Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogenesis ; 6(5): e336, 2017 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-28504693

RESUMO

High-grade glioma (HGG) is an incurable brain cancer. The transcriptomes of cells within HGG tumors are highly heterogeneous. This renders the tumors unresponsive or able to adapt to therapeutics targeted at single pathways, thereby causing treatment failure. To overcome this, we focused on cyclin-dependent kinase 7 (CDK7), a ubiquitously expressed molecule involved in two major drivers of HGG pathogenesis: cell cycle progression and RNA polymerase-II-based transcription. We tested the activity of THZ1, an irreversible CDK7 inhibitor, on patient-derived primary HGG cell lines and ex vivo HGG patient tissue slices, using proliferation assays, microarray analysis, high-resolution respirometry, cell cycle analysis and in vivo tumor orthografts. The cellular processes affected by CDK7 inhibition were analyzed by reverse transcriptase-quantitative PCR, western blot, flow cytometry and immunofluorescence. THZ1 perturbed the transcriptome and disabled CDK activation, leading to cell cycle arrest at G2 and DNA damage. THZ1 halted transcription of the nuclear-encoded mitochondrial ribosomal genes, reducing mitochondrial translation and oxidative respiration. It also inhibited the expression of receptor tyrosine kinases such as epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor-α (PDGFR-α), reducing signaling flux through the AKT, extracellular-signal-regulated kinase 1/2 (ERK1/2) and signal transducer and activator of transcription 3 (STAT3) downstream pathways. Finally, THZ1 disrupted nucleolar, Cajal body and nuclear speckle formation, resulting in reduced cytosolic translation and malfunction of the spliceosome and thus leading to aberrant mRNA processing. These findings indicate that CDK7 is crucial for gliomagenesis, validate CDK7 as a therapeutic target and provide new insight into the cellular processes that are affected by THZ1 and induce antitumor activity.

2.
Cell Death Discov ; 2: 16016, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27551510

RESUMO

Although mitochondrial DNA has been implicated in diseases such as cancer, its role remains to be defined. Using three models of tumorigenesis, namely glioblastoma multiforme, multiple myeloma and osteosarcoma, we show that mitochondrial DNA plays defining roles at early and late tumour progression. Specifically, tumour cells partially or completely depleted of mitochondrial DNA either restored their mitochondrial DNA content or actively recruited mitochondrial DNA, which affected the rate of tumorigenesis. Nevertheless, non-depleted tumour cells modulated mitochondrial DNA copy number at early and late progression in a mitochondrial DNA genotype-specific manner. In glioblastoma multiforme and osteosarcoma, this was coupled with loss and gain of mitochondrial DNA variants. Changes in mitochondrial DNA genotype affected tumour morphology and gene expression patterns at early and late progression. Importantly, this identified a subset of genes that are essential to early progression. Consequently, mitochondrial DNA and commonly expressed early tumour-specific genes provide novel targets against tumorigenesis.

3.
Cell Death Dis ; 6: e1664, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25719248

RESUMO

Mitochondrial DNA (mtDNA) copy number is strictly regulated during differentiation so that cells with a high requirement for ATP generated through oxidative phosphorylation have high mtDNA copy number, whereas those with a low requirement have few copies. Using immunoprecipitation of DNA methylation on 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC), which distinguish between de novo DNA methylation and demethylation, respectively, we set out to determine whether DNA methylation at exon 2 of the human mtDNA-specific polymerase (DNA polymerase gamma A (POLGA)) regulates cell-specific mtDNA copy number in highly proliferative and terminally differentiated cells. Highly proliferative cancer and pluripotent and multipotent cells possessed low mtDNA copy number and were highly methylated at exon 2 of POLGA in contrast to post-mitotic cells. Unlike neural stem cells, cancer cells were unable to differentiate and remained extensively DNA methylated at exon 2 of POLGA. However, mtDNA depletion of cancer cells reduced DNA methylation at exon 2 of POLGA as they replenished mtDNA to form tumours in mice. Glioblastoma cells treated with the DNA demethylation agent 5-azacytidine over 28 days of astrocyte-induced differentiation demethylated exon 2 of POLGA leading to increased mtDNA copy number and expression of the astrocyte endpoint marker glial fibrillary acidic protein (GFAP). However, the demethylation agent vitamin C (VitC) was unable to sustain increased mtDNA copy number and differentiation, as was the case when VitC was withdrawn after short-term treatment. These data demonstrate that DNA demethylation of POLGA is an essential regulator of mtDNA copy number and cellular fate and that cancer cells are only able to modulate DNA methylation of POLGA and mtDNA copy number in the presence of a DNA demethylation agent that inhibits de novo methyltransferase 1 activity.


Assuntos
Metilação de DNA/genética , DNA Mitocondrial/genética , DNA Polimerase Dirigida por DNA/genética , Ácido Ascórbico/farmacologia , Azacitidina/farmacologia , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Variações do Número de Cópias de DNA/genética , Metilação de DNA/efeitos dos fármacos , DNA Polimerase gama , DNA Polimerase Dirigida por DNA/metabolismo , Humanos , Imunoprecipitação , Reação em Cadeia da Polimerase em Tempo Real
4.
Oncogene ; 34(41): 5277-87, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25659577

RESUMO

A truncation mutant of the epidermal growth factor receptor, EGFRvIII, is commonly expressed in glioma, an incurable brain cancer. EGFRvIII is tumorigenic, in part, through its transactivation of other receptor tyrosine kinases (RTKs). Preventing the effects of this transactivation could form part of an effective therapy for glioma; however, the mechanism by which the transactivation occurs is unknown. Focusing on the RTK MET, we show that MET transactivation in U87MG human glioma cells in vitro is proportional to EGFRvIII activity and involves MET heterodimerization associated with a focal adhesion kinase (FAK) scaffold. The transactivation of certain other RTKs was, however, independent of FAK. Simultaneously targeting EGFRvIII (with panitumumab) and the transactivated RTKs themselves (with motesanib) in an intracranial mouse model of glioma resulted in significantly greater survival than with either agent alone, indicating that cotargeting these RTKs has potent antitumor efficacy and providing a strategy for treating EGFRvIII-expressing gliomas, which are usually refractory to treatment.


Assuntos
Neoplasias Encefálicas/metabolismo , Receptores ErbB/fisiologia , Glioma/metabolismo , Ativação Transcricional , Analgésicos/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Apoptose , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Feminino , Quinase 1 de Adesão Focal/metabolismo , Glioma/tratamento farmacológico , Glioma/genética , Indóis/farmacologia , Camundongos Endogâmicos BALB C , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Oligonucleotídeos , Panitumumabe , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncogene ; 34(13): 1658-66, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24747966

RESUMO

A feature of many gliomas is the amplification of the epidermal growth factor receptor (EGFR), resulting in its overexpression. Missense mutations or deletions within the extracellular domain are associated with this amplification and can lead to constitutive activation of the receptor, with the Domain I/II deletion, EGFRvIII, being the most common. These changes have also been associated with increased sensitivity to EGFR inhibition using small molecule inhibitors. We have expressed, in human glioma cells, EGFR containing four glioma-specific EGFR missense mutations within Domain IV (C620Y, C624F, C628Y and C636Y) to analyze their biological properties and sensitivity to EGFR inhibition. One of these mutants, C620Y, exhibited an enhanced basal phosphorylation, which was partially dependent on an EGFR-ligand autocrine loop. All Domain IV mutants responded equally as well as wildtype EGFR (wtEGFR) to ligand stimulation. Biochemical analysis revealed that a pre-formed, disulfide-bonded dimer associated with these mutations was underglycosylated, inactive and cytoplasmically retained. Ligand stimulation resulted in the formation of a tyrosine-phosphorylated, disulfide-bonded dimer for all Domain IV mutants but not for wtEGFR. Following treatment with the next-generation, irreversible pan-ErbB inhibitor dacomitinib, the C620Y, C624F and EGFRvIII mutants were inactivated, covalently dimerized and were retained in the cytoplasm, resulting in cell-surface receptor loss and, for C620Y and C624F, decreased binding of EGF. Dacomitinib treatment significantly reduced the in vivo growth of human glioma xenografts bearing C620Y, but not wtEGFR. Collectively, these data indicate that the unique biochemical traits of Domain IV EGFR cysteine mutants can be exploited for enhanced sensitivity to EGFR small molecule inhibitors, with potential clinical applications.


Assuntos
Receptores ErbB/genética , Glioma/tratamento farmacológico , Mutação , Multimerização Proteica , Quinazolinonas/uso terapêutico , Animais , Linhagem Celular Tumoral , Cisteína , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Feminino , Glioma/genética , Glioma/patologia , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Estrutura Terciária de Proteína , Quinazolinonas/farmacologia
6.
Cell Death Discov ; 1: 15028, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27551460

RESUMO

MAPK-activated protein kinase 2 (MK2) is a checkpoint kinase involved in the DNA damage response. MK2 inhibition enhances the efficacy of chemotherapeutic agents; however, whether MK2 inhibition alone, without concurrent chemotherapy, would attenuate survival of cancer cells has not been investigated. CMPD1 is a widely used non-ATP competitive inhibitor that prevents MK2 phosphorylation. We employed CMPD1 together with MK2 knock-down and ATP-competitive MK2 inhibitor III (MK2i) in a panel of glioblastoma cells to assess whether MK2 inhibition could induce cancer cell death. While CMPD1 was effective at selective killing of cancer cells, MK2i and MK2 knock-down had no effect on viability of glioblastoma cells. CMPD1 treatment induced a significant G2/M arrest but MK2i-treated cells were only minimally arrested at G1 phase. Intriguingly, at doses that were cytotoxic to glioblastoma cells, CMPD1 did not inhibit phosphorylation of MK2 and of its downstream substrate Hsp27. These results suggest that CMPD1 exhibits cytotoxic activity independently of MK2 inhibition. Indeed, we identified tubulin as a primary target of the CMPD1 cytotoxic activity. This study demonstrates how functional and mechanistic studies with appropriate selection of test compounds, combining genetic knock-down and pharmacological inhibition, coordinating timing and dose levels enabled us to uncover the primary target of an MK2 inhibitor commonly used in the research community. Tubulin is emerging as one of the most common non-kinase targets for kinase inhibitors and we propose that potential tubulin-targeting activity should be assessed in preclinical pharmacology studies of all novel kinase inhibitors.

7.
Hum Reprod ; 29(7): 1375-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24812320

RESUMO

Non-tubal ectopic pregnancies are a rare subgroup of ectopic pregnancies implanted at sites other than the Fallopian tube. Mortality from non-tubal ectopic pregnancies is higher compared with that for tubal ectopic pregnancies, and they are becoming more common, partly due to the rising incidence of Caesarean sections and use of assisted reproductive technologies. Non-tubal ectopic pregnancies can be especially difficult to treat. Surgical treatment is complex, and follow-up after medical treatment is usually protracted. There is therefore a need for more effective medical therapies to resolve non-tubal ectopic pregnancies and reduce operative intervention. We have recently reported successful use of combination gefitinib (an orally available epidermal growth factor receptor inhibitor) and methotrexate for treatment of tubal pregnancies. To our knowledge, this combination has not been used to treat non-tubal pregnancies. Here we report the use of combination gefitinib and methotrexate to treat eight women with stable, non-tubal ectopic pregnancies at two tertiary academic teaching hospitals (Edinburgh, UK and Melbourne, Australia); five interstitial and three Caesarean section scar ectopic pregnancies. Pretreatment serum hCG levels ranged from 2458 to 48 550 IU/l, and six women had pretreatment hCG levels >5000 IU/l. The women were co-administered 1-2 doses of i.m. methotrexate (50 mg/m² on Day 1, ± Day 4 or Day 7) with seven once daily doses of oral gefitinib (250 mg). The women were monitored until complete resolution of the ectopic pregnancy, defined as a serum hCG <15 IU/l. Time to resolution (days from first methotrexate dose until serum hCG <15 IU/l), safety and tolerability, complication rates and subsequent fertility outcomes were also recorded. All eight women were successfully treated with combination gefitinib and methotrexate. The most common side effects were transient acne/rash and diarrhoea, known side effects of gefitinib. All women promptly resumed menstruation and importantly, three women subsequently conceived spontaneously. Two have delivered a healthy infant at term and the third is currently in her second trimester of pregnancy. Hence, our case series supports a future clinical trial to determine the efficacy of combination gefitinib and methotrexate to treat non-tubal ectopic pregnancies.


Assuntos
Metotrexato/administração & dosagem , Gravidez Ectópica/tratamento farmacológico , Quinazolinas/administração & dosagem , Abortivos não Esteroides/administração & dosagem , Erupções Acneiformes/induzido quimicamente , Adulto , Cesárea/efeitos adversos , Gonadotropina Coriônica/sangue , Tubas Uterinas/fisiopatologia , Feminino , Gefitinibe , Humanos , Gravidez , Resultado da Gravidez , Ultrassonografia Pré-Natal , Adulto Jovem
8.
Cell Death Differ ; 20(12): 1644-53, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23995230

RESUMO

As stem cells undergo differentiation, mitochondrial DNA (mtDNA) copy number is strictly regulated in order that specialized cells can generate appropriate levels of adenosine triphosphate (ATP) through oxidative phosphorylation (OXPHOS) to undertake their specific functions. It is not understood whether tumor-initiating cells regulate their mtDNA in a similar manner or whether mtDNA is essential for tumorigenesis. We show that human neural stem cells (hNSCs) increased their mtDNA content during differentiation in a process that was mediated by a synergistic relationship between the nuclear and mitochondrial genomes and results in increased respiratory capacity. Differentiating multipotent glioblastoma cells failed to match the expansion in mtDNA copy number, patterns of gene expression and increased respiratory capacity observed in hNSCs. Partial depletion of glioblastoma cell mtDNA rescued mtDNA replication events and enhanced cell differentiation. However, prolonged depletion resulted in impaired mtDNA replication, reduced proliferation and induced the expression of early developmental and pro-survival markers including POU class 5 homeobox 1 (OCT4) and sonic hedgehog (SHH). The transfer of glioblastoma cells depleted to varying degrees of their mtDNA content into immunocompromised mice resulted in tumors requiring significantly longer to form compared with non-depleted cells. The number of tumors formed and the time to tumor formation was relative to the degree of mtDNA depletion. The tumors derived from mtDNA depleted glioblastoma cells recovered their mtDNA copy number as part of the tumor formation process. These outcomes demonstrate the importance of mtDNA to the initiation and maintenance of tumorigenesis in glioblastoma multiforme.


Assuntos
Neoplasias Encefálicas/genética , Variações do Número de Cópias de DNA/genética , DNA Mitocondrial/genética , Glioblastoma/genética , Animais , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Respiração Celular/genética , Replicação do DNA/genética , Regulação Neoplásica da Expressão Gênica , Proteína Glial Fibrilar Ácida/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Regulação para Cima/genética
9.
Br J Cancer ; 99(2): 294-304, 2008 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-18594532

RESUMO

TRAIL/Apo-2L has shown promise as an anti-glioma drug, based on investigations of TRAIL sensitivity in established glioma cell lines, but it is not known how accurately TRAIL signalling pathways of glioma cells in vivo are reproduced in these cell lines in vitro. To replicate as closely as possible the in vivo behaviour of malignant glioma cells, 17 early passage glioma cell lines and 5 freshly resected gliomas were exposed to TRAIL-based agents and/or chemotherapeutic drugs. Normal human hepatocytes and astrocytes and established glioma cell lines were also tested. Cross-linked TRAIL, but not soluble TRAIL, killed both normal cell types and cells from three tumours. Cells from only one glioma were killed by soluble TRAIL, although only inefficiently. High concentrations of cisplatin were lethal to glioma cells, hepatocytes and astrocytes. Isolated combinations of TRAIL and chemotherapy drugs were more toxic to particular gliomas than normal cells, but no combination was generally selective for glioma cells. This study highlights the widespread resistance of glioma cells to TRAIL-based agents, but suggests that a minority of high-grade glioma patients may benefit from particular combinations of TRAIL and chemotherapy drugs. In vitro sensitivity assays may help identify effective drug combinations for individual glioma patients.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Glioma/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Adulto , Idoso , Antineoplásicos/administração & dosagem , Astrócitos/efeitos dos fármacos , Carboplatina/administração & dosagem , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Ensaios de Seleção de Medicamentos Antitumorais , Etoposídeo/administração & dosagem , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioma/patologia , Hepatócitos/efeitos dos fármacos , Humanos , Lomustina/administração & dosagem , Masculino , Glicoproteínas de Membrana/administração & dosagem , Pessoa de Meia-Idade , Procarbazina/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Temozolomida , Fator de Necrose Tumoral alfa/administração & dosagem , Vincristina/administração & dosagem
10.
Biochem Pharmacol ; 71(10): 1422-34, 2006 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-16522318

RESUMO

The tyrphostin 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) is a potent and specific inhibitor of EGFR tyrosine kinase whose favourable preclinical profile supports progression towards clinical trials. Microphysiometric evaluation revealed a short (<24 min) effective inhibition of cellular receptor response to EGF challenge in BaF/ERX cells indicating a need to maintain sustained levels of inhibitor. Initial pharmacokinetic evaluation in mice of novel AG1478 formulations in a beta-cyclodextrin (Captisol) showed monoexponential elimination from plasma (half-life 30 min) following subcutaneous administration. A two-fold dose escalation gave a 2.4-fold increase in the total AUC. Bolus i.v. and 6 h continuous infusion were investigated in rats to mimic a more clinically relevant administration regimen. Drug elimination following bolus i.v. administration was biphasic (terminal elimination half-life 30-48 min). The linear relationship between dose and AUC(0-->infinity) (r2=0.979) enabled the prediction of infusion rates and doses for sustained delivery using continuous 6 h infusions, where steady state was reached in 120 min. Plasma levels of AG1478>10 microM were achieved over the duration of the infusion. At the lowest dose, plasma drug levels after the cessation of infusion declined with a half-life of approximately 43 min. EGFR activity, measured both by autophosphorylation and downstream signalling, was inhibited in a dose-dependent manner by injection of AG1478 in mice bearing xenografts of the human glioblastoma cell line U87MG.delta2-7, which expresses a constitutively active variant of the EGF receptor. Taken together, these experiments provide essential data to assess the anti-tumour efficacy of AG1478 and will assist in the rational design of dose regimens for clinical studies.


Assuntos
Inibidores Enzimáticos/farmacocinética , Receptores ErbB/antagonistas & inibidores , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Tirfostinas/farmacocinética , Animais , Área Sob a Curva , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Injeções Intravenosas , Injeções Subcutâneas , Camundongos , Estrutura Molecular , Quinazolinas , Ratos , Timidina/metabolismo , Tirfostinas/química , Tirfostinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Br J Cancer ; 92(6): 1069-77, 2005 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-15770208

RESUMO

We report the generation of a chimeric monoclonal antibody (ch806) with specificity for an epitope on the epidermal growth factor receptor (EGFR) that is different from that targeted by all other anti-EGFR therapies. Ch806 antibody is reactive to both de2-7 and overexpressed wild-type (wt) EGFR but not native EGFR expressed in normal tissues at physiological levels. Ch806 was stably expressed in CHO (DHFR -/-) cells and purified for subsequent characterisation and validated for use in preliminary immunotherapy investigations. Ch806 retained the antigen binding specificity and affinity of the murine parental antibody. Furthermore, ch806 displayed enhanced antibody-dependent cellular cytotoxicity against target cells expressing the 806 antigen in the presence of human effector cells. Ch806 was successfully radiolabelled with both iodine-125 and indium-111 without loss of antigen binding affinity or specificity. The radioimmunoconjugates were stable in the presence of human serum at 37 degrees C for up to 9 days and displayed a terminal half-life (T(1/2beta)) of approximately 78 h in nude mice. Biodistribution studies undertaken in BALB/c nude mice bearing de2-7 EGFR-expressing or amplified EGFR-expressing xenografts revealed that (125)I-labelled ch806 failed to display any significant tumour retention. However, specific and prolonged tumour localisation of (111)In-labelled ch806 was demonstrated with uptake of 31%ID g(-1) and a tumour to blood ratio of 5 : 1 observed at 7 days postinjection. In vivo therapy studies with ch806 demonstrated significant antitumour effects on established de2-7 EGFR xenografts in BALB/c nude mice compared to control, and both murine 806 and the anti-EGFR 528 antibodies. These results support a potential therapeutic role of ch806 in the treatment of suitable EGFR-expressing tumours, and warrants further investigation of the potential of ch806 as a therapeutic agent.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Receptores ErbB/imunologia , Neoplasias Experimentais/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Anticorpos Monoclonais/farmacocinética , Células CHO , Cricetinae , Feminino , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual , Transplante Heterólogo
12.
Cancer Res ; 61(14): 5349-54, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11454673

RESUMO

A mutant epidermal growth factor receptor (variously called DeltaEGFR, de2-7 EGFR, or EGFRvIII) containing a deletion of 267 amino acids of the extracellular domain is frequently highly expressed in human malignant gliomas and has been reported for cancers of the lung, breast, and prostate. We tested the efficacy of a novel monoclonal anti-DeltaEGFR antibody, mAb 806, on the growth of intracranial xenografted gliomas in nude mice. Systemic treatment with mAb 806 significantly reduced the volume of tumors and increased the survival of mice bearing xenografts of U87 MG.DeltaEGFR, LN-Z308.DeltaEGFR, or A1207.DeltaEGFR gliomas, each of which expresses high levels of DeltaEGFR. In contrast, mAb 806 treatment was ineffective with mice bearing the parental U87 MG tumors, which expressed low levels of endogenous wild-type EGFR, or U87 MG.DK tumors, which expressed high levels of kinase-deficient DeltaEGFR. A slight increase of survival of mice xenografted with a wild-type EGFR-overexpressing U87 MG glioma (U87 MG.wtEGFR) was effected by mAb 806 concordant with its weak cross-reactivity with such cells. Treatment of U87 MG.DeltaEGFR tumors in mice with mAb 806 caused decreases in both tumor growth and angiogenesis, as well as increased apoptosis. Mechanistically, in vivo mAb 806 treatment resulted in reduced phosphorylation of the constitutively active DeltaEGFR and caused down-regulated expression of the apoptotic protector, Bcl-XL. These data provide preclinical evidence that mAb 806 treatment may be a useful biotherapeutic agent for those aggressive gliomas that express DeltaEGFR.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Receptores ErbB/genética , Glioblastoma/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Divisão Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/mortalidade , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Nus , Mutação , Neovascularização Patológica/prevenção & controle , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Análise de Sobrevida , Taxa de Sobrevida , Transplante Heterólogo , Resultado do Tratamento , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína bcl-X
13.
Cancer Res ; 61(14): 5355-61, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11454674

RESUMO

The monoclonal antibody (mAb) 806 was raised against the delta2-7 epidermal growth factor receptor (de2-7 EGFR or EGFRvIII), a truncated version of the EGFR commonly expressed in glioma. Unexpectedly, mAb 806 also bound the EGFR expressed by cells exhibiting amplification of the EGFR gene but not to cells or normal tissue expressing the wild-type receptor in the absence of gene amplification. The unique specificity of mAb 806 offers an advantage over current EGFR antibodies, which all display significant binding to the liver and skin in humans. Therefore, we examined the antitumor activity of mAb 806 against human tumor xenografts grown in nude mice. The growth of U87 MG xenografts, a glioma cell line that endogenously expresses approximately 10(5) EGFRs in the absence of gene amplification, was not inhibited by mAb 806. In contrast, mAb 806 significantly inhibited the growth of U87 MG xenografts transfected with the de2-7 EGFR in a dose-dependent manner using both preventative and established tumor models. Significantly, U87 MG cells transfected with the wild-type EGFR, which increased expression to approximately 10(6) EGFRs/cell and mimics the situation of gene amplification, were also inhibited by mAb 806 when grown as xenografts in nude mice. Xenografts treated with mAb 806 all displayed large areas of necrosis that were absent in control tumors. This reduced xenograft viability was not mediated by receptor down-regulation or clonal selection because levels of antigen expression were similar in control and treated groups. The antitumor effect of mAb 806 was not restricted to U87 MG cells because the antibody inhibited the growth of new and established A431 xenografts, a cell line expressing >10(6) EGFRs/cell. This study demonstrates that mAb 806 possesses significant antitumor activity.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Receptores ErbB/genética , Neoplasias Experimentais/tratamento farmacológico , Animais , Anticorpos Monoclonais/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Receptores ErbB/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Genótipo , Humanos , Imuno-Histoquímica , Camundongos , Mutação , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Testes de Precipitina , Ligação Proteica , Transplante Heterólogo , Resultado do Tratamento , Células Tumorais Cultivadas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Neurochem ; 72(1): 1-9, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9886048

RESUMO

Myelin oligodendrocyte glycoprotein (MOG) is a quantitatively minor component of CNS myelin whose function remains relatively unknown. As MOG is an autoantigen capable of producing a demyelinating multiple sclerosis-like disease in mice and rats, much of the research directed toward MOG has been immunological in nature. Although the function of MOG is yet to be elucidated, there is now a relatively large amount of biochemical and molecular data relating to MOG. Here we summarize this information and include our recent findings pertaining to the cloning of the marsupial MOG gene. On the basis of this knowledge we suggest three possible functions for MOG: (a) a cellular adhesive molecule, (b) a regulator of oligodendrocyte microtubule stability, and (c) a mediator of interactions between myelin and the immune system, in particular, the complement cascade. Given that antibodies to MOG and to the myelin-specific glycolipid galactocerebroside (Gal-C) both activate the same signaling pathway leading to MBP degradation, we propose that there is a direct interaction between the membrane-associated regions of MOG and Gal-C. Such an interaction may have important consequences regarding the membrane topology and function of both molecules. Finally, we examine how polymorphisms and/or mutations to the MOG gene could contribute to the pathogenesis of multiple sclerosis.


Assuntos
Doenças Desmielinizantes/metabolismo , Glicoproteína Associada a Mielina/química , Glicoproteína Associada a Mielina/genética , Animais , Dados de Sequência Molecular , Glicoproteína Associada a Mielina/metabolismo , Homologia de Sequência de Aminoácidos
15.
J Mol Med (Berl) ; 75(2): 77-88, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9083925

RESUMO

Myelin oligodendrocyte glycoprotein (MOG) is a member of the immunoglobulin superfamily expressed exclusively in central nervous system (CNS) myelin. While the function of MOG is unknown, a number of studies have shown that immune responses to MOG contribute to the autoimmune-mediated demyelination seen in animals immunized with whole CNS tissue. This paper summarizes our recent studies, which unequivocally demonstrate that MOG by itself is able to generate both an encephalitogenic T cell response and an autoantibody response in Lewis rats and in several strains of mice. In Lewis rats the injection of both native MOG and MOG35-55 peptide produces a paralytic relapsing-remitting neurological disease with extensive plaque-like demyelination. The antibody response to MOG35-55 was highly restricted, as no reactivity to either other MOG peptides or myelin proteins could be detected. Fine epitope mapping showed that antibody from serum and cerebrospinal fluid of injected rats reacted strongly to MOG37-46, which is contiguous to the dominant T cell epitope contained within MOG44-55. NOD/Lt and C57BL/6 mice were also susceptible to severe neurological disease following injection with recombinant MOG or MOG35-55 peptide, indicating that this specific CNS autoantigen, or some of its determinants, can induce a pathogenic response across animal species. Severe paralysis and extensive demyelination were seen in both strains, but NOD/Lt mice experienced a chronic relapsing disease whereas C57BL/6 mice had a chronic non-remitting disease. Moreover, transfer of MOG35-55 T cells into naive NOD/Lt mice also produced severe neurological impairment as well as histological lesions. These results emphasize that a synergism between a T cell-response and anti-MOG antibodies may be important for the development of severe demyelinating disease. This, together with our demonstration that there is a predominant T cell response to MOG in patients with multiple sclerosis, clearly indicates that MOG is probably an important target autoantigen in this disease.


Assuntos
Autoantígenos/imunologia , Esclerose Múltipla/imunologia , Glicoproteína Associada a Mielina/imunologia , Animais , Autoantígenos/fisiologia , Linfócitos B/imunologia , Sistema Nervoso Central , Encefalomielite Autoimune Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Proteínas da Mielina , Glicoproteína Associada a Mielina/fisiologia , Glicoproteína Mielina-Oligodendrócito , Oligopeptídeos/imunologia , Ratos , Ratos Endogâmicos Lew , Linfócitos T/imunologia
16.
Dev Neurosci ; 19(1): 69-78, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9078435

RESUMO

The assembly and function of central nervous system (CNS) myelin requires the coordinated expression of several myelin-specific proteins, including myelin oligodendrocyte glycoprotein (MOG). Despite the recent cloning of MOG, the function of this molecule is still unknown. Because MOG is a late marker of oligodendrocyte maturation and is exclusively expressed in the CNS on the outermost lamellae of the myelin membrane, it is possible that this molecule plays an important role in the control and maintenance of myelination. Furthermore, as a member of the immunoglobulin superfamily that carries the L2/HNK-1 epitope, it has also been suggested that MOG is involved in cell-cell interaction, perhaps functioning as an adhesive molecule for bundles of nerve fibres. In order to further delineate the role of MOG throughout development we have analysed, by immunoblotting, the developmental appearance and accumulation pattern of MOG in the CNS of three mammalian species. We have also purified MOG to homogeneity from five different species including rat, guinea pig, bovine, monkey and human. Immunoblotting revealed two major MOG bands at 28 and 55 kD in all species. The 55 kD band appears to be a dimer of the lower band although treatment with 2-mercaptoethanol or EDTA failed to abolish it. Purified MOG from all species also displayed faint reactivity with bands at 36, 48 and 78 kD. While the 78 kD band may represent a trimer of MOG, the identity of the other bands remains unknown. Developmental studies in mouse, rat, guinea pig and bovine showed at as for other myelin proteins, MOG displayed a caudorostral gradient of expression, appearing in the spinal cord before the brain. The sensitivity of the detection system used here allowed us to detect MOG protein earlier than in previous reports such that its presence was clearly demonstrated in the CNS of mice and rats at 14 and 10 days after birth, respectively. Analysis of MOG expression in a novel transgenic mouse model that has both delayed and reduced myelination revealed that, like other myelin proteins, MOG expression was delayed compared with normal littermates. These results demonstrate that the expression of MOG is similar in all species and is regulated in a manner consistent with other myelin-specific proteins.


Assuntos
Envelhecimento/metabolismo , Glicoproteína Associada a Mielina/metabolismo , Animais , Bovinos , Cricetinae , Eletroforese em Gel de Poliacrilamida , Cobaias , Haplorrinos , Humanos , Camundongos , Camundongos Mutantes Neurológicos/crescimento & desenvolvimento , Camundongos Mutantes Neurológicos/metabolismo , Camundongos Transgênicos , Proteínas da Mielina/metabolismo , Glicoproteína Associada a Mielina/isolamento & purificação , Glicoproteína Mielina-Oligodendrócito , Ratos , Especificidade da Espécie , Distribuição Tecidual
17.
Mol Immunol ; 34(1): 33-8, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9182874

RESUMO

Myelin oligodendrocyte glycoprotein (MOG) is a myelin-specific protein restricted to the central nervous system (CNS). While MOG is considered a putative autoantigen in MS, its function(s) in myelin is unknown. As CNS myelin is able to activate the classical complement pathway, it must contain a Clq-binding/activating protein but the identity of this protein has not been reported. The data in this paper clearly demonstrate that MOG specifically binds Clq in a dose-dependent and saturating manner. This calcium-dependent interaction is mediated by the extracellular immunoglobulin-like domain of MOG. This MOG domain contains an amino acid motif similar to the core Clq-binding sequence previously identified in IgG antibodies. Purified MOG also inhibited the antibody-dependent lysis of RBC by complement. Taken together, these results demonstrate that MOG binds Clq near the IgG binding site and may be the protein responsible for complement activation in myelin. This direct interaction between a myelin-specific protein and Clq has significant implications for CNS inflammation and could be particularly important in demyelinating diseases such as multiple sclerosis.


Assuntos
Doenças do Sistema Nervoso Central/patologia , Complemento C1q/metabolismo , Glicoproteína Associada a Mielina/metabolismo , Oligodendroglia/metabolismo , Animais , Cálcio/fisiologia , Doenças do Sistema Nervoso Central/imunologia , Ensaio de Atividade Hemolítica de Complemento , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Espaço Extracelular/química , Humanos , Inflamação/imunologia , Proteína Básica da Mielina/imunologia , Proteínas da Mielina , Glicoproteína Associada a Mielina/química , Glicoproteína Mielina-Oligodendrócito , Ligação Proteica/imunologia , Estrutura Terciária de Proteína , Ratos
18.
Int Immunol ; 8(11): 1667-74, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8943561

RESUMO

Previous studies from our laboratory have demonstrated a predominant response to myelin oligodendrocyte glycoprotein (MOG) in patients with multiple sclerosis (MS) and showed that this molecule is able to induce in Lewis rats a chronic relapsing MS-like disease with extensive demyelination. To further study the possibility that MOG is a primary target antigen in MS, we have begun to investigate the encephalitogenicity and antibody response of different sequences of the extracellular domains of MOG in Lewis rats. We report that none of the synthetic peptides encompassing the MOG amino acid sequences 1-21, 67-87, 104-117 and 202-218 were encephalitogenic. In contrast, a single injection of MOG35-55 was able to induce severe neurological signs associated with inflammation and demyelination. All rats injected with MOG peptides 1-21, 35-55, 67-87 and 202-218 developed a high level of antibodies to their respective immunizing peptides as detected by ELISA and immunoblotting. Although all MOG peptide antisera reacted with immunoblots of native MOG separated under reducing conditions, only anti-MOG35-55 and anti-MOG202-218 antibodies reacted to native MOG, when tested under nonreducing conditions. These results indicate that the MOG35-55 peptide, which is found in the extracellular Ig V-like domain of MOG, is not only an encephalitogenic epitope but could also be an important determinant for initiating antibody-mediated demyelination. As indicated by the absence of reactivity to the other MOG peptides tested, as well as other central nervous system myelin proteins including myelin basic protein and proteolipid protein, the antibody response produced by MOG peptides is highly restricted.


Assuntos
Autoanticorpos/biossíntese , Glicoproteína Associada a Mielina/administração & dosagem , Glicoproteína Associada a Mielina/imunologia , Peptídeos/administração & dosagem , Peptídeos/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Superfície/administração & dosagem , Antígenos de Superfície/imunologia , Reações Cruzadas , Encefalomielite Autoimune Experimental , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Injeções Intramusculares , Dados de Sequência Molecular , Proteínas da Mielina , Glicoproteína Mielina-Oligodendrócito , Ratos , Ratos Endogâmicos Lew
19.
J Immunol ; 157(2): 919-26, 1996 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8752946

RESUMO

We have recently shown that a single injection of myelin oligodendrocyte glycoprotein (MOG), or the MOG35-55 peptide, produces a relapsing-remitting neurologic disease with extensive plaque-like demyelination. Given the features that this new autoimmune demyelinating model has in common with the clinicopathologic manifestations of multiple sclerosis, we have examined the Ab reactivity to native MOG and MOG35-55 peptide during the course of the disease in Lewis rats. Following immunization with MOG35-55, varied clinical symptoms were observed; these included hind and foreleg paralysis and various degrees of balance impairment. Disease progression also varied: 3 out of 21 animals had a single mild disease episode; 4 out of 21 had a mild relapsing-remitting disease; and 14 out of 21 had severe relapsing-remitting disease. Ab reactivity to MOG35-55 and native MOG was first detected in all rats 4 wk postimmunization and persisted throughout the 12 wk of observation. The Ab response was highly restricted with no reactivity to other peptides encompassing different extracellular segments of MOG. Fine epitope mapping showed that Ab from serum and cerebrospinal fluid of injected rats reacted strongly to MOG37-46 and to a lesser extent to MOG43-50. Although significant levels of anti-MOG Abs appeared necessary for the development of demyelinating lesions, their presence in blood and cerebrospinal fluid alone was not sufficient to produce severe clinical symptoms. These results demonstrate that the MOG35-55 peptide is highly encephalitogenic and can induce strong T and B cell responses. It is probably the complex interaction between these T and B cells that determines the severity of disease in individual rats.


Assuntos
Linfócitos B/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Epitopos/imunologia , Esclerose Múltipla/imunologia , Glicoproteína Associada a Mielina/imunologia , Oligodendroglia/imunologia , Peptídeos/imunologia , Sequência de Aminoácidos , Animais , Formação de Anticorpos , Especificidade de Anticorpos , Linfócitos B/metabolismo , Doença Crônica , Epitopos/líquido cefalorraquidiano , Membro Posterior , Hipersensibilidade Tardia/imunologia , Soros Imunes/análise , Injeções , Masculino , Dados de Sequência Molecular , Esclerose Múltipla/líquido cefalorraquidiano , Proteínas da Mielina , Glicoproteína Associada a Mielina/administração & dosagem , Glicoproteína Mielina-Oligodendrócito , Peptídeos/administração & dosagem , Peptídeos/líquido cefalorraquidiano , Ratos , Ratos Endogâmicos Lew , Recidiva
20.
J Immunol ; 154(10): 5536-41, 1995 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7537310

RESUMO

Chronic relapsing experimental autoimmune encephalomyelitis, a demyelinating disease induced by injection of central nervous system (CNS) tissue, is widely used as a model for multiple sclerosis. However, it is unclear which Ag or combination of Ags in the CNS induce the demyelinating immune response. We now show in Lewis rats that a single injection of myelin oligodendrocyte glycoprotein, a specific CNS myelin component, or an appropriately derived myelin oligodendrocyte glycoprotein peptide produces a relapsing remitting neurologic disease with extensive plaque-like demyelination. Igs from affected animals reacted specifically with myelin oligodendrocyte glycoprotein and stimulated a myelin protease activity, leading to myelin basic protein degradation. The demonstrated involvement of myelin oligodendrocyte glycoprotein as a new demyelinating neural Ag may provide a deeper insight into the pathogenesis of multiple sclerosis and its treatment.


Assuntos
Encefalomielite Autoimune Experimental/induzido quimicamente , Glicoproteínas de Membrana/toxicidade , Proteínas da Mielina/toxicidade , Glicoproteína Associada a Mielina , Sequência de Aminoácidos , Animais , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Soros Imunes/imunologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/imunologia , Dados de Sequência Molecular , Proteína Básica da Mielina/metabolismo , Proteínas da Mielina/química , Proteínas da Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/toxicidade , Ratos , Ratos Endogâmicos Lew
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...