the role of activating transcription factor 3 (ATF3) remains elusive. Brucine is a weak alkaline indole alkaloid extracted from the seeds of Strychnos nux-vomica, which has shown potent antitumor activity against various tumors, including glioma. In this study, we showed that brucine inhibited glioma cell growth in vitro and in vivo, which was paralleled by nuclear translocation of ATF3, lipid peroxidation, and increases of iron and H2O2. Furthermore, brucine-induced lipid peroxidation was inhibited or exacerbated when intracellular iron was chelated by deferoxamine (500 µM) or improved by ferric ammonium citrate (500 µM). Suppression of lipid peroxidation with lipophilic antioxidants ferrostatin-1 (50 µM) or liproxstatin-1 (30 µM) rescued brucine-induced glioma cell death. Moreover, knockdown of ATF3 prevented brucine-induced accumulation of iron and H2O2 and glioma cell death. We revealed that brucine induced ATF3 upregulation and translocation into nuclei via activation of ER stress. ATF3 promoted brucine-induced H2O2 accumulation via upregulating NOX4 and SOD1 to generate H2O2 on one hand, and downregulating catalase and xCT to prevent H2O2 degradation on the other hand. H2O2 then contributed to brucine-triggered iron increase and transferrin receptor upregulation, as well as lipid peroxidation. This was further verified by treating glioma cells with exogenous H2O2 alone. Moreover, H2O2 reversely exacerbated brucine-induced ER stress. Taken together, ATF3 contributes to brucine-induced glioma cell ferroptosis via increasing H2O2 and iron.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Antineoplásicos/uso terapêutico , Ferroptose/efeitos dos fármacos , Peróxido de Hidrogênio/metabolismo , Ferro/metabolismo , Estricnina/análogos & derivados , Sistema y+ de Transporte de Aminoácidos/metabolismo , Animais , Antineoplásicos/farmacologia , Catalase/metabolismo , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , NADPH Oxidase 4/metabolismo , Neoplasias/tratamento farmacológico , Estricnina/farmacologia , Estricnina/uso terapêutico , Superóxido Dismutase-1/metabolismo , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Biochem Biophys Res Commun ; 518(3): 590-597, 2019 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-31445705

RESUMO

RSL3 is a type of small molecular compound which can inactivate glutathione peroxidase 4 (GPX4) and induce ferroptosis, but its role in glioma cell death remains unclear. In this study, we found RSL3 inhibited the viabilities of glioma cells and induced glioma cell death in a dose-dependent manner. In vitro studies revealed that RSL3-induced cell death was accompanied with the changes of autophagy-associated protein levels and was alleviated by pretreatment of 3-Methyladenine, bafilomycin A1 and knockdown of ATG5 with siRNA. The ATP and pyruvate content as well as the protein levels of HKII, PFKP, PKM2 were decreased in cells treated by RSL3, indicating that RSL3 induced glycolysis dysfunction in glioma cells. Moreover, supplement of exterior sodium pyruvate, which was a final product of glycolysis, not only inhibited the changes of autophagy-associated protein levels caused by RSL3, but also prevented RSL3-induced cell death. In vivo data suggested that the inhibitory effect of RSL3 on the growth of glioma cells was associated with glycolysis dysfunction and autophagy activation. Taken together, RSL3 induced autophagic cell death in glioma cells via causing glycolysis dysfunction.


Assuntos
Antineoplásicos/farmacologia , Morte Celular Autofágica/efeitos dos fármacos , Carbolinas/farmacologia , Glioma/tratamento farmacológico , Glicólise/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Carbolinas/uso terapêutico , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Glioma/metabolismo , Glioma/patologia , Glutationa Peroxidase/antagonistas & inibidores , Glutationa Peroxidase/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ratos
6.
Front Pharmacol ; 13: 919636, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36408275

RESUMO

Anesthetics are essential for cancer surgery, but accumulated research have proven that some anesthetics promote the occurrence of certain cancers, leading to adverse effects in the lives of patients. Although anesthetic technology is mature, there is no golden drug selection standard for surgical cancer treatment. To afford the responsibility of human health, a more specific regimen for cancer resection is indeed necessary. Immunosuppression in oncologic surgery has an adverse influence on the outcomes of patients. The choice of anesthetic strategies influences perioperative immunity. Among anesthetics, propofol has shown positive effects on immunity. Apart from that, propofol's anticancer effect has been generally reported, which makes it more significant in oncologic surgery. However, the immunoregulative function of propofol is not reorganized well. Herein, we have summarized the impact of propofol on different immunocytes, proposed its potential mechanism for the positive effect on cancer immunity, and offered a conceivable hypothesis on its regulation to postoperative inflammation. We conclude that the priority of propofol is high in oncologic surgery and propofol may be a promising immunomodulatory drug for tumor therapy.

7.
Oncol Lett ; 20(4): 57, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32793311

RESUMO

Erastin is a small molecular compound that induces ferroptosis by binding to voltage-dependent anion-selective channel protein (VDAC)2, VDAC3 and solute carrier family 7 member 5 inhibiting the cystine/glutamate antiporter. However, to the best of our knowledge, the mechanism of erastin-induced breast cancer cell death remains unclear. In present study aimed to explore the underlying mechanisms of the antitumor effects of erastin on breast cancer cells. Cellular viability was assessed using an MTT assay, a lactate dehydrogenase cytotoxicity assay kit was used to determine the cell death rate, the intracellular Fe2+ levels were determined using an iron colorimetric assay kit and western blotting was used to estimate the changes of autophagy-associated proteins levels. The present study demonstrated that erastin inhibited the viability of breast cancer cells and induced breast cancer cell death in a dose-dependent manner. Additionally, autophagy was activated by erastin, as demonstrated by upregulated expression levels of autophagy-associated proteins in breast cancer cells. Bafilomycin A1, 3-methyladenine and knockdown of autophagy related (ATG)5 with small interfering RNA prevented erastin-induced breast cancer cell death and inhibited the erastin-induced changes in the expression levels of the autophagy-associated proteins beclin1, ATG5, ATG12, microtubule-associated proteins 1A/1B light chain 3B (LC3B) and P62. Furthermore, erastin-induced breast cancer cell death was inhibited by an iron chelator, deferoxamine, which inhibited the increases of erastin-induced iron levels and inhibited the erastin-induced changes in the expression levels of the autophagy-related proteins beclin1, ATG5, ATG12, LC3B and P62. In summary, erastin triggered autophagic death in breast cancer cells by increasing intracellular iron levels.

8.
Cell Death Dis ; 11(8): 630, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32801360

RESUMO

Induction of lethal autophagy has become a strategy to eliminate glioma cells, but it remains elusive whether autophagy contributes to cell death via causing mitochondria damage and nuclear translocation of apoptosis inducing factor (AIF). In this study, we find that silibinin induces AIF translocation from mitochondria to nuclei in glioma cells in vitro and in vivo, which is accompanied with autophagy activation. In vitro studies reveal that blocking autophagy with 3MA, bafilomycin A1 or by knocking down ATG5 with SiRNA inhibits silibinin-induced mitochondrial accumulation of superoxide, AIF translocation from mitochondria to nuclei and glioma cell death. Mechanistically, silibinin activates autophagy through depleting ATP by suppressing glycolysis. Then, autophagy improves intracellular H2O2 via promoting p53-mediated depletion of GSH and cysteine and downregulation of xCT. The increased H2O2 promotes silibinin-induced BNIP3 upregulation and translocation to mitochondria. Knockdown of BNIP3 with SiRNA inhibits silibinin-induced mitochondrial depolarization, accumulation of mitochondrial superoxide, and AIF translocation from mitochondria to nuclei, as well as prevents glioma cell death. Furthermore, we find that the improved H2O2 reinforces silibinin-induced glycolysis dysfunction. Collectively, autophagy contributes to silibinin-induced glioma cell death via promotion of oxidative stress-mediated BNIP3-dependent nuclear translocation of AIF.


Assuntos
Fator de Indução de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Núcleo Celular/metabolismo , Glioma/metabolismo , Glioma/patologia , Proteínas de Membrana/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Silibina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Glutationa/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Silibina/química , Proteína Supressora de Tumor p53/metabolismo
9.
Stereotact Funct Neurosurg ; 87(2): 114-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19223698

RESUMO

AIMS: The aim of this study was to assess the clinical effects of Gamma Knife radiosurgery (GKS) for recurrent and residual meningeal hemangiopericytomas (M-HPC). METHODS: Between December 1994 and December 2006, 22 patients with recurrent and residual M-HPC with 58 foci underwent GKS at the Gamma Knife Center of Beijing Neurosurgical Institute. Of these 22 patients, 13 patients (59.1%) were males and 9 patients (40.9%) were females. The mean age was 40.9 years (range 16-64 years). The mean volume of these tumors was 5.4 cm(3) (range 0.1-37.2 cm(3)). The mean tumor margin dose was 13.5 Gy (range 10.0-20.0 Gy). The mean tumor central dose was 28.2 Gy (range 21.8-35.0 Gy). The mean prescription isodose line was 48.4% (range 30.0-70.0%). RESULTS: The mean period of follow-up was 26.0 months (range 5-90 months). Of these 22 patients, intracranial metastases developed in 7 patients (31.8%), extracranial metastases developed in 3 patients (13.6%). Four patients died. The mean life expectancy of these 22 patients was 67.7 months (range 7-192 months). Of these 58 foci, radiological follow-up showed that 25 foci (43.1%) nearly disappeared, 13 foci (22.4%) shrunk, 14 foci (24.1%) remained stable and 6 foci (10.3%) enlarged. The overall tumor control rate was 89.7%. CONCLUSION: GKS provides an effective and safe adjunct management for postoperative small-to-moderate sized M-HPC and plays an important role in controlling recurrent and residual M-HPC to avoid a repeat surgical resection.


Assuntos
Neoplasias Meníngeas/cirurgia , Meningioma/cirurgia , Recidiva Local de Neoplasia/cirurgia , Neoplasia Residual/cirurgia , Radiocirurgia , Adolescente , Adulto , Terapia Combinada , Feminino , Seguimentos , Humanos , Imageamento por Ressonância Magnética , Masculino , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/radioterapia , Meningioma/patologia , Meningioma/radioterapia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Neoplasia Residual/patologia , Taxa de Sobrevida , Adulto Jovem
10.
Cancer Lett ; 467: 58-71, 2019 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-31560934

RESUMO

Chromatinolysis refers to enzymatic degradation of nuclear DNA and is regarded as one of the crucial events leading to cell death. Mixed-lineage kinase domain-like protein (MLKL) has been identified as a key executor of necroptosis, but it remains unclear whether MLKL contributes to necroptosis via regulation of chromatinolysis. In this study, we find that shikonin induces MLKL activation and chromatinolysis in glioma cells in vitro and in vivo, which are accompanied with nuclear translocation of AIF and γ-H2AX formation. In vitro studies reveal that inhibition of MLKL with its specific inhibitor NSA or knockdown of MLKL with siRNA abrogates shikonin-induced glioma cell necroptosis, as well as chromatinolysis. Mechanistically, activated MLKL targets mitochondria and triggers excessive generation of mitochondrial superoxide, which promotes AIF translocation into nucleus via causing mitochondrial depolarization and aggravates γ-H2AX formation via improving intracellular accumulation of ROS. Inhibition of nuclear level of AIF by knockdown of AIF with siRNA or mitigation of γ-H2AX formation by suppressing ROS with antioxidant NAC effectively prevents shikonin-induced chromatinolysis. Then, we found that RIP3 accounts for shikonin-induced activation of MLKL, and activated MLKL reversely up-regulates the protein level of CYLD and promotes the activation of RIP1 and RIP3. Taken together, our data suggest that MLKL contributes to shikonin-induced glioma cell necroptosis via promotion of chromatinolysis, and shikonin induces a positive feedback between MLKL and its upstream signals RIP1 and RIP3.


Assuntos
DNA de Neoplasias/química , Glioma/tratamento farmacológico , Naftoquinonas/administração & dosagem , Proteínas Quinases/metabolismo , Animais , Fator de Indução de Apoptose/metabolismo , Linhagem Celular Tumoral , Fragmentação do DNA , DNA de Neoplasias/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Mitocôndrias/metabolismo , Naftoquinonas/farmacologia , Necroptose , Ratos , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Lett ; 428: 21-33, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29702192

RESUMO

Ferroptosis is a form of programmed cell death decided by iron-dependent lipid peroxidation, but its role in glioma cell death remains unclear. In this study, we found Pseudolaric acid B (PAB) inhibited the viabilities of glioma cells in vitro and in vivo, which was accompanied by abnormal increases of intracellular ferrous iron, H2O2 and lipid peroxidation, as well as depletion of GSH and cysteine. In vitro studies revealed that the lipid peroxidation and the cell death caused by PAB were both inhibited by iron chelator deferoxamine, but exacerbated by supplement of ferric ammonium citrate. Inhibition of lipid peroxidation with ferrostatin-1 or GSH rescued PAB-induced cell death. Morphologically, the cells treated with PAB presented intact membrane, shrunken mitochondria with increased membrane density, and normal-sized nucleus without chromatin condensation. Mechanistically, PAB improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2 and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT pathway, which further exacerbated accumulation of H2O2 and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Diterpenos/farmacologia , Glioma/tratamento farmacológico , Peroxidação de Lipídeos/efeitos dos fármacos , Sistema y+ de Transporte de Aminoácidos/metabolismo , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/transplante , Cicloexilaminas/farmacologia , Modelos Animais de Doenças , Diterpenos/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/patologia , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Ferro/metabolismo , Camundongos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NADPH Oxidase 4/metabolismo , Fenilenodiaminas/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/efeitos dos fármacos
12.
Cancer Lett ; 425: 31-42, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29608987

RESUMO

RIP1 and RIP3 are necroptosis initiators, but their roles in regulation of glycolysis remain elusive. In this study, we found shikonin activated RIP1 and RIP3 in glioma cells in vitro and in vivo, which was accompanied with glycolysis suppression. Further investigation revealed that shikonin-induced decreases of glucose-6-phosphate and pyruvate and downregulation of HK II and PKM2 were significantly prevented when RIP1 or RIP3 was pharmacologically inhibited or genetically knocked down with SiRNA. Moreover, shikonin also triggered accumulation of intracellular H2O2 and depletion of GSH and cysteine. Mitigation of intracellular H2O2 via supplement of GSH reversed shikonin-induced glycolysis suppression. The role of intracellular H2O2 in regulation of glycolysis suppression was further confirmed in the cells treated with exogenous H2O2. Notably, inhibition of RIP1 or RIP3 prevented intracellular H2O2 accumulation, which was correlated with preventing shikonin-induced downregulation of x-CT and depletion of GSH and cysteine. In addition, supplement of pyruvate effectively inhibited shikonin- or exogenous H2O2-induced accumulation of intracellular H2O2 and glioma cell death. Taken together, we demonstrated in this study that RIP1 and RIP3 contributed to shikonin-induced glycolysis suppression via increasing intracellular H2O2.


Assuntos
Glioma/tratamento farmacológico , Glicólise/efeitos dos fármacos , Peróxido de Hidrogênio/metabolismo , Naftoquinonas/administração & dosagem , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Linhagem Celular Tumoral , Cisteína/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/metabolismo , Glutationa/metabolismo , Humanos , Camundongos , Naftoquinonas/farmacologia , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Neurosurg ; 105 Suppl: 144-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18503348

RESUMO

OBJECT: The authors sought to assess the clinical effect of Gamma Knife surgery (GKS) for trigeminal schwannomas. METHODS: Between December 1994 and December 2003, 69 patients with trigeminal schwannomas underwent GKS, and 58 patients were followed up and reviewed at the Beijing Neurosurgical Institute. The mean target volume was 4.6 cm3. The mean peripheral dose was 13.1 Gy, and the mean central dose was 28.3 Gy. The mean radiological follow-up period was 42.5 months. Radiological follow-up demonstrated near-complete disappearance of the tumors in four patients (6.9%), in 34 patients (58.6%) a reduction was seen, in 16 patients (27.6%) no change was observed, and in four patients (6.9%) an enlargement was revealed. The overall tumor control rate was 93.1%. Improvement of presenting neurological symptoms was observed in 28 patients (48.3%), stabilization of presenting neurological symptoms was observed in 23 patients (39.6%), continued progression of presenting neurological symptoms was observed in seven patients (12.1%), and transient cranial nerve dysfunction was observed in six patients (10.4%). Among 13 patients with secondary trigeminal neuralgia, 10 patients had significant improvement or disappearance of trigeminal neuralgia after GKS. CONCLUSIONS: Gamma Knife surgery provides an effective and safe primary and/or adjunct treatment for patients with small- to moderate-sized trigeminal schwannomas, with a low risk of iatrogenic cranial neuropathy and great improvement of clinical symptoms.


Assuntos
Neoplasias dos Nervos Cranianos/cirurgia , Neurilemoma/cirurgia , Radiocirurgia , Doenças do Nervo Trigêmeo/cirurgia , Adolescente , Adulto , Idoso , Criança , Estudos de Coortes , Neoplasias dos Nervos Cranianos/complicações , Neoplasias dos Nervos Cranianos/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neurilemoma/complicações , Neurilemoma/patologia , Estudos Retrospectivos , Resultado do Tratamento , Doenças do Nervo Trigêmeo/complicações , Doenças do Nervo Trigêmeo/patologia , Carga Tumoral , Adulto Jovem
15.
Chin Med J (Engl) ; 115(2): 238-41, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11940340

RESUMO

OBJECTIVE: To investigate the mechanism of multidrug resistance (MDR) in a human glioma cell and methods for overcoming multi-drug resistance. METHODS: MDR cell line C(6)/adr was established. The expression of the mdr-1 gene and its P-glycoprotein (P-gp) in the C(6)/adr cell line was observed by RT-PCR and flow cytometry. The reversal of MDR by verapamil, erythromycin, dihydropyridine, P-gp monoclonal antibody and Salvia miltiorrhiza (SM) was studied by microtiter tetrazolium (MTT) assay or by high performance liquid chromatographic assay. RESULTS: The mdr-1 gene of the C(6)/adr cell line was positive, over-expressing P-gp. The drug-resistance of the C(6)/adr cell lines could be partly reversed by 2 - 6 microg/ml of verapamil, 50 - 100 microg/ml of erythromycin, or 5 microg/ml of dihydropyridine. As concentration increased, they had a better effect. Among these drugs, 100 microg/ml of erythromycin had the best result of reversal. Dihydropyridine 1 microg/ml, P-gp monoclonal antibody and SM had no effect. CONCLUSION: The mdr-1 gene and its expression might be associated with the MDR of glioma cells. Verapamil, erythromycin and dihydropyridine could reverse the MDR of glioma cells.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Sobrevivência Celular/genética , Relação Dose-Resposta a Droga , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/genética , Eritromicina/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Células Tumorais Cultivadas/efeitos dos fármacos , Verapamil/farmacologia
17.
Biochem Biophys Res Commun ; 325(4): 1240-5, 2004 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-15555559

RESUMO

The decreased antitumor immune response significantly contributes to the progression of glioma. To evaluate whether the antitumor immunity is restored by stable co-expression of IL-18 and Fas receptor, we retrovirally introduced these two genes into rat C6 glioma cells. We found that IL-18-transduced glioma cells secreted IL-18 and induced PBMC IFN-gamma production in vitro. We also found that Fas-transduced glioma cells were susceptible to Fas-mediated apoptosis. In vivo, we found that IL-18 expression and Fas expression synergistically inhibited C6 cell tumorigenesis with the glioma cells being subcutaneously injected in rat flank. Furthermore, we found that co-expression of IL-18 and Fas also produced a marked survival advantage with the rats being intracerebrally implanted with the glioma cells. Finally, we demonstrated that FasL-dependent PBMC cytotoxicity participated in the anti-glioma immunity induced by IL-18 and Fas expression. Taken together, these findings demonstrate that increasing IL-18 production in tumor microenvironment and prompting functional Fas receptor expression of tumor cells could enhance FasL-dependent cytotoxic antitumor immunity.


Assuntos
Glioma/metabolismo , Glioma/terapia , Interleucina-18/metabolismo , Glicoproteínas de Membrana/metabolismo , Engenharia de Proteínas/métodos , Receptor fas/metabolismo , Animais , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Proteína Ligante Fas , Técnicas de Transferência de Genes , Glioma/genética , Glioma/patologia , Interleucina-18/genética , Ratos , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Análise de Sobrevida , Resultado do Tratamento , Receptor fas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA