Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Pharmacol Exp Ther ; 374(1): 52-61, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32327529

RESUMO

The analgesic potency of morphine-6-glucuronide (M6G) has been shown to be 50-fold higher than morphine after intracerebral injection. However, the brain penetration of M6G is significantly lower than morphine, thus limiting its usefulness in pain management. Here, we created new entities by the conjugation of the angiopep-2 peptide (An2) that crosses the blood-brain barrier (BBB) by low-density lipoprotein receptor-related protein 1 receptor-mediated transcytosis with either morphine or M6G. We demonstrated improvement of BBB permeability of these new entities compared with that of unconjugated M6G and morphine. Intravenous or subcutaneous administration of the An2-M6G conjugate exerted greater and more sustained analgesic activity than equivalent doses of either morphine or M6G. Likewise, subcutaneous An2-morphine induced a delayed but prolonged antinociceptive effect. The effects of these conjugates on the gastrointestinal tract motility were also evaluated. An2-morphine significantly reduced the intestinal transit time, whereas An2-M6G exhibited a reduced constipation profile, as compared with an equimolar dose of morphine. In summary, we have developed new brain-penetrant opioid conjugates exhibiting improved analgesia to side effect ratios. These results thus support the use of An2-carrier peptides as an innovative BBB-targeting technology to deliver effective drugs, such as M6G, for pain management. SIGNIFICANCE STATEMENT: The metabolite morphine-6-glucuronide (M6G) does not efficiently cross the blood-brain barrier. The low-density lipoprotein receptor-related protein 1 peptide ligand angiopep-2 may serve as an effective drug delivery system to the brain. Here, we demonstrated that the coupling of M6G to angiopep-2 peptide (An2) improves its brain penetration and significantly increases its analgesic potency. The An2-M6G conjugate has a favorable side effect profile that includes reduction of developing constipation. An2-M6G exhibits a unique pharmacodynamic profile with a better therapeutic window than morphine.


Assuntos
Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Barreira Hematoencefálica/metabolismo , Derivados da Morfina/química , Derivados da Morfina/metabolismo , Peptídeos/química , Administração Intravenosa , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Transporte Biológico , Motilidade Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Derivados da Morfina/administração & dosagem , Derivados da Morfina/farmacologia , Nociceptividade/efeitos dos fármacos
2.
Anal Bioanal Chem ; 409(5): 1425-1433, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27873002

RESUMO

Mucopolysaccharidosis type II (Hunter's disease) mouse model (IdS-KO) was investigated by both imaging mass spectrometry (IMS) and immunohistochemistry (IHC) performed on the same tissue sections. For this purpose, IdS-KO mice brain sections were coated with sublimated 1,5-diaminonaphtalene and analyzed by high spatial resolution IMS (5 µm) and anti-GM3 IHC on the same tissue sections to characterize the ganglioside monosialated ganglioside (GM) deposits found in Hunter's disease. IMS analysis have found that two species of GM3 and GM2 that are only different due to the length of their fatty acid residue (stearic or arachidic residue) were overexpressed in the IdS-KO mice compared to a control mouse. GM3 and GM2 were characterized by on-tissue exact mass and MS/MS compared to a GM3 standard. Realignment of both IMS and IHC data sets further confirmed the observed regioselective signal previously detected by providing direct correlation of the IMS image for the two GM3 overly expressed MS signals with the anti-GM3 IHC image. Furthermore, these regioselective GM MS signals were also found to have highly heterogeneous distributions within the GM3-IHC staining. Some deposits showed high content in GM3 and GM2 stearic species (r = 0.74) and others had more abundant GM3 and GM2 arachidic species (r = 0.76). Same-section analysis of Hunter's disease mouse model by both high spatial resolution IMS and IHC provides a more in-depth analysis of the composition of the GM aggregates while providing spatial distribution of the observed molecular species. Graphical Abstract Ganglioside imaging mass spectrometry followed by immunohistochemistry performed on the same tissue section.


Assuntos
Encéfalo/metabolismo , Gangliosídeo G(M2)/metabolismo , Gangliosídeo G(M3)/metabolismo , Imuno-Histoquímica/métodos , Mucopolissacaridose II/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Animais , Camundongos , Camundongos Knockout
3.
J Cell Mol Med ; 14(12): 2827-39, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19818094

RESUMO

New and effective therapeutics that cross the blood-brain barrier (BBB) are critically needed for treatment of many brain diseases. We characterize here a novel drug development platform that is broadly applicable for the development of new therapeutics with increased brain penetration. The platform is based on the Angiopep-2 peptide, a sequence derived from ligands that bind to low-density lipoprotein receptor-related protein-1 (LRP-1), a receptor expressed on the BBB. Fluorescent imaging studies of a Cy5.5Angiopep-2 conjugate and immunohistochemical studies of injected Angiopep-2 in mice demonstrated efficient transport across the BBB into brain parenchyma and subsequent co-localization with the neuronal nuclei-selective marker NeuN and the glial marker glial fibrillary acidic protein (GFAP). Uptake of [(¹²5I]-Angiopep-2 into brain endothelial cells occurred by a saturable mechanism involving LRP-1. The primary sequence and charge of Angiopep-2 were crucial for its passage across the BBB. Overall, the results demonstrate the significant potential of this platform for the development of novel neurotherapeutics.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos , Peptídeos/metabolismo , Animais , Antígenos Nucleares/análise , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Carbocianinas , Células Endoteliais/metabolismo , Proteína Glial Fibrilar Ácida/análise , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/análise , Transporte Proteico , Ensaio Radioligante , Ratos , Transcitose
4.
J Neurochem ; 106(4): 1534-44, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18489712

RESUMO

The blood-brain barrier (BBB) restricts the entry of proteins as well as potential drugs to cerebral tissues. We previously reported that a family of Kunitz domain-derived peptides called Angiopeps can be used as a drug delivery system for the brain. Here, we further characterize the transcytosis ability of these peptides using an in vitro model of the BBB and in situ brain perfusion. These peptides, and in particular Angiopep-2, exhibited higher transcytosis capacity and parenchymal accumulation than do transferrin, lactoferrin, and avidin. Angiopep-2 transport and accumulation in brain endothelial cells were unaffected by the P-glycoprotein inhibitor, cyclosporin A, indicating that this peptide is not a substrate for the efflux pump P-glycoprotein. However, competition studies show that activated alpha(2)-macroglobulin, a specific ligand for the low-density lipoprotein receptor-related protein-1 (LRP1) and Angiopep-2 can share the same receptor. In addition, LRP1 was detected in glioblastomas and brain metastases from lung and skin cancers. Fluorescent microscopy also revealed that Alexa488-Angiopep-2 co-localized with LRP1 in brain endothelial cell monolayers. Overall, these results suggest that Angiopep-2 transport across the BBB is, in part, mediated by LRP1.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/fisiologia , Sistemas de Liberação de Medicamentos/métodos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Oligopeptídeos/fisiologia , Fragmentos de Peptídeos/metabolismo , Somatostatina/análogos & derivados , Animais , Barreira Hematoencefálica/metabolismo , Bovinos , Linhagem Celular Tumoral , Células Cultivadas , Vetores Genéticos/administração & dosagem , Vetores Genéticos/metabolismo , Humanos , Fragmentos de Peptídeos/administração & dosagem , Peptídeos Cíclicos , Transporte Proteico/fisiologia , Ratos , Somatostatina/fisiologia
5.
Biochem Biophys Res Commun ; 372(3): 440-6, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18485890

RESUMO

We have investigated the involvement of P-glycoprotein (P-gp)/caveolin-1 interaction in the regulation of brain endothelial cells (EC) migration and tubulogenesis. P-gp overexpression in MDCK-MDR cells was correlated with enhanced cell migration whereas treatment with P-gp inhibitors CsA or PSC833 reduced it. Transfection of RBE4 rat brain endothelial cells with mutated versions of MDR1, in the caveolin-1 interaction motif, decreased the interaction between P-gp and caveolin-1, enhanced P-gp transport activity and cell migration. Moreover, down-regulation of caveolin-1 in RBE4 cells by siRNA against caveolin-1 stimulated cell migration. Interestingly, the inhibition of P-gp/caveolin-1 interaction increased also EC tubulogenesis. Furthermore, decrease of P-gp expression by siRNA inhibited EC tubulogenesis. These data indicate that the level of P-gp/caveolin-1 interaction can modulate brain endothelial angiogenesis and P-gp dependent cell migration.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Encéfalo/irrigação sanguínea , Caveolina 1/metabolismo , Movimento Celular , Células Endoteliais/fisiologia , Neovascularização Fisiológica , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Caveolina 1/genética , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Ciclosporina/farmacologia , Ciclosporinas/farmacologia , Cães , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , RNA Interferente Pequeno/genética , Ratos
6.
J Pharmacol Exp Ther ; 324(3): 1064-72, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18156463

RESUMO

By controlling access to the brain, the blood-brain barrier (BBB) restricts the entry of proteins and potential drugs to cerebral tissues. We demonstrate here the transcytosis ability of aprotinin and peptides derived from Kunitz domains using an in vitro model of the BBB and in situ brain perfusion. Aprotinin transcytosis across bovine brain capillary endothelial cell (BBCEC) monolayers is at least 10-fold greater than that of holo-transferrin. Sucrose permeability was unaffected by high concentrations of aprotinin, indicating that transcytosis of aprotinin was unrelated to changes in the BBCEC monolayer integrity. Alignment of the amino acid sequence of aprotinin with the Kunitz domains of human proteins allowed the identification and design of a family of peptides, named Angiopeps. These peptides, and in particular Angiopep-2, exhibit higher transcytosis capacity and parenchyma accumulation than aprotinin. Overall, these results suggest that these Kunitz-derived peptides could be advantageously used as a new brain delivery system for pharmacological agents that do not readily enter the brain.


Assuntos
Encéfalo/metabolismo , Química Farmacêutica/métodos , Sistemas de Liberação de Medicamentos/métodos , Peptídeos/administração & dosagem , Peptídeos/genética , Sequência de Aminoácidos/genética , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Técnicas de Cocultura , Humanos , Camundongos , Dados de Sequência Molecular , Peptídeos/farmacocinética , Ratos
7.
Brain Res Mol Brain Res ; 135(1-2): 93-103, 2005 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15857672

RESUMO

Protein l-isoaspartyl methyltransferase (PIMT) functions as a repair enzyme that acts upon damaged proteins bearing abnormal aspartyl residues. We previously reported that PIMT expression and activity are reduced by half in human epileptic hippocampus. Here we investigated PIMT regulation in astrocytic tumors, which are the most common human brain tumors. PIMT expression and enzyme activity were significantly decreased in all grades of human astrocytic tumors. More precisely, PIMT levels were significantly lower by 76% in pilocytic astrocytomas (grade I), 46% in astrocytomas (grade II), 69% in anaplastic astrocytomas (grade III), and a marked 80% in glioblastomas (grade IV) as compared to normal brains. RT-PCR analysis showed that levels of type I PIMT mRNA were up-regulated while those of type II PIMT mRNA were down-regulated in glioblastomas. Furthermore, the reduced PIMT levels correlated closely with a decrease in the number of neuron cells in astrocytic tumors as assessed by measuring the neuron-specific enolase level. Many proteins with abnormal aspartyl residues accumulated in brain tumors and some were specific to individual grades of astrocytic tumors. Similar results were obtained, either by measuring the reduction in PIMT activity and expression or by measuring the formation of abnormal proteins, in an orthotopic rat brain tumor model implanted with invasive CNS-1 glioma cells. The novelty of these findings was to provide the first evidence for a marked reduction of PIMT expression and activity during stage progression of astrocytic tumors in humans.


Assuntos
Neoplasias Encefálicas/enzimologia , Encéfalo/enzimologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glioma/enzimologia , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/metabolismo , Animais , Northern Blotting , Proteína Glial Fibrilar Ácida/metabolismo , Glioma/classificação , Humanos , Imuno-Histoquímica/métodos , Masculino , Metilação , Transplante de Neoplasias/métodos , Fosfopiruvato Hidratase/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
8.
Mol Cancer Ther ; 14(1): 129-40, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25492620

RESUMO

Anti-HER2 monoclonal antibodies (mAb) have been shown to reduce tumor size and increase survival in patients with breast cancer, but they are ineffective against brain metastases due to poor brain penetration. In previous studies, we identified a peptide, known as Angiopep-2 (An2), which crosses the blood-brain barrier (BBB) efficiently via receptor-mediated transcytosis, and, when conjugated, endows small molecules and peptides with this property. Extending this strategy to higher molecular weight biologics, we now demonstrate that a conjugate between An2 and an anti-HER2 mAb results in a new chemical entity, ANG4043, which retains in vitro binding affinity for the HER2 receptor and antiproliferative potency against HER2-positive BT-474 breast ductal carcinoma cells. Unlike the native mAb, ANG4043 binds LRP1 clusters and is taken up by LRP1-expressing cells. Measuring brain exposure after intracarotid delivery, we demonstrate that the new An2-mAb conjugate penetrates the BBB with a rate of brain entry (Kin) of 1.6 × 10(-3) mL/g/s. Finally, in mice with intracranially implanted BT-474 xenografts, systemically administered ANG4043 increases survival. Overall, this study demonstrates that the incorporation of An2 to the anti-HER2 mAb confers properties of increased uptake in brain endothelial cells as well as BBB permeability. These characteristics of ANG4043 result in higher exposure levels in BT-474 brain tumors and prolonged survival following systemic treatment. Moreover, the data further validate the An2-drug conjugation strategy as a way to create brain-penetrant biologics for neuro-oncology and other CNS indications.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Neoplasias Encefálicas/tratamento farmacológico , Imunoconjugados/administração & dosagem , Imunoconjugados/farmacocinética , Peptídeos/administração & dosagem , Peptídeos/farmacocinética , Receptor ErbB-2/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Antineoplásicos/síntese química , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Feminino , Células MCF-7 , Camundongos , Camundongos Nus , Peptídeos/síntese química , Peptídeos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Mol Neurobiol ; 30(2): 157-83, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15475625

RESUMO

The blood-brain barrier contributes to brain homeostasis by controlling the access of nutrients and toxic substances to the central nervous system (CNS). The acquired brain endothelial cells phenotype results from their sustained interactions with their microenvironment. The endothelial component is involved in the development and progression of most CNS diseases such as brain tumors, Alzheimer's disease, or stroke, for which efficient treatments remain to be discovered. The endothelium constitutes an attractive therapeutical target, particularly in the case of brain tumors, because of the high level of angiogenesis associated with this disease. Drug development based on targeting differential protein expression in the vasculature associated with normal tissues or with disease states holds great potential. This article highlights some of the growing body of evidence showing molecular differences between the vascular bed phenotype of normal and pathological endothelium, with a particular focus on brain tumor endothelium targets, which may play crucial roles in the development of brain cancers. Finally, an overview is presented of the emerging therapies for brain tumors that take the endothelial component into consideration.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/citologia , Sistemas de Liberação de Medicamentos/métodos , Endotélio Vascular/citologia , Inibidores da Angiogênese/administração & dosagem , Animais , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos
10.
Vascul Pharmacol ; 38(6): 339-48, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12529928

RESUMO

1. The blood-brain barrier (BBB) contributes to brain homeostastis and fulfills a protective function by controlling the access of solutes and toxic substances to the central nervous system (CNS). The efflux transporter P-glycoprotein (P-gp) is a key element of the molecular machinery that confers special permeability properties to the BBB. 2. P-gp, which was initially recognized for its ability to expel anticancer drugs from multidrug-resistant cancer cells, is strongly expressed in brain capillaries. Its expression in the BBB limits the accumulation of many hydrophobic molecules and potentially toxic substances in the brain. 3. The purpose of this review is to summarize the current state of knowledge about the expression of P-gp, its cellular localization as well as its possible functions in the BBB.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Barreira Hematoencefálica , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Transporte Biológico , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Ensaios Clínicos como Assunto , Resistencia a Medicamentos Antineoplásicos , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Microcirculação/citologia , Microcirculação/metabolismo
11.
J Clin Invest ; 124(3): 1199-213, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24531547

RESUMO

Neurotensin (NT) has emerged as an important modulator of nociceptive transmission and exerts its biological effects through interactions with 2 distinct GPCRs, NTS1 and NTS2. NT provides strong analgesia when administered directly into the brain; however, the blood-brain barrier (BBB) is a major obstacle for effective delivery of potential analgesics to the brain. To overcome this challenge, we synthesized chemical conjugates that are transported across the BBB via receptor-mediated transcytosis using the brain-penetrant peptide Angiopep-2 (An2), which targets LDL receptor-related protein-1 (LRP1). Using in situ brain perfusion in mice, we found that the compound ANG2002, a conjugate of An2 and NT, was transported at least 10 times more efficiently across the BBB than native NT. In vitro, ANG2002 bound NTS1 and NTS2 receptors and maintained NT-associated biological activity. In rats, i.v. ANG2002 induced a dose-dependent analgesia in the formalin model of persistent pain. At a dose of 0.05 mg/kg, ANG2002 effectively reversed pain behaviors induced by the development of neuropathic and bone cancer pain in animal models. The analgesic properties of ANG2002 demonstrated in this study suggest that this compound is effective for clinical management of persistent and chronic pain and establish the benefits of this technology for the development of neurotherapeutics.


Assuntos
Analgésicos/farmacologia , Nociceptividade/efeitos dos fármacos , Peptídeos/farmacologia , Succinimidas/farmacologia , Analgésicos/síntese química , Analgésicos/farmacocinética , Animais , Barreira Hematoencefálica/metabolismo , Neoplasias Ósseas/patologia , Permeabilidade Capilar , Linhagem Celular , Linhagem Celular Tumoral , Dor Crônica/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos , Formaldeído , Concentração Inibidora 50 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Peptídeos/síntese química , Peptídeos/farmacocinética , Ratos , Ratos Sprague-Dawley , Succinimidas/síntese química , Succinimidas/farmacocinética
12.
Mol Nutr Food Res ; 54(8): 1192-201, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20087857

RESUMO

Among the natural products shown to possess chemopreventive and anticancer properties, curcumin is one of the most potent. In the current study, we investigated the effects of this natural product on the growth of human glioma U-87 cells xenografted into athymic mice. We show here that curcumin administration exerted significant anti-tumor effects on subcutaneous and intracerebral gliomas as demonstrated by the slower tumor growth rate and the increase of animal survival time. While investigating the mechanism of its action in vivo, we observed that curcumin decreased the gelatinolytic activities of matrix metalloproteinase-9. Furthermore, treatment with curcumin inhibited glioma-induced angiogenesis as indicated by the decrease of endothelial cell marker from newly formed vessels and by the diminution of the concentration of hemoglobin in curcumin-treated tumors. We also demonstrate, using an in vitro model of blood-brain barrier, that curcumin can cross the blood-brain barrier to a high level. These are the first results showing that curcumin suppresses tumor growth of gliomas in xenograft models. The mechanisms of the anti-tumor effects of curcumin were related, at least partly, to the inhibition of glioma-induced angiogenesis.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticarcinógenos/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Curcumina/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/prevenção & controle , Neovascularização Patológica/prevenção & controle , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/farmacologia , Animais , Animais Recém-Nascidos , Anticarcinógenos/farmacocinética , Anticarcinógenos/farmacologia , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/farmacologia , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Curcumina/farmacocinética , Curcumina/farmacologia , Feminino , Glioblastoma/irrigação sanguínea , Glioblastoma/patologia , Humanos , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Nus , Ratos , Análise de Sobrevida , Transcitose , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Med Chem ; 53(7): 2814-24, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20210346

RESUMO

This report describes the synthesis and preliminary biological characterization of 2 (ANG1007) and 3 (ANG1009), two new chemical entities under development for the treatment of primary and secondary brain cancers. 2 consists of three doxorubicin molecules conjugated to Angiopep-2, a 19-mer peptide that crosses the blood-brain barrier (BBB) by an LRP-1 receptor-mediated transcytosis mechanism. 3 has a similar structure, with the exception that three etoposide moieties are conjugated to Angiopep-2. Both agents killed cancer cell lines in vitro with similar IC(50) values and with apparently similar cytotoxic mechanisms as unconjugated doxorubicin and etoposide. 2 and 3 exhibited dramatically higher BBB influx rate constants than unconjugated doxorubicin and etoposide and pooled within brain parenchymal tissue. Passage through the BBB was similar in Mdr1a (-/-) and wild type mice. These results provide further evidence of the potential of this drug development platform in the isolation of novel therapeutics with increased brain penetration.


Assuntos
Antineoplásicos/química , Antineoplásicos/metabolismo , Encéfalo/metabolismo , Doxorrubicina/análogos & derivados , Doxorrubicina/química , Doxorrubicina/metabolismo , Etoposídeo/análogos & derivados , Etoposídeo/química , Etoposídeo/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Sequência de Aminoácidos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/citologia , Encéfalo/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Etoposídeo/farmacologia , Etoposídeo/uso terapêutico , Etoposídeo/toxicidade , Feminino , Humanos , Concentração Inibidora 50 , Cinética , Masculino , Camundongos , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Peptídeos/toxicidade
14.
J Neurochem ; 101(1): 1-8, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17326770

RESUMO

p-glycoprotein (p-gp) is an ATP-binding cassette transporter and its overexpression is responsible for the acquisition of the multidrug resistance phenotype in human tumors. p-gp is localized at the blood-brain barrier and is involved in brain cytoprotection. Our previous work used immunoprecipitation to show that caveolin-1 can interact with p-gp. In this study, we provide evidence that caveolin-1 regulates p-gp transport activity in a rat brain endothelial cell line (RBE4). Down-regulation of caveolin-1 by siRNA reduced the interaction between p-gp and caveolin-1, followed by a decrease in [3H]-Taxol and [3H]-Vinblastine accumulation in RBE4 cells. The latter result showed that down-regulation of caveolin-1 enhanced p-gp transport activity. RBE4 cells were also transfected with Sarcoma in order to modulate caveolin-1 phosphorylation. Overexpression of Sarcoma, a protein tyrosine kinase, stimulated caveolin-1 phosphorylation and increased both [3H]-Taxol and [3H]-Vinblastine accumulation as well as Hoechst 33342 accumulation. Transfection of caveolin-1 inhibits p-gp transport activity. Conversely, transfection of the mutant cavY14F decreased the p-gp/caveolin-1 interaction and reduced accumulation of the two p-gp substrates. Thus, our data show that caveolin-1 regulates p-gp function through the phosphorylation state of caveolin-1 in endothelial cells from the blood-brain barrier.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Barreira Hematoencefálica/metabolismo , Caveolina 1/metabolismo , Células Endoteliais/metabolismo , Animais , Antineoplásicos/farmacologia , Caveolina 1/genética , Linhagem Celular Transformada , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Endoteliais/efeitos dos fármacos , Paclitaxel/farmacocinética , Fosforilação , Transporte Proteico/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Vimblastina/farmacocinética , Quinases da Família src/genética , Quinases da Família src/metabolismo
15.
J Neurochem ; 84(2): 316-24, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12558994

RESUMO

Endothelial cells (ECs) are new targets for tumor therapy. In this work, we purified endothelial cells from intracerebral and subcutaneous experimental gliomas as well as from normal brain in order to define some of the phenotypical differences between angiogenic and quiescent brain vasculature. We show that the multidrug resistance genes encoding drug efflux pumps at the brain endothelium are expressed differently in normal and tumoral vasculature. We also show that ECs from gliomas present increased activity of gelatinase B (MMP9), key enzyme in the angiogenic process. Importantly, we observe a different phenotype between ECs in the intracerebral and subcutaneous models. Our results provide molecular evidence of phenotypic distinction between tumoral and normal brain vasculature and indicate that the EC phenotype depends on interactions both with tumor cells and also with the microenvironment.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Encéfalo/irrigação sanguínea , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Glioma/irrigação sanguínea , Metaloproteinases da Matriz/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Biomarcadores Tumorais/biossíntese , Divisão Celular/efeitos dos fármacos , Movimento Celular , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Endotélio Vascular/citologia , Ativação Enzimática/fisiologia , Masculino , Camundongos , Transplante de Neoplasias , Fenótipo , Ratos , Ratos Endogâmicos Lew , Frações Subcelulares/química , Frações Subcelulares/metabolismo , Células Tumorais Cultivadas
16.
Biochem Biophys Res Commun ; 324(2): 937-45, 2004 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-15474518

RESUMO

The organosulfur compounds (OSCs), present in garlic, are studied for their protective effect against human cancers. P-glycoprotein (P-gp) and multidrug resistance protein 2 (Mrp2) are two transporters involved in the defense of cells and in the development of multidrug resistance. Whereas OSCs increase glutathione S-transferase activity (GST), Mrp2 plays a role in the transport of glutathione (GSH)-conjugates. In this study, we have investigated the effect of two OSCs, diallyl disulfide (DADS) and S-allyl cysteine (SAC), on P-gp and Mrp2 expression in renal brush-border membranes. By Western blot analysis, our results show that DADS induces Mrp2 expression (by 7-fold), which correlates with the rise of GST activity and GSH levels. Surprisingly, a co-administration of OSC with cisplatin, an anticancer drug, significantly increased Mrp2 gene and protein expression (by 30-fold), suggesting that DADS could potentiate the effects of cisplatin. Interestingly, SAC and cisplatin in co-treatment decreased P-gp protein expression and mdr1b isoform mRNA levels. In addition, modulation of the mdr1b isoform and Mrp2 by cisplatin was completely abolished by a glutathione precursor, N-acetyl cysteine. These results indicate that OSCs present in a garlic-rich diet might alter chemotherapeutic treatments using P-gp or Mrp2 substrates.


Assuntos
Compostos Alílicos/farmacologia , Anticarcinógenos/farmacologia , Cisteína/análogos & derivados , Dissulfetos/farmacologia , Alho/metabolismo , Proteínas Mitocondriais/biossíntese , Extratos Vegetais/farmacologia , Proteínas Ribossômicas/biossíntese , Proteínas de Saccharomyces cerevisiae/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Acetilcisteína/metabolismo , Compostos Alílicos/metabolismo , Animais , Western Blotting , Membrana Celular/metabolismo , Cisplatino/farmacologia , Cisteína/metabolismo , Dissulfetos/metabolismo , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Rim/metabolismo , Córtex Renal/metabolismo , Fígado/metabolismo , Masculino , Microvilosidades/metabolismo , Proteínas Mitocondriais/genética , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Ribossômicas/genética , Proteínas de Saccharomyces cerevisiae/genética
17.
J Neurosci Res ; 75(2): 291-299, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14705150

RESUMO

Primary brain tumors, particularly glioblastomas (GB), remain a challenge for oncology. An element of the malignant brain tumors' aggressive behavior is the fact that GB are among the most densely vascularized tumors. To determine some of the molecular regulations occuring at the brain tumor endothelium level during tumoral progression would be an asset in understanding brain tumor biology. Caveolin-1 is an essential structural constituent of caveolae that has been implicated in mitogenic signaling, oncogenesis, and angiogenesis. In this work we investigated regulation of caveolin-1 expression in brain endothelial cells (ECs) under angiogenic conditions. In vitro, brain EC caveolin-1 is down-regulated by angiogenic factors treament and by hypoxia. Coculture of brain ECs with tumoral cells induced a similar down-regulation. In addition, activation of the p42/44 MAP kinase is demonstrated. By using an in vivo brain tumor model, we purified ECs from gliomas as well as from normal brain to investigate possible regulation of caveolin-1 expression in tumoral brain vasculature. We show that caveolin-1 expression is strikingly down-regulated in glioma ECs, whereas an increase of phosphorylated caveolin-1 is observed. Whole-brain radiation treatment, a classical way in which GB is currently being treated, resulted in increased caveolin-1 expression in tumor isolated ECs. The level of tumor cells spreading around newly formed blood vessels was also elevated. The regulation of caveolin-1 expression in tumoral ECs may reflect the tumoral vasculature state and correlates with angiogenesis kinetics.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Caveolinas/metabolismo , Células Endoteliais/metabolismo , Glioma/irrigação sanguínea , Neovascularização Patológica/metabolismo , Proteínas Angiogênicas/farmacologia , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Cavéolas/efeitos da radiação , Caveolina 1 , Caveolinas/efeitos da radiação , Linhagem Celular Tumoral , Técnicas de Cocultura , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Regulação para Baixo/efeitos da radiação , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/efeitos da radiação , Glioma/metabolismo , Glioma/radioterapia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Masculino , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/efeitos da radiação , Metástase Neoplásica/fisiopatologia , Metástase Neoplásica/radioterapia , Neovascularização Patológica/fisiopatologia , Neovascularização Patológica/radioterapia , Fosforilação/efeitos da radiação , Ratos , Ratos Endogâmicos Lew
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA