Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 17(14): 13121-13136, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37432926

RESUMO

Ex vivo-loaded white blood cells (WBC) can transfer cargo to pathological foci in the central nervous system (CNS). Here we tested affinity ligand driven in vivo loading of WBC in order to bypass the need for ex vivo WBC manipulation. We used a mouse model of acute brain inflammation caused by local injection of tumor necrosis factor alpha (TNF-α). We intravenously injected nanoparticles targeted to intercellular adhesion molecule 1 (anti-ICAM/NP). We found that (A) at 2 h, >20% of anti-ICAM/NP were localized to the lungs; (B) of the anti-ICAM/NP in the lungs >90% were associated with leukocytes; (C) at 6 and 22 h, anti-ICAM/NP pulmonary uptake decreased; (D) anti-ICAM/NP uptake in brain increased up to 5-fold in this time interval, concomitantly with migration of WBCs into the injured brain. Intravital microscopy confirmed transport of anti-ICAM/NP beyond the blood-brain barrier and flow cytometry demonstrated complete association of NP with WBC in the brain (98%). Dexamethasone-loaded anti-ICAM/liposomes abrogated brain edema in this model and promoted anti-inflammatory M2 polarization of macrophages in the brain. In vivo targeted loading of WBC in the intravascular pool may provide advantages of coopting WBC predisposed to natural rapid mobilization from the lungs to the brain, connected directly via conduit vessels.


Assuntos
Sistemas de Liberação de Medicamentos , Pulmão , Camundongos , Animais , Pulmão/metabolismo , Encéfalo/metabolismo , Lipossomos/metabolismo , Leucócitos/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo
2.
Commun Biol ; 4(1): 299, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33674772

RESUMO

We describe the design, kinetic properties, and structures of engineered subtilisin proteases that degrade the active form of RAS by cleaving a conserved sequence in switch 2. RAS is a signaling protein that, when mutated, drives a third of human cancers. To generate high specificity for the RAS target sequence, the active site was modified to be dependent on a cofactor (imidazole or nitrite) and protease sub-sites were engineered to create a linkage between substrate and cofactor binding. Selective proteolysis of active RAS arises from a 2-step process wherein sub-site interactions promote productive binding of the cofactor, enabling cleavage. Proteases engineered in this way specifically cleave active RAS in vitro, deplete the level of RAS in a bacterial reporter system, and also degrade RAS in human cell culture. Although these proteases target active RAS, the underlying design principles are fundamental and will be adaptable to many target proteins.


Assuntos
Engenharia de Proteínas , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Subtilisina/metabolismo , Células HEK293 , Humanos , Cinética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Proteólise , Proteínas Proto-Oncogênicas p21(ras)/genética , Especificidade por Substrato , Subtilisina/genética
3.
Pharmaceutics ; 13(2)2021 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-33578893

RESUMO

Achieving intracellular delivery of protein therapeutics within cells remains a significant challenge. Although custom formulations are available for some protein therapeutics, the development of non-toxic delivery systems that can incorporate a variety of active protein cargo and maintain their stability, is a topic of great relevance. This study utilized ionic polyphosphazenes (PZ) that can assemble into supramolecular complexes through non-covalent interactions with different types of protein cargo. We tested a PEGylated graft copolymer (PZ-PEG) and a pyrrolidone containing linear derivative (PZ-PYR) for their ability to intracellularly deliver FITC-avidin, a model protein. In endothelial cells, PZ-PYR/protein exhibited both faster internalization and higher uptake levels than PZ-PEG/protein, while in cancer cells both polymers achieved similar uptake levels over time, although the internalization rate was slower for PZ-PYR/protein. Uptake was mediated by endocytosis through multiple mechanisms, PZ-PEG/avidin colocalized more profusely with endo-lysosomes, and PZ-PYR/avidin achieved greater cytosolic delivery. Consequently, a PZ-PYR-delivered anti-F-actin antibody was able to bind to cytosolic actin filaments without needing cell permeabilization. Similarly, a cell-impermeable Bax-BH3 peptide known to induce apoptosis, decreased cell viability when complexed with PZ-PYR, demonstrating endo-lysosomal escape. These biodegradable PZs were non-toxic to cells and represent a promising platform for drug delivery of protein therapeutics.

4.
J Control Release ; 301: 54-61, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-30871995

RESUMO

New advances in intra-arterial (IA) catheters offer clinically proven local interventions in the brain. Here we tested the effect of combining local IA delivery and vascular immunotargeting. Microinjection of tumor necrosis factor alpha (TNFα) in the brain parenchyma causes cerebral overexpression of Inter-Cellular Adhesion Molecule-1 (ICAM-1) in mice. Systemic intravenous injection of ICAM-1 antibody (anti-ICAM-1) and anti-ICAM-1/liposomes provided nearly an order of magnitude higher uptake in the inflamed vs normal brain (from ~0.1 to 0.8%ID/g for liposomes). Local injection of anti-ICAM-1 and anti-ICAM-1/liposomes via carotid artery catheter provided an additional respective 2-fold and 5-fold elevation of uptake in the inflamed brain vs levels attained by IV injection. The uptake in the inflamed brain of respective untargeted IgG counterparts was markedly lower (e.g., uptake of anti-ICAM-1/liposomes was 100-fold higher vs IgG/liposomes). These data affirm the specificity of the combined effect of the first pass and immunotargeting. Intravital real-time microscopy via cranial window revealed that anti-ICAM-1/liposomes, but not IgG/liposomes bind to the lumen of blood vessels in the inflamed brain within minutes after injection. This straightforward framework provides the basis for translational efforts towards local vascular drug targeting to the brain.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Encéfalo/metabolismo , Encefalite/metabolismo , Molécula 1 de Adesão Intercelular/imunologia , Animais , Anticorpos Monoclonais/farmacocinética , Transporte Biológico , Encéfalo/irrigação sanguínea , Encefalite/induzido quimicamente , Lipossomos , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Nanoestruturas/administração & dosagem , Poliestirenos/administração & dosagem , Poliestirenos/farmacocinética , Fator de Necrose Tumoral alfa
5.
Biomacromolecules ; 18(6): 2000-2011, 2017 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-28525259

RESUMO

A series of biodegradable drug delivery polymers with intrinsic multifunctionality have been designed and synthesized utilizing a polyphosphazene macromolecular engineering approach. Novel water-soluble polymers, which contain carboxylic acid and pyrrolidone moieties attached to an inorganic phosphorus-nitrogen backbone, were characterized by a suite of physicochemical methods to confirm their structure, composition, and molecular sizes. All synthesized polyphosphazenes displayed composition-dependent hydrolytic degradability in aqueous solutions at neutral pH. Their formulations were stable at lower temperatures, potentially indicating adequate shelf life, but were characterized by accelerated degradation kinetics at elevated temperatures, including 37 °C. It was found that synthesized polyphosphazenes are capable of environmentally triggered self-assembly to produce nanoparticles with narrow polydispersity in the size range of 150-700 nm. Protein loading capacity of copolymers has been validated via their ability to noncovalently bind avidin without altering biological functionality. Acid-induced membrane-disruptive activity of polyphosphazenes has been established with an onset corresponding to the endosomal pH range and being dependent on polymer composition. The synthesized polyphosphazenes facilitated cell-surface interactions followed by time-dependent, vesicular-mediated, and saturable internalization of a model protein cargo into cancer cells, demonstrating the potential for intracellular delivery.


Assuntos
Ácidos Carboxílicos/química , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Pirrolidinonas/química , Animais , Avidina/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Composição de Medicamentos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Nanopartículas/ultraestrutura , Compostos Organofosforados/síntese química , Compostos Organofosforados/farmacologia , Tamanho da Partícula , Polímeros/síntese química , Polímeros/farmacologia , Suínos
6.
J Control Release ; 238: 221-230, 2016 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-27473764

RESUMO

Intercellular adhesion molecule 1 (ICAM-1) is a cell-surface protein overexpressed in many diseases and explored for endocytosis and transcytosis of drug delivery systems. All previous evidence demonstrating ICAM-1-mediated transport of therapeutics into or across cells was obtained using nanocarriers or conjugates coupled to multiple copies of anti-ICAM antibodies or peptides. Yet, transport of therapeutics linked to non-multivalent anti-ICAM ligands has never been shown, since multivalency was believed to be necessary to induce transport. Our goal was to explore whether non-multivalent binding to ICAM-1 could drive endocytosis and/or transcytosis of model cargo in different cell types. We found that anti-ICAM was specifically internalized by all tested ICAM-1-expressing cells, including epithelial, fibroblast and neuroblastoma cells, primary or established cell lines. Uptake was inhibited at 4°C and in the presence of an inhibitor of the ICAM-1-associated pathway, rather than inhibitors of the clathrin or caveolar routes. We observed minimal transport of anti-ICAM to lysosomes, yet prominent and specific transcytosis across epithelial monolayers. Finally, we coupled a model cargo (the enzyme horseradish peroxidase (HRP)) to anti-ICAM and separated a 1:2 antibody:enzyme conjugate for non-multivalent ICAM-1 targeting. Similar to anti-ICAM, anti-ICAM-HRP was specifically internalized and transported across cells, which rendered intra- and trans-cellular enzyme activity. Therefore, non-multivalent ICAM-1 targeting also provides transport of cargoes into and across cells, representing a new alternative for future therapeutic applications via this route.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Peroxidase do Rábano Silvestre/administração & dosagem , Peroxidase do Rábano Silvestre/farmacocinética , Imunoconjugados/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Animais , Células CACO-2 , Clatrina/metabolismo , Endocitose , Células Endoteliais/metabolismo , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/farmacocinética , Lisossomos/metabolismo , Camundongos
7.
J Control Release ; 210: 39-47, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-25966362

RESUMO

Targeting nanocarriers (NC) to endothelial cell adhesion molecules including Platelet-Endothelial Cell Adhesion Molecule-1 (PECAM-1 or CD31) improves drug delivery and pharmacotherapy of inflammation, oxidative stress, thrombosis and ischemia in animal models. Recent studies unveiled that hydrodynamic conditions modulate endothelial endocytosis of NC targeted to PECAM-1, but the specificity and mechanism of effects of flow remain unknown. Here we studied the effect of flow on endocytosis by human endothelial cells of NC targeted by monoclonal antibodies Ab62 and Ab37 to distinct epitopes on the distal extracellular domain of PECAM. Flow in the range of 1-8dyn/cm(2), typical for venous vasculature, stimulated the uptake of spherical Ab/NC (~180nm diameter) carrying ~50 vs 200 Ab62 and Ab37 per NC, respectively. Effect of flow was inhibited by disruption of cholesterol-rich plasmalemma domains and deletion of PECAM-1 cytosolic tail. Flow stimulated endocytosis of Ab62/NC and Ab37/NC via eliciting distinct signaling pathways mediated by RhoA/ROCK and Src Family Kinases, respectively. Therefore, flow stimulates endothelial endocytosis of Ab/NC in a PECAM-1 epitope specific manner. Using ligands of binding to distinct epitopes on the same target molecule may enable fine-tuning of intracellular delivery based on the hemodynamic conditions in the vascular area of interest.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Portadores de Fármacos/administração & dosagem , Epitopos/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Anticorpos Monoclonais/química , Linhagem Celular Tumoral , Células Cultivadas , Portadores de Fármacos/química , Endocitose , Células Endoteliais da Veia Umbilical Humana , Humanos , Nanopartículas/administração & dosagem , Nanopartículas/química , Poliestirenos/administração & dosagem , Poliestirenos/química , Reologia , Estresse Mecânico
8.
Pharm Res ; 32(4): 1264-78, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25319100

RESUMO

PURPOSE: Delivery of therapeutics to neurons is paramount to treat neurological conditions, including many lysosomal storage disorders. However, key aspects of drug-carrier behavior in neurons are relatively unknown: the occurrence of non-canonical endocytic pathways (present in other cells); whether carriers that traverse the blood-brain barrier are, contrarily, retained within neurons; if neuron-surface receptors are accessible to bulky carriers compared to small ligands; or if there are differences regarding neuronal compartments (neuron body vs. neurites) pertaining said parameters. We have explored these questions using model polymer nanocarriers targeting intercellular adhesion molecule-1 (ICAM-1). METHODS: Differentiated human neuroblastoma cells were incubated with anti-ICAM-coated polystyrene nanocarriers and analyzed by fluorescence microscopy. RESULTS: ICAM-1 expression and nanocarrier binding was enhanced in altered (TNFα) vs. control conditions. While small ICAM-1 ligands (anti-ICAM) preferentially accessed the cell body, anti-ICAM nanocarriers bound with faster kinetics to neurites, yet reached similar saturation over time. Anti-ICAM nanocarriers were also endocytosed with faster kinetics and lower saturation levels in neurites. Non-classical cell adhesion molecule (CAM) endocytosis ruled uptake, and neurite-to-cell body transport was inferred. Nanocarriers trafficked to lysosomes, delivering active enzymes (dextranase) with substrate reduction in a lysosomal-storage disease model. CONCLUSION: ICAM-1-targeting holds potential for intracellular delivery of therapeutics to neurons.


Assuntos
Dextranase/administração & dosagem , Portadores de Fármacos/química , Endocitose/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/metabolismo , Lisossomos/metabolismo , Nanoestruturas/química , Neurônios/efeitos dos fármacos , Transporte Biológico , Linhagem Celular Tumoral , Dextranase/farmacocinética , Dextranase/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Doenças por Armazenamento dos Lisossomos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/metabolismo , Especificidade por Substrato
9.
J Vis Exp ; (80): e50638, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24192611

RESUMO

Sub-micrometer carriers (nanocarriers; NCs) enhance efficacy of drugs by improving solubility, stability, circulation time, targeting, and release. Additionally, traversing cellular barriers in the body is crucial for both oral delivery of therapeutic NCs into the circulation and transport from the blood into tissues, where intervention is needed. NC transport across cellular barriers is achieved by: (i) the paracellular route, via transient disruption of the junctions that interlock adjacent cells, or (ii) the transcellular route, where materials are internalized by endocytosis, transported across the cell body, and secreted at the opposite cell surface (transyctosis). Delivery across cellular barriers can be facilitated by coupling therapeutics or their carriers with targeting agents that bind specifically to cell-surface markers involved in transport. Here, we provide methods to measure the extent and mechanism of NC transport across a model cell barrier, which consists of a monolayer of gastrointestinal (GI) epithelial cells grown on a porous membrane located in a transwell insert. Formation of a permeability barrier is confirmed by measuring transepithelial electrical resistance (TEER), transepithelial transport of a control substance, and immunostaining of tight junctions. As an example, ~200 nm polymer NCs are used, which carry a therapeutic cargo and are coated with an antibody that targets a cell-surface determinant. The antibody or therapeutic cargo is labeled with (125)I for radioisotope tracing and labeled NCs are added to the upper chamber over the cell monolayer for varying periods of time. NCs associated to the cells and/or transported to the underlying chamber can be detected. Measurement of free (125)I allows subtraction of the degraded fraction. The paracellular route is assessed by determining potential changes caused by NC transport to the barrier parameters described above. Transcellular transport is determined by addressing the effect of modulating endocytosis and transcytosis pathways.


Assuntos
Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Intestinal/metabolismo , Nanopartículas/administração & dosagem , Nanopartículas/metabolismo , Células CACO-2 , Permeabilidade da Membrana Celular , Impedância Elétrica , Mucosa Gástrica/citologia , Humanos , Absorção Intestinal , Mucosa Intestinal/citologia , Radioisótopos do Iodo/química , Marcação por Isótopo , Coloração e Rotulagem/métodos , Junções Íntimas/metabolismo , alfa-Galactosidase/administração & dosagem , alfa-Galactosidase/farmacocinética
10.
J Biomater Sci Polym Ed ; 24(8): 912-26, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23647248

RESUMO

Thiolated polymers containing disulfide linkages are commonly researched in gene delivery with the assumption that the thiolated complexes form disulfide bonds. This study investigates the extent of disulfide linking in a thiol-containing polymer and determines the impact that free thiols have on the polymer's delivery potential. A fluorescent cationic polymer containing thiol pendant chains was prepared from poly(allylamine) and 2-iminothiolate (Traut's reagent). Polymer fluorescence was determined by UV plate readings and fluorescent microscopy. Transfection efficiency and cytotoxicity were assessed in MCF-7 breast cancer cells. Results show that thiolated polymers exhibited fluorescence at ex/em ∼595/620. Fluorescent measurements, microscopy imaging, and DNA electrophoresis show that thiolated polymers are not internalized by cells in a culture, yet, they bind to the cell surface, perhaps valuable for applications requiring cell adhesion.


Assuntos
Ácido Ditionitrobenzoico/química , Técnicas de Transferência de Genes , Imidoésteres/química , Poliaminas/química , Contagem de Células , Linhagem Celular , Membrana Celular/metabolismo , DNA/química , DNA/genética , Humanos , Ligação Proteica , Transfecção
11.
J Control Release ; 163(1): 25-33, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-22698938

RESUMO

Bioavailability of oral drugs, particularly large hydrophilic agents, is often limited by poor adhesion and transport across gastrointestinal (GI) epithelial cells. Drug delivery systems, such as sub-micrometer polymer carriers (nanocarriers, NCs) coupled to affinity moieties that target GI surface markers involved in transport, may improve this aspect. To explore this strategy, we coated 100-nm polymer particles with an antibody to ICAM-1 (a protein expressed on the GI epithelium and other tissues) and evaluated targeting, uptake, and transport in human GI epithelial cells. Fluorescence and electron microscopy, and radioisotope tracing revealed that anti-ICAM NCs specifically bound to cells in culture, were internalized via CAM-mediated endocytosis, trafficked by transcytosis across cell monolayers without disrupting the permeability barrier or cell viability, and enabled transepithelial transport of a model therapeutic enzyme (α-galactosidase, deficient in lysosomal Fabry disease). These results indicate that ICAM-1 targeting may provide delivery of therapeutics, such as enzymes, to and across the GI epithelium.


Assuntos
Portadores de Fármacos/administração & dosagem , Imunoglobulina G/administração & dosagem , Molécula 1 de Adesão Intercelular/metabolismo , Nanoestruturas/administração & dosagem , alfa-Galactosidase/administração & dosagem , Transporte Biológico , Células CACO-2 , Trato Gastrointestinal/citologia , Humanos , Molécula 1 de Adesão Intercelular/imunologia , Poliestirenos/administração & dosagem , Fator de Necrose Tumoral alfa/farmacologia
12.
Biomacromolecules ; 13(5): 1331-9, 2012 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-22515194

RESUMO

Polymers have attracted much attention as potential gene delivery vectors due to their chemical and structural versatility. However, several challenges associated with polymeric carriers, including low transfection efficiencies, insufficient cargo release, and high cytotoxicity levels have prevented clinical implementation. Strong electrostatic interactions between polymeric carriers and DNA cargo can prohibit complete cargo release within the cell. As a result, cargo DNA never reaches the cell's nucleus where gene expression takes place. In addition, highly charged cationic polymers have been correlated with high cytotoxicity levels, making them unsuitable carriers in vivo. Using poly(allylamine) (PAA) as a model, we investigated how pH-sensitive disulfide cross-linked polymer networks can improve the delivery potential of cationic polymer carriers. To accomplish this, we conjugated thiol-terminated pendant chains onto the primary amines of PAA using 2-iminothiolane, developing three new polymer vectors with 5, 13, or 20% thiol modification. Unmodified PAA and thiol-conjugated polymers were tested for their ability to bind and release plasmid DNA, their capacity to protect genetic cargo from enzymatic degradation, and their potential for endolysosomal escape. Our results demonstrate that polymer-plasmid complexes (polyplexes) formed by the 13% thiolated polymer demonstrate the greatest delivery potential. At high N/P ratios, all thiolated polymers (but not unmodified counterparts) were able to resist decomplexation in the presence of heparin, a negatively charged polysaccharide used to mimic in vivo polyplex-protein interactions. Further, all thiolated polymers exhibited higher buffering capacities than unmodified PAA and, therefore, have a greater potential for endolysosomal escape. However, 5 and 20% thiolated polymers exhibited poor DNA binding-release kinetics, making them unsuitable carriers for gene delivery. The 13% thiolated polymers, on the other hand, displayed high DNA binding efficiency and pH-sensitive release.


Assuntos
DNA/química , Técnicas de Transferência de Genes , Vetores Genéticos/química , Poliaminas/química , Compostos de Sulfidrila/química , Sítios de Ligação , Plasmídeos/química
13.
J Control Release ; 149(3): 323-31, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21047542

RESUMO

Fabry disease, due to the deficiency of α-galactosidase A (α-Gal), causes lysosomal accumulation of globotriaosylceramide (Gb3) in multiple tissues and prominently in the vascular endothelium. Although enzyme replacement therapy (ERT) by injection of recombinant α-Gal improves the disease outcome, the effects on the vasculopathy associated with life-threatening cerebrovascular, cardiac and renal complications are still limited. We designed a strategy to enhance the delivery of α-Gal to organs and endothelial cells (ECs). We targeted α-Gal to intercellular adhesion molecule 1 (ICAM-1), a protein expressed on ECs throughout the vasculature, by loading this enzyme on nanocarriers coated with anti-ICAM (anti-ICAM/α-Gal NCs). In vitro radioisotope tracing showed efficient loading of α-Gal on anti-ICAM NCs, stability of this formulation under storage and in model physiological fluids, and enzyme release in response to lysosome environmental conditions. In mice, the delivery of (125)I-α-Gal was markedly enhanced by anti-ICAM/(125)I-α-Gal NCs in brain, kidney, heart, liver, lung, and spleen, and transmission electron microscopy showed anti-ICAM/α-Gal NCs attached to and internalized into the vascular endothelium. Fluorescence microscopy proved targeting, endocytosis and lysosomal transport of anti-ICAM/α-Gal NCs in macro- and micro-vascular ECs and a marked enhancement of Gb3 degradation. Therefore, this ICAM-1-targeting strategy may help improve the efficacy of therapeutic enzymes for Fabry disease.


Assuntos
Portadores de Fármacos/química , Endotélio Vascular/citologia , Doença de Fabry/tratamento farmacológico , Molécula 1 de Adesão Intercelular/imunologia , alfa-Galactosidase/administração & dosagem , alfa-Galactosidase/uso terapêutico , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Antígenos Glicosídicos Associados a Tumores/metabolismo , Linhagem Celular , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Terapia de Reposição de Enzimas , Doença de Fabry/enzimologia , Doença de Fabry/patologia , Humanos , Lisossomos/metabolismo , Lisossomos/patologia , Lisossomos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , alfa-Galactosidase/farmacocinética
14.
J Pharmacol Exp Ther ; 317(3): 1161-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16505161

RESUMO

Targeting of diagnostic and therapeutic agents to endothelial cells (ECs) provides an avenue to improve treatment of many maladies. For example, intercellular adhesion molecule 1 (ICAM-1), a constitutive endothelial cell adhesion molecule up-regulated in many diseases, is a good determinant for endothelial targeting of therapeutic enzymes and polymer nanocarriers (PNCs) conjugated with anti-ICAM (anti-ICAM/PNCs). However, intrinsic and extrinsic factors that control targeting of anti-ICAM/PNCs to ECs (e.g., anti-ICAM affinity and PNC valency and flow) have not been defined. In this study we tested in vitro and in vivo parameters of targeting to ECs of anti-ICAM/PNCs consisting of either prototype polystyrene or biodegradable poly(lactic-coglycolic) acid polymers (approximately 200 nm diameter spheres carrying approximately 200 anti-ICAM molecules). Anti-ICAM/PNCs, but not control IgG/PNCs 1) rapidly (t1/2 approximately 5 min) and specifically bound to tumor necrosis factor-activated ECs in a dose-dependent manner (Bmax approximately 350 PNC/cell) at both static and physiological shear stress conditions and 2) bound to ECs and accumulated in the pulmonary vasculature after i.v. injection in mice. Anti-ICAM/PNCs displayed markedly higher EC affinity versus naked anti-ICAM (Kd approximately 80 pM versus approximately 8 nM) in cell culture and, probably because of this factor, higher value (185.3 +/- 24.2 versus 50.5 +/- 1.5% injected dose/g) and selectivity (lung/blood ratio 81.0 +/- 10.9 versus 2.1 +/- 0.02, in part due to faster blood clearance) of pulmonary targeting. These results 1) show that reformatting monomolecular anti-ICAM into high-affinity multivalent PNCs boosts their vascular immuno-targeting, which withstands physiological hydrodynamics and 2) support potential anti-ICAM/PNCs utility for medical applications.


Assuntos
Anticorpos Monoclonais/farmacologia , Portadores de Fármacos/química , Células Endoteliais/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Nanoestruturas/química , Polímeros/química , Animais , Afinidade de Anticorpos , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Glicolatos/química , Humanos , Molécula 1 de Adesão Intercelular/imunologia , Ácido Láctico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Poliestirenos/química , Ratos , Ratos Sprague-Dawley
15.
J Cell Sci ; 116(Pt 8): 1599-609, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12640043

RESUMO

Antibody conjugates directed against intercellular adhesion molecule (ICAM-1) or platelet-endothelial cell adhesion molecule (PECAM-1) have formed the basis for drug delivery vehicles that are specifically recognized and internalized by endothelial cells. There is increasing evidence that ICAM-1 and PECAM-1 may also play a role in cell scavenger functions and pathogen entry. To define the mechanisms that regulate ICAM-1 and PECAM-1 internalization, we examined the uptake of anti-PECAM-1 and anti-ICAM-1 conjugates by endothelial cells. We found that the conjugates must be multimeric, because monomeric anti-ICAM-1 and anti-PECAM-1 are not internalized. Newly internalized anti-ICAM-1 and anti-PECAM-1 conjugates did not colocalize with either clathrin or caveolin, and immunoconjugate internalization was not reduced by inhibitors of clathrin-mediated or caveolar endocytosis, suggesting that this is a novel endocytic pathway. Amiloride and protein kinase C (PKC) inhibitors, agents known to inhibit macropinocytosis, reduced the internalization of clustered ICAM-1 and PECAM-1. However, expression of dominant-negative dynamin-2 constructs inhibited uptake of clustered ICAM-1. Binding of anti-ICAM-1 conjugates stimulated the formation of actin stress fibers by human umbilical vein endothelial cells (HUVEC). Latrunculin, radicicol and Y27632 also inhibited internalization of clustered ICAM-1, suggesting that actin rearrangements requiring Src kinase and Rho kinase (ROCK) were required for internalization. Interestingly, these kinases are part of the signal transduction pathways that are activated when circulating leukocytes engage endothelial cell adhesion molecules, suggesting the possibility that CAM-mediated endocytosis is regulated using comparable signaling pathways.


Assuntos
Endocitose/fisiologia , Endotélio Vascular/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Amidas/farmacologia , Amilorida/farmacologia , Anticorpos Monoclonais/metabolismo , Caveolina 1 , Caveolinas/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Clatrina/farmacologia , Dinaminas/genética , Dinaminas/fisiologia , Endocitose/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/imunologia , Lactonas/farmacologia , Macrolídeos , Modelos Biológicos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Piridinas/farmacologia , Tiazóis/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA