Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Cancer ; 18(1): 1016, 2018 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-30348128

RESUMO

BACKGROUND: The Insulin-like growth factor (IGF) pathway plays a role in tumour development and progression. In vivo, IGF1 activity is regulated by the IGF binding proteins (IGFBPs). IGFBP4 inhibits the activity of IGF1 but proteolytic cleavage by pregnancy-associated plasma protein-A (PAPP-A) releases active IGF1. A modified IGFBP4, dBP4, which was resistant to PAPP-A cleavage but retained IGF1 binding capacity, was engineered, expressed in Human Embryonic Kidney (HEK) 293 cells and purified. This study examined the effects of dBP4 on IGF1-induced cell migration, invasion and angiogenesis in vitro. The effect of intra-tumour injections of dBP4 on tumour angiogenesis and metastasis was examined using the 4T1.2luc orthotopic model of breast cancer. METHODS: PAPP-A resistance and IGF binding capacity of dBP4 were characterized by Western blot and surface plasmon resonance, respectively. 4T1.2luc are mouse mammary adenocarcinoma cells transfected with luciferase to allow in vivo imaging. The effect of dBP4 on IGF1-induced Akt activation in 4T1.2luc cells was assessed by Western blot. Cell migration and invasion assays were performed using 4T1.2luc cells. Angiokit™ assays and Matrigel® implants were used to assess the effects of dBP4 on angiogenesis in vitro and in vivo, respectively. An orthotopic breast cancer model - 4T1.2luc cells implanted in the mammary fat pad of BALB/c mice - was used to assess the effect of intra tumour injection of purified dBP4 on tumour angiogenesis and metastasis. Tumour growth and lung metastasis were examined by in vivo imaging and tumour angiogenesis was evaluated by CD31 immunohistochemistry. RESULTS: Our engineered, PAPP-A resistant IGFBP4 (dBP4) retained IGF1 binding capacity and inhibited IGF1 activation of Akt as well as IGF1-induced migration and invasion by 4T1.2 mammary adenocarcinoma cells. dBP4 inhibited IGF1-induced angiogenesis in vitro and in Matrigel implants in vivo. Direct intra-tumour injection of soluble dBP4 reduced angiogenesis in 4T1.2 luc mammary tumours tumour and reduced lung metastasis. CONCLUSION: A PAPP-A resistant IGFBP4, dBP4, inhibits angiogenesis and metastasis in 4T1.2 mammary fat pad tumours. This study highlights the therapeutic potential of dBP4 as an approach to block the tumour-promoting actions of IGF1.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Neovascularização Patológica/metabolismo , Proteína Plasmática A Associada à Gravidez/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Metástase Neoplásica , Fosforilação , Proteólise , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes
2.
Org Biomol Chem ; 14(39): 9278-9286, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27722734

RESUMO

Antimicrobial Peptides (AMPs) have unique anticancer properties, but their clinical application is currently limited by an inadequate margin of safety. A prodrug strategy associated with a combination therapy approach could address this limitation by increasing their therapeutic index and their efficacy. Accordingly, the first targeted anticancer polymeric prodrug candidates of AMPs, intended for combination therapy with another polymeric prodrug of an approved antineoplastic agent (doxorubicin), were synthesized as either a PEG-based dual-release prodrug or two individual pegylated prodrugs. The latter are based on a cathepsin B-labile peptide linker and an acid-sensitive acyl hydrazone bond for the AMP and doxorubicin prodrugs, respectively. Anticancer activities and toxicity differentials achieved with the free peptide and its polymer conjugates against ovarian, cancer and non-malignant, cells, indicate that protease-dependent reversible pegylation could be implemented to increase the therapeutic indices of AMPs in cancer therapy. The results obtained also show that this approach can be developed if the releasable PEG linker can be optimised to conciliate the attributes and restrictions of pegylation against proteases. In addition, combination of the polymeric prodrugs of the AMP and of doxorubicin provides additive antitumor effects which could be exploited to enhance the efficacy of the AMP candidate.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Pró-Fármacos/química , Peptídeos Catiônicos Antimicrobianos/química , Protocolos de Quimioterapia Combinada Antineoplásica/química , Catepsina B/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Polietilenoglicóis/química , Polímeros/química , Pró-Fármacos/síntese química , Pró-Fármacos/farmacologia
3.
Br J Cancer ; 106(3): 525-37, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22233923

RESUMO

BACKGROUND: Mammographic microcalcifications represent one of the most reliable features of nonpalpable breast cancer yet remain largely unexplored and poorly understood. METHODS: We report a novel model to investigate the in vitro mineralisation potential of a panel of mammary cell lines. Primary mammary tumours were produced by implanting tumourigenic cells into the mammary fat pads of female BALB/c mice. RESULTS: Hydroxyapatite (HA) was deposited only by the tumourigenic cell lines, indicating mineralisation potential may be associated with cell phenotype in this in vitro model. We propose a mechanism for mammary mineralisation, which suggests that the balance between enhancers and inhibitors of physiological mineralisation are disrupted. Inhibition of alkaline phosphatase and phosphate transport prevented mineralisation, demonstrating that mineralisation is an active cell-mediated process. Hydroxyapatite was found to enhance in vitro tumour cell migration, while calcium oxalate had no effect, highlighting potential consequences of calcium deposition. In addition, HA was also deposited in primary mammary tumours produced by implanting the tumourigenic cells into the mammary fat pads of female BALB/c mice. CONCLUSION: This work indicates that formation of mammary HA is a cell-specific regulated process, which creates an osteomimetic niche potentially enhancing breast tumour progression. Our findings point to the cells mineralisation potential and the microenvironment regulating it, as a significant feature of breast tumour development.


Assuntos
Adenocarcinoma/patologia , Neoplasias da Mama/patologia , Calcinose/patologia , Neoplasias Mamárias Experimentais/patologia , Fosfatase Alcalina/metabolismo , Animais , Carbonato de Cálcio/metabolismo , Oxalato de Cálcio/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Durapatita/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Fosfatos/metabolismo
4.
Gene Ther ; 16(3): 329-39, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19177133

RESUMO

The efficacy of current anti-cancer gene therapies is limited by the inability of gene vectors to penetrate the poorly vascularized, hypoxic regions of tumors, leaving these sites untreated. We describe a new approach for targeting gene therapy to these sites, which employs an attenuated strain of the non-pathogenic bacterium, Salmonella typhimurium, carrying an exogenous (that is, reporter or therapeutic) gene under the regulation of a new, highly hypoxia-inducible promoter (FF+20(*)). This bacterial vector was seen to rapidly migrate into, and thrive in, hypoxic areas of both mammary tumor spheroids grown in vitro and orthotopic mammary tumors after systemic injection. Using the reporter gene construct, FF+20(*)-lacZ, we show that bacterial expression of high levels of beta-galactosidase occurred only in hypoxic/necrotic sites of spheroids and tumors. We then replaced the reporter gene with one encoding a novel cytotoxic protein (HlyE) and showed that this was also expressed by bacteria only in hypoxic regions of murine mammary tumors. This resulted in a marked increase in tumor necrosis and reduced tumor growth. Our system represents a promising new strategy for delivering gene therapy to poorly vascularized regions of tumors and shows, for the first time, the efficacy of HlyE as an anti-tumor agent.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/farmacocinética , Neoplasias Mamárias Experimentais/terapia , Salmonella typhimurium/genética , Animais , Morte Celular , Hipóxia Celular/fisiologia , Técnicas de Cocultura , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Feminino , Marcação de Genes , Genes Reporter , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Necrose , Esferoides Celulares , Distribuição Tecidual , Células Tumorais Cultivadas
5.
Br J Cancer ; 101(2): 278-86, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19536088

RESUMO

BACKGROUND: Insulin-like growth factor 1 (IGF1) promotes breast cancer and disease progression. Bioavailability of IGF1 is modulated by IGF-binding proteins (IGFBPs). IGFBP4 inhibits IGF1 activity but cleavage by pregnancy-associated plasma protein-A (PAPP-A) protease releases active IGF1. METHODS: Expression of IGF pathway components and PAPP-A was assessed by western blot or RT-PCR. IGFBP4 (dBP4) resistant to PAPP-A cleavage, but retaining IGF-binding capacity, was used to block IGF activity in vivo. 4T1.2 mouse mammary adenocarcinoma cells transfected with empty vector, vector expressing wild-type IGFBP4 or vector expressing dBP4 were implanted in the mammary fat pad of BALB/c mice and tumour growth was assessed. Tumour angiogenesis and endothelial cell apoptosis were assessed by immunohistochemistry. RESULTS: 4T1.2 cells expressed the IGF1R receptor and IGFBP4. PAPP-A was expressed within mammary tumours but not by 4T1.2 cells. Proliferation and vascular endothelial growth factor (VEGF) production by 4T1.2 cells was increased by IGF1(E3R) (recombinant IGF1 resistant to binding by IGFBPs) but not by wild-type IGF1. IGF1-stimulated microvascular endothelial cell proliferation was blocked by recombinant IGFBP4. 4T1.2 tumours expressing dBP4 grew significantly more slowly than controls or tumours expressing wild-type IGFBP4. Inhibition of tumour growth by dBP4 was accompanied by the increased endothelial cell apoptosis. CONCLUSION: Protease-resistant IGFBP4 blocks IGF activity, tumour growth and angiogenesis.


Assuntos
Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Modelos Animais de Doenças , Humanos , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteína Plasmática A Associada à Gravidez/biossíntese , Proteína Plasmática A Associada à Gravidez/metabolismo , Receptor IGF Tipo 1/biossíntese , Proteínas Recombinantes/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese
6.
Physiol Meas ; 29(3): 375-88, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18367812

RESUMO

A fibre optic spectrophotometer front-end system for measuring corneas to overcome shortcomings associated with existing instruments was tested. The system allowed prompt measurement postmortem, minimizing beam pathlength to reduce the effects of scatter and unwanted refraction and eliminated optical interfaces and cuvette media. Rabbit corneas were excised immediately postmortem and placed on a detecting fibre optic coupled to an Ocean Optics spectrophotometer and illuminated by a deuterium-halogen source. The compact instrument with its small beam size allowed tissue profiling at test points across the corneal surface and efficient interchange for comparison of different tissues. This simplified system operation allowed rapid tissue altering to study induced changes on transmittance. The corneal transmittance data showed a consistent sharp cut-off at 320 nm in the ultraviolet radiation (UVR) spectrum, which decayed rapidly from postmortem swelling. Inter- and intra-corneal consistency was demonstrated by comparing data from different regions of the same cornea and those from opposite eyes. Changes to the spectra, particularly in the UVB below 300 nm, were evident when the corneal epithelium was removed, indicating that this layer is not the only corneal UVR filter. The new system reduced much of the variability associated with previous methods, as it rapidly measured corneal transmittance postmortem. Data are in broad agreement with published transmittance curves. The removal of the corneal epithelium revealed a substantial stromal contribution to the overall corneal UVR absorption, suggesting that corneas with pathologically or iatrogenically thinned stromas are less effective UVR blockers.


Assuntos
Córnea/fisiologia , Animais , Calibragem , Epitélio Corneano/fisiologia , Tecnologia de Fibra Óptica , Luz , Coelhos , Espectrofotometria Infravermelho , Espectrofotometria Ultravioleta , Raios Ultravioleta
7.
Gene ; 145(2): 227-30, 1994 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-8056336

RESUMO

A 1190-bp DNA fragment, designated X14, was isolated from a Bursaphelenchus xylophilus (isolate J10) genomic library. Used as a probe for DNA profiling, this fragment identifies the pinewood nematode (Bursaphelenchus spp.) at both the species and isolate level. The DNA profiles of a number of different species and isolates hybridised with the X14 probe, are presented. The nucleotide sequence of the 1190-bp probe was determined. Secondary structure analysis identified a large imperfect inverted repeat (IR) element within the probe which could form a large stem-loop structure in the RNA.


Assuntos
Nematoides/classificação , Nematoides/genética , Animais , Sequência de Bases , Sondas de DNA , Biblioteca Genômica , Dados de Sequência Molecular , Infecções por Nematoides/epidemiologia , Doenças das Plantas/parasitologia , Especificidade da Espécie , Árvores/parasitologia
8.
J Nematol ; 25(3): 406-15, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19279787

RESUMO

We have evaluated the potential of DNA-based methods to identify and differentiate Bursaphelenchus spp. and isolates. The isolation of a DNA probe, designated X14, and development of a DNA fingerprinting method for the identification and differentiation of Bursaphelenchus species and strains is described. Polymerase chain reaction (PCR) amplification of DNA isolated from Bursaphelenchus species using two primers derived from the sequence of the cloned repetitive DNA fragment X14 resulted in multiple band profiles. A 4-kb fragment thus amplified from B. xylophilus DNA was not amplified from B. mucronatus or B. fraudulentus DNA. In addition to this fragment, several other fragments are amplified from the three species. The banding patterns obtained allowed species identification and may have value in determining taxonomic affinities.

10.
Ir J Med Sci ; 178(2): 201-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19340516

RESUMO

BACKGROUND: Cyclo-oxygenase-2 (COX-2) is up-regulated in malignant tumours rendering it an attractive target for cancer therapeutics. However, whether long-term antagonism maintains its initial efficacy on established tumours is unclear. METHODS: 4T1 cells were injected into the mammary fat pad of BALB/c mice (n = 8). Once tumour deposits were established, animals were randomized into two equal groups to receive either a selective COX-2 inhibitor (SC-236) or a drug vehicle. Further animals similarly treated (n = 7) were studied in diuresis cages allowing urine capture and analysis by mass spectrometry to determine Prostaglandin F-1 levels (PGF-1). In addition, both wild-type receiving SC-236 and COX-2 knockout mice receiving either SC 236 or vehicle were subjected to the same studies to determine whether tumour-derived or host-derived (stromal) COX-2 was the critical element. Finally, BALB/c mice with 4T1 tumours (n = 7) were treated with a combination of COX-2 and lipoxygenase (LOX) inhibition to attenuate this escape phenomenon. RESULTS: While selective COX-2 inhibition initially retarded tumour growth, a rapid increase in tumour growth rate occurred later (day 9). This escape phenomenon correlated with an increase in urinary PGF-1 levels. An identical trend was also observed whether COX-2 knockout mice received SC-236 or not, suggesting that this effect is due to increased tumour-derived COX-2 production rather than recovery of host COX-2 functional capacity. Finally, dual inhibition of COX and LOX pathways attenuated this escape process. CONCLUSION: The anti-neoplastic effects of selective COX-2 inhibition may not be sustained as tumours demonstrate an escape capacity. However, this phenomenon maybe attenuated by a combination of COX/LOX inhibitors.


Assuntos
Neoplasias da Mama/enzimologia , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Ciclo-Oxigenase 2/metabolismo , Lipoxigenase/metabolismo , Animais , Ciclo-Oxigenase 2/efeitos dos fármacos , Progressão da Doença , Feminino , Lipoxigenase/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Distribuição Aleatória
11.
Ir J Med Sci ; 177(4): 359-65, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18855097

RESUMO

BACKGROUND: Endotoxin (LPS), a cell wall constituent of gram-negative bacteria, is a potent inflammatory stimulus. We demonstrated that laparotomy increases primary tumour growth and experimental lung metastases, implicating endotoxin as a causative factor. We hypothesised that the anti-endotoxin agent, rBPI(21) would block surgery-induced tumour growth. METHODS: Mammary adenocarcinoma cells were injected into female BALB/c mice to establish lung metastases. Mice were randomised into three groups receiving anaesthesia, laparotomy or laparotomy and rBPI(21) treatment on day 14. Animals were killed on day 19, lungs harvested and blood obtained. Number and size of lung metastases were recorded. Apoptosis, mitosis and microvessel density within metastases were assessed and VEGF measured. CONCLUSIONS: Laparotomy increased metastatic growth, decreased tumour cell apoptosis, increased tumour cell proliferation, increased microvessel density and circulating VEGF. LPS blockade by rBPI(21) attenuated this increased growth and decreased proliferation, increased apoptosis, decreased micro-vessel density and circulating VEGF. This suggests that rBPI(21), has clinical potential in attenuating surgery enhanced tumour growth, especially in patients with a history of cancer undergoing laparotomy.


Assuntos
Adenocarcinoma/terapia , Peptídeos Catiônicos Antimicrobianos/imunologia , Endotoxinas/antagonistas & inibidores , Inflamação/complicações , Laparotomia/efeitos adversos , Proteínas de Membrana/imunologia , Proteínas Recombinantes/uso terapêutico , Adenocarcinoma/etiologia , Adenocarcinoma/imunologia , Animais , Apoptose , Proteínas de Bactérias/imunologia , Proteínas Sanguíneas/imunologia , Técnicas de Cultura de Células , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Inflamação/fisiopatologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica/patologia , Distribuição Aleatória , Proteínas Recombinantes/imunologia , Aderências Teciduais/complicações , Fator A de Crescimento do Endotélio Vascular
12.
J Cell Mol Med ; 9(4): 777-94, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16364190

RESUMO

Vascular endothelial growth factor (VEGF) was originally identified as an endothelial cell specific growth factor stimulating angiogenesis and vascular permeability. Some family members, VEGF C and D, are specifically involved in lymphangiogenesis. It now appears that VEGF also has autocrine functions acting as a survival factor for tumour cells protecting them from stresses such as hypoxia, chemotherapy and radiotherapy. The mechanisms of action of VEGF are still being investigated with emerging insights into overlapping pathways and cross-talk between other receptors such as the neuropilins which were not previously associated with angiogenesis. VEGF plays an important role in embryonic development and angiogenesis during wound healing and menstrual cycle in the healthy adult. VEGF is also important in a number of both malignant and non-malignant pathologies. As it plays a limited role in normal human physiology, VEGF is an attractive therapeutic target in diseases where VEGF plays a key role. It was originally thought that in pathological conditions such as cancer, VEGF functioned solely as an angiogenic factor, stimulating new vessel formation and increasing vascular permeability. It has since emerged it plays a multifunctional role where it can also have autocrine pro-survival effects and contribute to tumour cell chemoresistance. In this review we discuss the established role of VEGF in angiogenesis and the underlying mechanisms. We discuss its role as a survival factor and mechanisms whereby angiogenesis inhibition improves efficacy of chemotherapy regimes. Finally, we discuss the therapeutic implications of targeting angiogenesis and VEGF receptors, particularly in cancer therapy.


Assuntos
Neovascularização Patológica , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Antineoplásicos/farmacologia , Artrite Reumatoide/metabolismo , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Diabetes Mellitus/metabolismo , Éxons , Regulação da Expressão Gênica , Humanos , Hipóxia , Ligantes , Modelos Biológicos , Neoplasias/metabolismo , Ligação Proteica , Isoformas de Proteínas , Psoríase/metabolismo , Reprodução , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
13.
Br J Cancer ; 92(2): 328-33, 2005 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-15655556

RESUMO

There is increasing evidence that vascular endothelial growth factor (VEGF) has autocrine as well as paracrine functions in tumour biology. Vascular endothelial growth factor-mediated cell survival signalling occurs via the classical tyrosine kinase receptors Flt-1, KDR/Flk-1 and the more novel neuropilin (NP) receptors, NP-1 and NP-2. A 24-mer peptide, which binds to neuropilin-1, induced apoptosis of murine and human breast carcinoma cells, whereas a peptide directed against KDR had no effect. Both anti-NP1 and anti-KDR peptides induced endothelial cell apoptosis. Confocal microscopy using 5-(6)-carboxyfluorescein-labelled peptides showed that anti-NP1 bound to both tumour and endothelial cells, whereas anti-KDR bound endothelial cells only. This study demonstrates that NP-1 plays an essential role in autocrine antiapoptotic signalling by VEGF in tumour cells and that NP1-blockade induces tumour cell and endothelial cell apoptosis. Specific peptides can therefore be used to target both autocrine (tumour cells) and paracrine (endothelial cells) signalling by VEGF.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias da Mama/metabolismo , Neuropilina-1/metabolismo , Peptídeos/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Adenocarcinoma/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Citometria de Fluxo , Humanos , Camundongos , Microscopia Confocal , Neuropilina-1/imunologia , Peptídeos/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
J Surg Res ; 69(1): 145-9, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9202661

RESUMO

Interleukin-6 is associated with poor prognosis in breast cancer. Expression of GP96, a glucose regulated stress protein, is related to drug resistance in tumor cells. Interleukin-6 has previously been shown to induce GP96 expression in a murine myeloblastic cell line. BT474 or MDA-MB231 cells were incubated with recombinant Interleukin-6 (100 to 750 U/ml) for 24 hr. To establish a time course for GP96 induction, MDA-MB231 cells were incubated with 250 U/ml recombinant interleukin-6 for 0-48 hr. Following incubation, cells were washed twice in phosphate-buffered saline (PBS) and cell lysates were prepared by adding 100 microliters of PBS and freezing at -20 degrees C. GP96 was assessed by immunoblotting. Breast tumor tissue and histologically normal breast tissue were obtained within 1 hr of resection and flash frozen in liquid nitrogen. Tissue was homogenized in ice-cold PBS and cell debris was pelleted by centrifugation at 300g at 4 degrees C for 5 min. Supernatants were collected and assayed for interleukin-6 by ELISA, and GP96 by immunoblotting. Both interleukin-6 (P < 0.001) and GP96 are elevated in breast tumor tissue compared to histologically normal tissue. Interleukin-6 (> or = 250 U/ml for > or = 12 hr) induces GP96 in the metastatic breast cancer cell line, MDA-MB231, but has no effect on GP96 levels in the primary cell line, BT474. Elevated interleukin-6 in breast tumors may induce GP96 expression in tumor cells conferring a survival advantage by rendering them resistant to cytotoxic therapy and other forms of stress.


Assuntos
Neoplasias da Mama/metabolismo , Interleucina-6/farmacologia , Glicoproteínas de Membrana/metabolismo , Mama/metabolismo , Neoplasias da Mama/secundário , Feminino , Humanos , Interleucina-6/metabolismo , Proteínas Recombinantes , Valores de Referência , Células Tumorais Cultivadas
15.
Biochem Biophys Res Commun ; 210(3): 753-9, 1995 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-7763249

RESUMO

A major component of the infectious particle causing spongiform encephalopathies or prion diseases is an aberrant isoform (PrPSc) of a glycosyl-phosphatidylinositol (GPI)-anchored cell surface protein, PrPC. The cellular processes involved in the formation of PrPSc are unclear but involve the internalization of PrPC prior to conversion. Here, we demonstrate that PrPC is associated with caveolin, a structural protein component of caveolae. We show that PrPC and caveolin share similar detergent characteristics and copurify in linear sucrose gradients. PrPC was protected from proteinase K digestion in the caveolin fraction but solubilizing the caveolae prior to proteinase K digestion rendered PrPC susceptible to proteinase K digestion. Our results indicate a physical association between PrPC and caveolin in N2a cells. The implication of these results in relation to prion biogenesis is discussed.


Assuntos
Caveolinas , Proteínas de Membrana/análise , Neuroblastoma/ultraestrutura , Proteínas PrPC/análise , Animais , Caveolina 1 , Linhagem Celular , Membrana Celular/ultraestrutura , Centrifugação com Gradiente de Concentração , Detergentes , Imunofluorescência , Glicosilfosfatidilinositóis/análise , Proteínas de Membrana/isolamento & purificação , Camundongos , Proteínas PrPC/isolamento & purificação , Solubilidade , Células Tumorais Cultivadas
16.
Br J Surg ; 88(8): 1105-9, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11488797

RESUMO

BACKGROUND: Angiogenesis is critical for tumour growth and metastasis. The switch to the angiogenic phenotype depends on the net balance between positive and negative angiogenic factors released by the tumour. It was hypothesized that patients with oesophageal cancer would express raised serum levels of vascular endothelial growth factor (VEGF) which would return to normal values with neoadjuvant chemoradiotherapy. METHODS: Forty-four patients with oesophageal cancer who were selected for treatment with neoadjuvant chemoradiotherapy had blood samples taken before treatment, during chemoradiotherapy, before operation, on days 1, 3 and 5 after surgery, and 3 months after resection. Serum levels of VEGF were measured. Values were correlated with response to treatment. Controls were patients who were undergoing surgery for non-malignant conditions. RESULTS: Serum VEGF levels were raised in patients with oesophageal cancer compared with age-matched controls (mean 247 versus 1157 pg/ml; P < 0.01). VEGF levels were unaffected by neoadjuvant treatment but fell significantly on the first day after operation (652 versus 1057 pg/ml before operation; P < 0.05). No decrease occurred in control patients. VEGF levels had returned to preoperative levels by day 5. A similar postoperative rise in VEGF levels was seen in the control subjects (1194 pg/ml on day 5 versus 71 pg/ml before operation; P = 0.001). There was no correlation between VEGF level and response to treatment or tumour stage. VEGF levels had decreased significantly at 3 months following tumour resection (594 versus 1558 pg/ml on day 5; P = 0.03). CONCLUSION: VEGF levels are raised in patients with oesophageal cancer and are unaltered by neoadjuvant treatment, suggesting an additional source other than tumour cells for this proangiogenic agent.


Assuntos
Adenocarcinoma/sangue , Carcinoma de Células Escamosas/sangue , Fatores de Crescimento Endotelial/sangue , Neoplasias Esofágicas/sangue , Linfocinas/sangue , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/terapia , Quimioterapia Adjuvante/métodos , Terapia Combinada/métodos , Neoplasias Esofágicas/irrigação sanguínea , Neoplasias Esofágicas/terapia , Feminino , Humanos , Masculino , Neovascularização Patológica , Cuidados Pré-Operatórios/métodos , Radioterapia Adjuvante/métodos , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
17.
Ann Surg Oncol ; 5(3): 271-8, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9607631

RESUMO

BACKGROUND: Breast tumors contain high numbers of infiltrating macrophages. The role and function of these cells within the tumor remain unclear, but a number of studies have found an association between poor prognosis and macrophage content in human breast cancer. Both hypoxia and TGFbeta-1 have been shown to regulate VEGF in other cell types. We hypothesized that breast tumor-associated macrophages produce VEGF and that macrophage production of this factor is regulated by both hypoxia and TGFbeta-1. METHODS: Paraffin-embedded breast tumor sections were stained immunohistochemically with anti-VEGF, anti-CD68, and anti-cytokeratin. Monocytes were matured for 3 days in 20% autologous plasma and activated with 1000 U/mL interferon-gamma for 24 hours. Supernatants were assayed for VEGF protein by ELISA. Total RNA was isolated from cells and reverse transcribed to cDNA, which was used as a template in PCR reactions for VEGF and beta-actin. RESULTS: Both tumor cells and tumor macrophages produce VEGF in human breast tumors. Hypoxia increases VEGF protein and mRNA levels in monocyte-derived macrophages, whereas TGFbeta-1 increases VEGF protein but not mRNA under hypoxic growth conditions. CONCLUSIONS: Breast tumor-associated macrophages may contribute to the angiogenic activity of human breast tumors by producing VEGF. Macrophage production of VEGF is upregulated by hypoxia and TGFbeta-1, both of which occur in the tumor environment. Macrophage production of VEGF is regulated at both the mRNA and protein levels.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Hipóxia Celular/fisiologia , Fatores de Crescimento Endotelial/metabolismo , Linfocinas/metabolismo , Macrófagos/metabolismo , RNA Mensageiro/genética , RNA Neoplásico/genética , Fator de Crescimento Transformador beta/fisiologia , Neoplasias da Mama/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Monócitos , Reação em Cadeia da Polimerase , Prognóstico , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Br J Cancer ; 91(2): 359-65, 2004 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-15213717

RESUMO

Cyclooxygenase-2 (COX-2) expression is increased in breast cancer and surgery has been shown to increase the growth of metastatic tumours. We investigated the effect of selective COX-2 inhibition on the growth of metastases in either an experimental metastasis model or following excision of a murine primary breast tumour. 50,000 4T1 mammary carcinoma cells were injected into the mammary fat pad of female BALB/c mice. When the mean TD reached 8+/-0.4 mm, tumours were excised and the mice were randomised into two groups (n=12 per group) to receive daily intraperitoneal injections of the selective COX-2 inhibitor, SC-236 or drug vehicle for 14 days. Alternatively, experimental metastases were established by tail-vein injection of 50,000 4T1 cells. Mice received either the selective COX-2 inhibitor, SC-236 or drug vehicle for 14 days (n=12 per group). SC-236 treatment significantly reduced tumour burden, the number and size of spontaneous metastases following primary tumour excision. SC-236 treatment also reduced tumour burden, the number and size of experimental metastases. Immunohistochemical staining demonstrated that COX-2 inhibition reduced microvessel density and increased apoptosis within both spontaneous and experimental metastases. These data clearly demonstrate that the selective COX-2 inhibitor, SC-236, has potent antimetastatic activity against both spontaneous metastases arising following primary tumour excision and experimental metastases.


Assuntos
Adenocarcinoma/tratamento farmacológico , Apoptose/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/uso terapêutico , Isoenzimas/antagonistas & inibidores , Neoplasias Mamárias Experimentais/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Pirazóis/uso terapêutico , Sulfonamidas/uso terapêutico , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/secundário , Animais , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Feminino , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/prevenção & controle , Prostaglandina-Endoperóxido Sintases
19.
Ann Surg Oncol ; 4(8): 621-7, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9416408

RESUMO

BACKGROUND: Angiogenesis is essential for tumor growth and metastasis. Vascular endothelial growth factor (VEGF) is the most potent angiogenic factor identified to date. TGF beta-1 acts as an indirect angiogenic agent. METHODS: VEGF and TGF beta-1 were measured in the serum of breast cancer patients and age-matched controls and in tumor tissue of cancer patients by ELISA. VEGF protein and mRNA expression by breast tumor cell lines were examined, and the effect of TGF beta-1 on VEGF production in these cells was assessed. RESULTS: VEGF levels were significantly higher (P = .03) in the serum of patients with breast cancer compared to age-matched controls. A positive correlation was found between serum (r = 0.539) and tumor tissue (r = 0.688) levels of VEGF and TGF beta-1. Metastatic MDA-MB-231 breast cancer cells produce more VEGF than do the primary BT474 cells. TGF beta-1 significantly (P < .05) increased production of VEGF. CONCLUSIONS: Breast cancer cells constitutively produce VEGF protein and mRNA. There is a relationship between VEGF and TGF beta-1 levels in breast cancer patients, and TGF beta-1 regulates VEGF expression by breast cancer cells.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Fatores de Crescimento Endotelial/metabolismo , Linfocinas/metabolismo , Neovascularização Patológica/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Análise de Variância , Estudos de Casos e Controles , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Br J Cancer ; 85(2): 273-8, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11461089

RESUMO

Tumour progression is regulated by the balance of proliferation and apoptosis in the tumour cell population. To date, the role of vascular endothelial growth factor (VEGF) in tumour growth has been attributed to the induction of angiogenesis. VEGF has been shown to be a survival factor for endothelial cells, preventing apoptosis by inducing Bcl-2 expression. In both murine (4T1) and human (MDA-MB-231) metastatic mammary carcinoma cell lines, we found that VEGF upregulated Bcl-2 expression and anti-VEGF antibodies reduced Bcl-2 expression. These alterations in Bcl-2 expression were reflected by the levels of tumour cell apoptosis. VEGF resulted in reduced tumour cell apoptosis, whereas its inhibition with anti-VEGF neutralizing antibodies induced apoptosis directly in tumour cells. Therefore, in addition to its role in angiogenesis and vessel permeability, VEGF acts as a survival factor for tumour cells, inducing Bcl-2 expression and inhibiting tumour cell apoptosis.


Assuntos
Adenocarcinoma/patologia , Apoptose/fisiologia , Neoplasias da Mama/patologia , Fatores de Crescimento Endotelial/fisiologia , Linfocinas/fisiologia , Neoplasias Mamárias Experimentais/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Regulação para Cima/fisiologia , Animais , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Células Tumorais Cultivadas , Regulação para Cima/imunologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA