Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Gene Med ; 12(9): 755-65, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20821746

RESUMO

BACKGROUND: Using the tumour type specific human osteocalcin (hOC) promoter, we have previously reported strong promoter activation in hormone independent prostate cancer cells in vitro. In the present study, we present a comparative study of the tissue specific promoter prostate specific membrane antigen (PSMA), and the tumour-type specific hOC promoter driving the inducible nitric oxide synthase (iNOS) transgene using both in vitro and in vivo models. METHODS: In vitro cytotoxicity was assessed by clonogenic assay. Quantification of nitric oxide expression was determined by the Griess test. In vivo anti-tumour efficacy was determined by tumour growth delay following direct intra-tumoural injection of the constructs into PC3 xenografts. In addition, tumours were dissected post mortem and examined for morphological differences as well as changes in apoptotic protein expression. RESULTS: PSMA/iNOS produced cytotoxicity in both androgen dependant and independent cell lines. Nitric oxide quantification confirmed that increased cytotoxicity was directly associated with nitric oxide production. Tumour growth delays were observed in all groups treated with the iNOS-expressing constructs ranging from 10.7 days for the hOC/iNOS single dose treatment group to a maximum of 52.2 days for the hOC/iNOS multiple dose group. Intra-tumoural assessment of iNOS and cleaved poly (ADP-ribose) polymerase protein expression demonstrated a significant up-regulation of both proteins, indicating cytotoxicity mediated through the intrinsic apoptotic pathway. CONCLUSIONS: Highly significant tumour growth delay coupled with no detrimental side-effects were observed following treatment with the PSMA/iNOS and hOC/iNOS constructs. We consider that these findings provide a basis for the development of systemically delivered PSMA/iNOS or hOC/iNOS targeting early stage and advanced prostate cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Regulação da Expressão Gênica/fisiologia , Terapia Genética , Neoplasias Hormônio-Dependentes/terapia , Óxido Nítrico Sintase Tipo II/genética , Neoplasias da Próstata/terapia , Animais , Antígenos de Superfície/genética , Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Feminino , Glutamato Carboxipeptidase II/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Estadiamento de Neoplasias , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Osteocalcina/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transgenes/fisiologia , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Gene Med ; 11(2): 160-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19062185

RESUMO

BACKGROUND: Nitric oxide (NO.) derived from donor drugs has been shown to be an effective chemosensitizer in vitro. We investigated the combination of inducible nitric oxide synthase (iNOS) gene transfer, driven by a strong constitutive promoter (cytomegalovirus; CMV) with the DNA cross-linking agent cisplatin in mouse and human tumour cell lines. METHODS: Proof of principal experiments were performed in the radiation-induced fibrosarcoma-1 (RIF-1) murine cell line. Cells were transfected with constitutively expressed CMV/iNOS plasmid DNA using a cationic lipid vector, before exposure to cisplatin. In vivo efficacy was determined in an intradermal RIF-1 tumour model, with intraperitoneal administration of cisplatin. Additionally, treatment potential was investigated in various human tumour cell lines including human prostate (DU145 and PC3) and human colon (HT29 and HCT116) cancer cell lines. Experimental endpoints were established using western blot, Greiss test, clonogenic assay and tumour growth delay. RESULTS: Transfection of RIF-1 tumour cells in vitro with the CMV/iNOS significantly enhanced the cytotoxicity of cisplatin (0.2-1.0 microM). In vivo transfer of CMV/iNOS by direct injection into established RIF-1 tumours caused a significant (p = 0.0027) delay in tumour growth. CMV/iNOS gene transfer in vitro resulted in the strong expression of iNOS DNA in all cell lines, and significantly increased levels of NO. in all cell lines except HCT116. CONCLUSIONS: Significant chemosensitization of cisplatin cytotoxicity was observed in the presence of NO. derived from the overexpression iNOS. We conclude that p53 status of the various cell lines was unlikely to be responsible for cisplatin-induced apoptosis.


Assuntos
Antineoplásicos/toxicidade , Cisplatino/toxicidade , Óxido Nítrico Sintase/genética , Animais , Apoptose , Linhagem Celular Tumoral , Terapia Combinada , Terapia Genética , Vetores Genéticos/genética , Humanos , Camundongos , Neoplasias Induzidas por Radiação/terapia , Transdução Genética , Transfecção
3.
Mol Carcinog ; 48(2): 110-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18618592

RESUMO

Cytochrome P450 1B1 (CYP1B1) mRNA is constitutively expressed in most normal extra-hepatic tissues; however the protein is not detectable in these tissues but is expressed in a wide variety of tumors. CYP1B1 is responsible for the activation of a number of carcinogens present in tobacco smoke and food. A surgical model of rat esophageal tumorigenesis, promoted by gastric or duodenal reflux was used to determine CYP1B1 expression in premalignant esophageal tissue. Immunohistochemistry was performed using a modified amplified fluorescein tyramide protocol. CYP1B1 was not observed in normal esophageal mucosa, submucosa, or muscularis mucosa. Animals exposed to gastric reflux developed mild hyperplasia. Varying degrees of hyperplasia were observed in the duodenal reflux group. All regions of hyperplasia showed moderate or strong CYP1B1 immunoreactivity. Duodenal reflux induced a small number of premalignant changes: immunoreactivity was absent from the epithelium of squamous dysplasia (0/10), Barrett's esophagus (0/7), and majority of dysplastic Barrett's esophagus (1/4). Moderate or strong immunoreactivity was observed in the majority (7/8) of squamous cell carcinomas (SCCs) in situ. Immunoreactivity was also observed in the lamina propria and submucosa in association with inflammation, regardless of the severity of inflammation. The expression of CYP1B1 in hyperplasia, SCCs in situ, or in association with inflammation may increase the production of carcinogenic metabolites, which may promote esophageal tumorigenesis.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Refluxo Duodenogástrico/complicações , Neoplasias Esofágicas/enzimologia , Refluxo Gastroesofágico/complicações , Animais , Especificidade de Anticorpos , Hidrocarboneto de Aril Hidroxilases/imunologia , Western Blotting , Citocromo P-450 CYP1B1 , Refluxo Duodenogástrico/enzimologia , Neoplasias Esofágicas/etiologia , Feminino , Refluxo Gastroesofágico/enzimologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
4.
Clin Cancer Res ; 14(5): 1502-9, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18316575

RESUMO

PURPOSE: A number of cytotoxic chemotherapy agents tested at low concentrations show antiangiogenic properties with limited cytotoxicity, e.g., cyclophosphamide, tirapazamine, and mitoxantrone. AQ4N is a bioreductive alkylaminoanthraquinone that is cytotoxic when reduced to AQ4; hence, it can be used to target hypoxic tumor cells. AQ4N is structurally similar to mitoxantrone and was evaluated for antiangiogenic properties without the need for bioreduction. EXPERIMENTAL DESIGN: The effect of AQ4N and fumagillin on human microvascular endothelial cells (HMEC-1) was measured using a variety of in vitro assays, i.e., 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, wound scrape, tubule formation, rat aortic ring, and invasion assays. Low-dose AQ4N (20 mg/kg) was also given in vivo to mice bearing a tumor in a dorsal skin flap. RESULTS: AQ4N (10(-11) to 10(-5) mol/L) had no effect on HMEC-1 viability. AQ4N (10(-9) to 10(-5)mol/L) caused a sigmoidal dose-dependent inhibition of endothelial cell migration in the wound scrape model. Fumagillin showed a similar response over a lower dose range (10(-13) to 10(-9) mol/L); however, the maximal inhibition was less (25% versus 43% for AQ4N). AQ4N inhibited HMEC-1 cell contacts on Matrigel (10(-8) to 10(-5) mol/L), HMEC-1 cell invasion, and sprouting in rat aorta explants. Immunofluorescence staining with tubulin, vimentim, dynein, and phalloidin revealed that AQ4N caused disruption to the cell cytoskeleton. When AQ4N (20 mg/kg) was given in vivo for 5 days, microvessels disappeared in LNCaP tumors grown in a dorsal skin flap. CONCLUSIONS: This combination of assays has shown that AQ4N possesses antiangiogenic effects in normoxic conditions, which could potentially contribute to antitumor activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Antraquinonas/farmacologia , Endotélio Vascular/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Pele/irrigação sanguínea , Animais , Antineoplásicos/farmacologia , Aorta/efeitos dos fármacos , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Neoplasias do Colo/tratamento farmacológico , Cicloexanos/farmacologia , Citoesqueleto/metabolismo , Combinação de Medicamentos , Ácidos Graxos Insaturados/farmacologia , Humanos , Laminina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microcirculação , Neovascularização Fisiológica , Neoplasias da Próstata/tratamento farmacológico , Proteoglicanas/metabolismo , Ratos , Ratos Wistar , Sesquiterpenos/farmacologia , Retalhos Cirúrgicos , Cicatrização/efeitos dos fármacos
5.
Endocrinology ; 149(11): 5724-34, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18669603

RESUMO

FKBP-like (FKBPL) protein is a novel immunophilin-like protein that plays a role in the cellular stress response. Its three tetratricopeptide repeat motifs are homologous to the heat shock protein 90 interaction sites of other immunophilins that have roles in steroid hormone receptor signaling. In this study, using biomolecular complementation and coimmunoprecipitation techniques, we show that FKBPL also colocalizes and interacts with the components of the heat shock protein 90-glucocorticoid receptor (GR) complex and demonstrate that the PPIase domain of FKBPL is important for the interaction between this complex and the dynein motor protein, dynamitin. Treatment of DU145 cells with the GR ligand, dexamethasone, induced a rapid and coordinated translocation of both GR and FKBPL to the nucleus; this response was perturbed when FKBPL was knocked down with a targeted small interfering RNA. Furthermore, overexpression of FKBPL increased GR protein levels and transactivation of a luciferase reporter gene in response to dexamethasone in DU145 cells. However, these responses were cell line dependent. In summary, these data suggest that FKBPL can be classed as a new member of the FKBP protein family with a role in steroid receptor complexes and signaling.


Assuntos
Imunofilinas/metabolismo , Imunofilinas/fisiologia , Receptores de Glucocorticoides/metabolismo , Receptores de Esteroides/fisiologia , Núcleo Celular/metabolismo , Humanos , Imunofilinas/antagonistas & inibidores , Imunofilinas/genética , Complexos Multiproteicos/metabolismo , Ligação Proteica , Transporte Proteico , RNA Interferente Pequeno/farmacologia , Receptores de Esteroides/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Ligação a Tacrolimo , Ativação Transcricional/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
6.
Oncol Res ; 17(3): 93-101, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18669161

RESUMO

The contribution of endothelial cell growth to angiogenesis has been widely studied; however, the involvement of pericytes is less well documented, especially in human tumors. In this study we aimed to quantify and assess the prognostic significance of pericyte coverage, the extent of hypoxia, and microvessel density (MVD) in normal bladder mucosa and urothelial carcinoma. Antibody to alpha-smooth muscle actin was used to assess the distribution of pericytes (mural/smooth muscle cells) in the microvessels of normal human bladder (n = 4) mucosa and in urothelial carcinoma (n = 47) samples; this was quantitated using microvessel pericyte index (MPI). The MVD was measured using two different methods (n = 47) and hypoxia was assessed using glucose transporter-1 (Glut-1) staining (n = 30). There was a 70% reduction in MPI in urothelial carcinomas compared to normal bladder mucosa (p < 0.0012); MPI did not correlate with tumor stage or grade. Ta and T1 superficial tumors were divided into two groups with a MPI of <15% or >15%. Progression-free survival was significantly shorter for tumors with MPI >15% (p = 0.0036). MVD had no prognostic value using either evaluation method. Glut-1 immunoreactivity was not prognostic in superficial urothelial carcinoma samples. Tumors with a higher MPI showed a greater Glut-1 immunoreactivity (p = 0.0051). Microvessels in urothelial carcinoma have a considerable loss of pericyte coverage compared to normal bladder mucosa. The data from this preliminary study indicate that progression-free survival was shorter in patients whose superficial tumors had higher pericyte coverage of the microvessels. This may be due to increased levels of hypoxia, as demonstrated by a significant increase in Glut-1 staining.


Assuntos
Carcinoma/patologia , Neovascularização Patológica/patologia , Pericitos/patologia , Neoplasias da Bexiga Urinária/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Permeabilidade Capilar , Carcinoma/irrigação sanguínea , Carcinoma/fisiopatologia , Contagem de Células , Feminino , Transportador de Glucose Tipo 1/análise , Humanos , Hipóxia/etiologia , Hipóxia/patologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/irrigação sanguínea , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/fisiopatologia , Estadiamento de Neoplasias , Neovascularização Patológica/fisiopatologia , Prognóstico , Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/fisiopatologia
7.
J Pharm Pharmacol ; 60(8): 999-1017, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18644193

RESUMO

Nitric oxide (NO(.)) is a reactive nitrogen radical produced by the NO synthase (NOS) enzymes; it affects a plethora of downstream physiological and pathological processes. The past two decades have seen an explosion in the understanding of the role of NO(.) biology, highlighting various protective and damaging modes of action. Much of the controversy surrounding the role of NO(.) relates to the differing concentrations generated by the three isoforms of NOS. Both calcium-dependent isoforms of the enzyme (endothelial and neuronal NOS) generate low-nanomolar/picomolar concentrations of NO(.). By contrast, the calcium-independent isoform (inducible NOS (iNOS)) generates high concentrations of NO(.), 2-3 orders of magnitude greater. This review summarizes the current literature in relation to iNOS gene therapy for the therapeutic benefit of various pathological conditions, including various states of vascular disease, wound healing, erectile dysfunction, renal dysfunction and oncology. The available data provide convincing evidence that manipulation of endogenous NO(.) using iNOS gene therapy can provide the basis for future clinical trials.


Assuntos
Terapia Genética/métodos , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Animais , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Disfunção Erétil/enzimologia , Disfunção Erétil/genética , Disfunção Erétil/terapia , Feminino , Técnicas de Transferência de Genes , Humanos , Nefropatias/enzimologia , Nefropatias/genética , Nefropatias/terapia , Masculino , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/terapia , Óxido Nítrico Sintase Tipo II/genética , Resultado do Tratamento , Cicatrização/genética
8.
Front Biosci ; 12: 3406-18, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17485309

RESUMO

Reactive nitrogen species play important roles in cell signalling, but when present at high concentrations they can subject cells to nitrosative stress, which may lead to cell death. Nitric oxide (NOx) is now recognized as playing important roles in cancer aetiology and progression and it can influence the outcome of cancer treatment. It is synthesised by the action of nitric oxide synthases (NOSs) on the amino acid arginine. Although NOx is not highly reactive with biological molecules, it reacts readily with other oxygen radicals to generate highly damaging reactive nitrogen species such as peroxynitrite, nitrogen dioxide and dinitrogen trioxide. These are potent inducers of apoptosis and necrosis. They may also inhibit DNA repair mechanisms, leading to mutation and carcinogenesis. Both inhibition and over-production of NOx have been investigated as strategies for cancer therapy. There is clear evidence that administration of competitive inhibitors of NOS can significantly slow the growth of solid tumors in rodent models, probably by reducing blood flow, and this creates a hypoxic environment that is conducive to the activation of bioreductive anticancer agents. Alternatively, generation of NOx concentrations in the high micromolar range by NOx donor drugs or gene therapy with inducible NOS is directly cytotoxic to cells and has been shown to inhibit tumor growth. At these high concentrations NOx is also an excellent sensitizer to radiation and to some chemotherapeutic agents, particularly cisplatin. Thus, manipulation of NOx levels in tumors offers exciting opportunities to improve the effectiveness of cancer treatment.


Assuntos
Neoplasias/terapia , Nitrosação , Radicais Livres , Humanos , Neoplasias/fisiopatologia , Óxido Nítrico/biossíntese
9.
Peptides ; 28(4): 781-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17321638

RESUMO

Multiple bradykinin-related peptides including a novel bradykinin structural variant, (Val(1))-bradykinin, have been identified from the defensive skin secretion of Guenther's frog, Hylarana guentheri by a tandem mass spectrometry method. Subsequently, four different preprobradykinin cDNAs, which encoded multiple bradykinin copies and its structural variants, were consistently cloned from a skin derived cDNA library. These preprobradykinin cDNAs showed little structural similarity with mammalian kininogens and the kininogens from the skin of toads, but have regions that are highly conserved in the kininogens from another ranid frog, Odorrana schmackeri. Alignment of these preprobradykinins revealed that preprobradykinin 1, 2 and 3 may derive from a single gene by alternative exon splicing.


Assuntos
Anuros/metabolismo , Bradicinina/isolamento & purificação , Peptídeos/isolamento & purificação , Pele/metabolismo , Sequência de Aminoácidos , Proteínas de Anfíbios/genética , Proteínas de Anfíbios/isolamento & purificação , Proteínas de Anfíbios/farmacologia , Animais , Anuros/genética , Sequência de Bases , Bradicinina/genética , Bradicinina/farmacologia , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Relação Dose-Resposta a Droga , Técnicas In Vitro , Cininogênios/química , Masculino , Dados de Sequência Molecular , Músculo Liso/efeitos dos fármacos , Peptídeos/genética , Peptídeos/farmacologia , Precursores de Proteínas/genética , Precursores de Proteínas/isolamento & purificação , Ratos , Ratos Wistar , Análise de Sequência de DNA , Espectrometria de Massas em Tandem
10.
Oncol Res ; 16(12): 569-74, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18351131

RESUMO

Nicotinamide is a potent radiosensitizer currently employed in the treatment of cancer of the bladder, head, and neck. Unfortunately, nicotinamide is also a potent emetic at the concentrations required for radiosensitization. Previously, we have demonstrated that nicotinamide-induced emesis is the direct result of decreased spontaneous peristaltic activity in the ileum. However, the effect of nicotinamide's metabolites on peristaltic activity have not been investigated, although several studies have unsuccessfully attempted to correlate the degree of emesis with the levels of the metabolites in plasma. Isolated rat ileum rings and rat tail arteries were perfused with oxygenated Krebs solution in an organ bath. Nicotinamide, 1-methynicotinamide, or N-oxide nicotinamide were introduced to the perfusate and changes in amplitude of spontaneous peristaltic activity or phenylephrine-induced vasoconstriction recorded. Nicotinamide inhibited peristalsis in the ileum and agonist-induced vasoconstriction in the rat tail arteries, as previously observed. However, the primary metabolites of nicotinamide were without effect. Gut smooth muscle and rat tail artery are sensitive to the relaxant effects of nicotinamide. The primary metabolites of nicotinamide are not vasoactive and do not blunt either spontaneous or agonist-induced contraction and are thus unlikely to contribute to the degree of emesis observed following nicotinamide administration.


Assuntos
Niacinamida/efeitos adversos , Radiossensibilizantes/efeitos adversos , Vômito/induzido quimicamente , Animais , Artérias/efeitos dos fármacos , Artérias/metabolismo , Íleo/efeitos dos fármacos , Íleo/metabolismo , Soluções Isotônicas , Masculino , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Peristaltismo/efeitos dos fármacos , Radiossensibilizantes/metabolismo , Ratos , Ratos Wistar , Cauda/irrigação sanguínea , Vasoconstrição/efeitos dos fármacos
11.
Cardiovasc Res ; 69(1): 207-17, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16165110

RESUMO

OBJECTIVE: Pericytes, located in close proximity to the underlying endothelium, form an integral component of the microvasculature. These cells are intimately involved in angiogenesis, which is of fundamental importance in many physiological and pathological processes. We evaluated the influence of pericyte-conditioned medium (PCM) on endothelial cell growth characteristics and modulation of endothelial gene expression. METHODS: Migration and tubule formation assays were performed in vitro to determine the effect of PCM on endothelial growth characteristics. cDNA microarray analysis was used to identify alterations in gene expression following exposure of human microvascular endothelial cells (HMEC-1) to PCM. Overexpression of PAI-1 using recombinant protein or transient transfection, and inhibition using an inhibitory antibody against PAI-1, were used to determine whether up- or down-regulation of this gene was responsible for the changes in endothelial cell characteristics observed in response to PCM exposure. RESULTS: We have shown that PCM exerts a dramatic inhibitory influence on endothelial cell migration in vitro. In addition, endothelial cells cultured on Matrigel and exposed to PCM were found to generate significantly fewer angiogenic branches. Microarray analysis of endothelial cells exposed to PCM identified PAI-1 as the gene showing the greatest level of differential expression (3.4-fold induction). Studies using an inhibitory antibody to PAI-1 suggest that induction of this protein by PCM is pivotal to the observed inhibitory influence on the migratory and angiogenic potential of HMEC-1. We further investigated this by overexpressing PAI-1, which was shown to have a potent inhibitory influence on EC migration and angiogenic branching, although the concentration of PAI-1 was clearly important. CONCLUSION: Collectively, these findings suggest that PCM contains a bioactive element(s) that controls both endothelial cell migration and tubule formation in vitro and that these responses may be partially controlled by increased endothelial cell expression of PAI-1.


Assuntos
Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Neovascularização Fisiológica , Pericitos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Northern Blotting/métodos , Western Blotting/métodos , Movimento Celular , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Inibidor 1 de Ativador de Plasminogênio/metabolismo
12.
Sci Rep ; 6: 19442, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26787230

RESUMO

Gold nanoparticle radiosensitization represents a novel technique in enhancement of ionising radiation dose and its effect on biological systems. Variation between theoretical predictions and experimental measurement is significant enough that the mechanism leading to an increase in cell killing and DNA damage is still not clear. We present the first experimental results that take into account both the measured biodistribution of gold nanoparticles at the cellular level and the range of the product electrons responsible for energy deposition. Combining synchrotron-generated monoenergetic X-rays, intracellular gold particle imaging and DNA damage assays, has enabled a DNA damage model to be generated that includes the production of intermediate electrons. We can therefore show for the first time good agreement between the prediction of biological outcomes from both the Local Effect Model and a DNA damage model with experimentally observed cell killing and DNA damage induction via the combination of X-rays and GNPs. However, the requirement of two distinct models as indicated by this mechanistic study, one for short-term DNA damage and another for cell survival, indicates that, at least for nanoparticle enhancement, it is not safe to equate the lethal lesions invoked in the local effect model with DNA damage events.


Assuntos
Ouro , Nanopartículas Metálicas , Imagem Molecular , Radiação , Linhagem Celular Tumoral , Células Cultivadas , Dano ao DNA/efeitos da radiação , Humanos , Microscopia Eletrônica de Transmissão por Filtração de Energia , Método de Monte Carlo , Doses de Radiação
13.
Oncol Res ; 15(2): 81-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16119005

RESUMO

Pentoxifylline (PTX) is currently used therapeutically as a tumor oxygenator where it been shown to increase tumor blood flow and potentiate ionizing radiation damage. The clinical benefits of PTX have been primarily attributed to its effect on the rheologic properties of whole blood, although there is speculation that the mechanism for PTX-induced increases in tumor oxygenation may be the direct result of reduced vascular resistance. Therefore, to address the issue of vascular (geometric) resistance directly, we examined the ability of PTX and its hydroxy metabolite, lisofylline (LF), to modulate phenylephrine (PE)-induced constriction in isolated rat tail arteries. PTX or LF significantly attenuated phenylphrine (PE)-induced vasoconstriction in a dose-dependent manner. The EC50 for LF and PTX were 336 and 466 microM, respectively. Gastrointestinal disturbances have been reported following oral ingestion of PTX. To clarify the mechanistic basis for this side effect we examined the potential of PTX to modulate spontaneous peristalsis in isolated rat ileum rings. PTX significantly attenuated the spontaneous contractions (oscillations) in a dose-dependent manner. In comparison to isolated rat arterial vessels, the ileum ring preparations were significantly more sensitive (eightfold) to the relaxing effects of PTX (EC50 58 microM). Our data suggest that PTX- or LF-induced changes in tumor blood flow may be the direct result of vascular smooth muscle relaxation. Furthermore, the gastrointestinal disturbances that have been reported in the literature may be a consequence of PTX-induced inhibition of gut peristalsis.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Íleo/metabolismo , Músculo Liso Vascular/metabolismo , Pentoxifilina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Vasoconstritores/farmacologia , Animais , Artérias/metabolismo , Artérias/patologia , Relação Dose-Resposta a Droga , Trato Gastrointestinal/metabolismo , Íleo/patologia , Masculino , Oscilometria , Oxigênio/metabolismo , Pentoxifilina/análogos & derivados , Peristaltismo/efeitos dos fármacos , Fenilefrina/farmacologia , Ratos , Ratos Wistar , Fatores de Tempo , Vasoconstrição/efeitos dos fármacos
14.
PLoS One ; 10(2): e0118187, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25689628

RESUMO

Therapeutic inhibition of poly(ADP-ribose) polymerase (PARP), as monotherapy or to supplement the potencies of other agents, is a promising strategy in cancer treatment. We previously reported that the first PARP inhibitor to enter clinical trial, rucaparib (AG014699), induced vasodilation in vivo in xenografts, potentiating response to temozolomide. We now report that rucaparib inhibits the activity of the muscle contraction mediator myosin light chain kinase (MLCK) 10-fold more potently than its commercially available inhibitor ML-9. Moreover, rucaparib produces additive relaxation above the maximal degree achievable with ML-9, suggesting that MLCK inhibition is not solely responsible for dilation. Inhibition of nitric oxide synthesis using L-NMMA also failed to impact rucaparib's activity. Rucaparib contains the nicotinamide pharmacophore, suggesting it may inhibit other NAD+-dependent processes. NAD+ exerts P2 purinergic receptor-dependent inhibition of smooth muscle contraction. Indiscriminate blockade of the P2 purinergic receptors with suramin abrogated rucaparib-induced vasodilation in rat arterial tissue without affecting ML-9-evoked dilation, although the specific receptor subtypes responsible have not been unequivocally identified. Furthermore, dorsal window chamber and real time tumor vessel perfusion analyses in PARP-1-/- mice indicate a potential role for PARP in dilation of tumor-recruited vessels. Finally, rucaparib provoked relaxation in 70% of patient-derived tumor-associated vessels. These data provide tantalising evidence of the complexity of the mechanism underlying rucaparib-mediated vasodilation.


Assuntos
Aorta/efeitos dos fármacos , Aorta/fisiologia , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Quinase de Cadeia Leve de Miosina/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Receptores Purinérgicos P2/metabolismo , Animais , Aorta/metabolismo , Carcinoma de Células Renais/irrigação sanguínea , Humanos , Neoplasias Renais/irrigação sanguínea , Masculino , Camundongos , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/deficiência , Ratos , Ratos Wistar , Vasodilatação/efeitos dos fármacos
15.
Cancer Gene Ther ; 10(1): 40-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12489027

RESUMO

The bioreductive drug, AQ4N, is metabolized under hypoxic conditions and has been shown to enhance the antitumor effects of radiation and chemotherapy drugs. We have investigated the role of cytochrome P450 3A4 (CYP3A4) in increasing the metabolism of AQ4N using a gene-directed enzyme prodrug therapy (GDEPT) strategy. RIF-1 murine tumor cells were transfected with a mammalian expression vector containing CYP3A4 cDNA. In vitro AQ4N metabolism, DNA damage, and clonogenic cell kill were assessed following exposure of transfected and parental control cells to AQ4N. The presence of exogenous CYP3A4 increased the metabolism of AQ4N and significantly enhanced the ability of the drug to cause DNA strand breaks and clonogenic cell death. Cotransfection of CYP reductase with CYP3A4 showed a small enhancement of the effect in the DNA damage assay only. A single injection of CYP3A4 into established RIF-1 murine tumors increased the metabolism of AQ4N, and when used in combination with radiation, three of nine tumors were locally controlled for >60 days. This is the first demonstration that CYPs alone can be used in a GDEPT strategy for bioreduction of the cytotoxic prodrug, AQ4N. AQ4N is the only CYP-activated bioreductive agent in clinical trials. Combination with a GDEPT strategy may offer a further opportunity for targeting radiation-resistant and chemo-resistant hypoxic tumor cells.


Assuntos
Antraquinonas/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Fibrossarcoma/terapia , Terapia Genética/métodos , Pró-Fármacos/metabolismo , Animais , Antraquinonas/farmacologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Biotransformação , Western Blotting , Hipóxia Celular , Terapia Combinada , Ensaio Cometa , Citocromo P-450 CYP3A , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , DNA de Neoplasias/efeitos dos fármacos , Fibrossarcoma/enzimologia , Humanos , Camundongos , Camundongos Endogâmicos C3H , NADPH-Ferri-Hemoproteína Redutase/genética , Pró-Fármacos/farmacologia , Doses de Radiação , Transfecção , Ensaio Tumoral de Célula-Tronco
16.
Peptides ; 23(9): 1547-55, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12217414

RESUMO

Bradykinin and (Thr(6))-bradykinin have been identified in the defensive skin secretion of the fire-bellied toad, Bombina orientalis. The homologous cDNAs for both peptides were cloned from a skin library using a 3'- and 5'-RACE strategy. Kininogen-1 (BOK-1) contained an open-reading frame of 167 amino acid residues encoding four repeats of bradykinin, and kininogen-2 (BOK-2) contained an open-reading frame of 161 amino acid residues encoding two repeats of (Thr(6))-bradykinin. Alignment of both precursor nucleotide and amino acid sequences revealed a high degree of structural similarity. These amphibian skin kininogens/preprobradykinins are not biologically analogous to mammalian kininogens.


Assuntos
Anuros/metabolismo , Bradicinina/química , Pele/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Bradicinina/genética , Cromatografia Líquida , Clonagem Molecular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Cininogênios/química , Cininogênios/metabolismo , Masculino , Espectrometria de Massas , Dados de Sequência Molecular , Músculo Liso/metabolismo , Fases de Leitura Aberta , Ratos , Ratos Wistar , Homologia de Sequência de Aminoácidos , Raios Ultravioleta
17.
Regul Pept ; 121(1-3): 65-72, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15256275

RESUMO

Structural homologues of vertebrate regulatory peptides found in defensive skin secretions of anuran amphibians often display enhanced bioactivity and receptor binding when compared with endogenous mammalian peptide ligands. Maximakinin, a novel N-terminally extended bradykinin (DLPKINRKGPRPPGFSPFR) from the skin venom of a Chinese toad (Bombina maxima), displays such activity enhancement when compared with bradykinin but is additionally highly selective for mammalian arterial smooth muscle bradykinin receptors displaying a 50-fold increase in molar potency in this smooth muscle type. In contrast, a 100-fold decrease in molar potency was observed at bradykinin receptors in intestinal and uterine smooth muscle preparations. Maximakinin has thus evolved as a "smart" defensive weapon in the toad with receptor/tissue selective targeting. Natural selection of amphibian skin venom peptides for antipredator defence, through inter-species delivery by an exogenous secretory mode, produces subtle structural stabilisation modifications that can potentially provide new insights for the design of selectively targeted peptide therapeutics.


Assuntos
Venenos de Anfíbios/farmacologia , Anuros , Artérias/efeitos dos fármacos , Bradicinina/química , Músculo Liso Vascular/efeitos dos fármacos , Peçonhas/química , Sequência de Aminoácidos , Venenos de Anfíbios/síntese química , Venenos de Anfíbios/química , Animais , Bioensaio , Antagonistas dos Receptores da Bradicinina , Dados de Sequência Molecular , Músculo Liso Vascular/fisiologia , Ratos , Receptores da Bradicinina/metabolismo , Especificidade por Substrato , Fatores de Tempo
18.
Regul Pept ; 116(1-3): 147-54, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14599726

RESUMO

We have isolated a novel bradykinin B(2)-receptor antagonist peptide, kinestatin, from toad (Bombina maxima) defensive skin secretion. Mass spectroscopy established a molecular mass of 931.56 Da and a provisional structure: pGlu-Leu/Ile-Pro-Gly-Leu/Ile-Gly-Pro-Leu/Ile-Arg.amide. The unmodified sequence, -QIPGLGPLRG-, was located at the C-terminus of a 116-amino-acid residue open-reading frame following interrogation of a sequenced B. maxima skin cDNA library database. This confirmed the presence of appropriate primary structural attributes for the observed post-translational modifications present on the mature peptide and established residue 2 as Ile and residues 5/8 as Leu. Kinestatin represents a prototype novel peptide from amphibian skin.


Assuntos
Anuros , Antagonistas de Receptor B2 da Bradicinina , Peptídeos/isolamento & purificação , Peptídeos/farmacologia , Pele/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Bradicinina/farmacologia , DNA Complementar/análise , DNA Complementar/genética , Masculino , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/genética , Fenilefrina/farmacologia , Ratos , Ratos Wistar , Receptor B2 da Bradicinina/metabolismo , Alinhamento de Sequência , Pele/metabolismo , Vasodilatação/efeitos dos fármacos
19.
J Biomed Biotechnol ; 2003(2): 110-137, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12721516

RESUMO

Cancer gene therapy has been one of the most exciting areas of therapeutic research in the past decade. In this review, we discuss strategies to restrict transcription of transgenes to tumour cells. A range of promoters which are tissue-specific, tumour-specific, or inducible by exogenous agents are presented. Transcriptional targeting should prevent normal tissue toxicities associated with other cancer treatments, such as radiation and chemotherapy. In addition, the specificity of these strategies should provide improved targeting of metastatic tumours following systemic gene delivery. Rapid progress in the ability to specifically control transgenes will allow systemic gene delivery for cancer therapy to become a real possibility in the near future.

20.
Oncol Res ; 14(10): 483-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15559762

RESUMO

Nicotinamide has been shown to be an effective tumor oxygenator in preclinical studies and is part of a promising clinical protocol for the treatment of cancer of the larynx. It has been known for some time that nicotinamide sensitizes tumors, at least in part, by modulating vascular smooth muscle contraction; however, the cellular target within the smooth muscle cell has yet to be identified. Our previous studies have eliminated targets within several agonist and antagonist signaling pathways in smooth muscle, suggesting that it must act at a point close to the contractile machinery of the cell. The present study investigated the effect of nicotinamide on four key steps responsible for force generation via actin/myosin interaction in the smooth muscle cell: calcium binding to calmodulin, calcium-calmodulin binding to smooth muscle myosin light chain kinase (MLCK) inhibitor peptide 480-501 (smMLCIP), modulation of MLCK-dependent signaling, and MLCK-induced phosphorylation of the regulatory myosin light chain, MLC20. Nicotinamide abolished the phosphorylation of MLC20, but had no significant effect on the other endpoints tested. We conclude that the vasorelaxant effects of nicotinamide are mediated mainly through inhibition of MLC20 phosphorylation, and that this could be a promising target for the development of novel tumor oxygenators to enhance radio- and chemotherapy.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Músculo Liso Vascular/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Niacinamida/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Cálcio/metabolismo , Calmodulina/metabolismo , Galinhas , Relação Dose-Resposta a Droga , Masculino , Modelos Biológicos , Contração Muscular , Niacinamida/metabolismo , Oxigênio/metabolismo , Peptídeos/química , Fenilefrina/farmacologia , Fosforilação , Ratos , Ratos Wistar , Transdução de Sinais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA