Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Control Release ; 144(3): 341-9, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20226822

ABSTRACT

In non-phagocytic cells such as endothelial cells, processing of liposomes and subsequent release of drug content is often inefficient due to the absence of professional processing machinery, which limits pharmacological efficacy. We therefore developed a liposome based drug delivery system with superior intracellular release characteristics. The design was based on long circulating conventional liposomes that were formulated with a cationic amphiphile, 1-methyl-4-(cis-9-dioleyl)methyl-pyridinium-chlorid (SAINT-C18). These so-called SAINT-O-Somes had a diameter of 100 nm, were as stable as conventionally formulated liposomes, and showed superior release of their content at pH conditions that liposomes encounter when they are endocytosed by cells. Attachment of anti-E-selectin specific antibodies to the distal end of surface grafted poly(ethylene glycol) resulted in immuno-SAINT-O-Somes that were as efficiently taken up by inflammation activated endothelial cells as conventional anti-E-selectin specific immunoliposomes. More importantly, intracellular release of calcein encapsulated in these targeted SAINT-O-Somes was 10 fold higher as compared to the release of calcein from conventional liposomes. For intracellular delivery siRNA into activated endothelial cells, formulation with SAINT-C18 was a necessity to induce a specific down-regulation of gene expression of VE-cadherin. Additionally, targeted doxorubicin loaded SAINT-O-Somes decreased endothelial cell viability significantly more than targeted conventional doxorubicin liposomes. SAINT-O-Somes therefore represent a new class of lipid based particles with superior drug release characteristics that can be applied for the efficacious intracellular delivery of hydrophilic drugs including siRNA.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Endothelial Cells/drug effects , Pyridinium Compounds/chemistry , RNA, Small Interfering/administration & dosage , Surface-Active Agents/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line , Cell Survival/drug effects , Cryoelectron Microscopy , Drug Compounding , E-Selectin/genetics , Endothelial Cells/metabolism , Flow Cytometry , Gene Expression/drug effects , Humans , Lipids/chemistry , Liposomes , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Particle Size , RNA, Small Interfering/pharmacokinetics
2.
J Control Release ; 141(2): 241-51, 2010 Jan 25.
Article in English | MEDLINE | ID: mdl-19766679

ABSTRACT

Applications of small-interfering RNA (siRNA) call for specific and efficient delivery of siRNA into particular cell types. We developed a novel, non-viral targeting system to deliver siRNA specifically into inflammation-activated endothelial cells. This was achieved by conjugating the cationic amphiphilic lipid SAINT to antibodies recognizing the inflammatory cell adhesion molecule E-selectin. These anti-E-selectin-SAINT lipoplexes (SAINTarg) maintained antigen recognition capacity of the parental antibody in vitro, and ex vivo in human kidney tissue slices subjected to inflammatory conditions. Regular SAINT mediated transfection resulted in efficient gene silencing in human microvascular endothelial cells (HMEC-1) and conditionally immortalized glomerular endothelial cells (ciGEnC). However, primary human umbilical vein endothelial cells (HUVEC) transfected poorly, a phenomenon that we could quantitatively correlate with a cell-type specific capacity to facilitate siRNA uptake. Importantly, SAINTarg increased siRNA uptake and transfection specificity for activated endothelial cells. Transfection with SAINTarg delivered significantly more siRNA into activated HUVEC, compared to transfection with non-targeted SAINT. The enhanced uptake of siRNA was corroborated by improved silencing of both gene- and protein expression of VE-cadherin in activated HUVEC, indicating that SAINTarg delivered functionally active siRNA into endothelial cells. The obtained results demonstrate a successful design of a small nucleotide carrier system with improved and specific siRNA delivery into otherwise difficult-to-transfect primary endothelial cells, which in addition reduced considerably the amount of siRNA needed for gene silencing.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Endothelial Cells/metabolism , Inflammation/metabolism , Kidney/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transfection , Aged , Antibodies, Monoclonal/metabolism , Binding Sites , Biological Transport , Cadherins/immunology , Cell Culture Techniques , Cells, Cultured , Endothelial Cells/immunology , Humans , Inflammation/immunology , Kidney/immunology , Male , Middle Aged , Particle Size , Pyridinium Compounds/metabolism , Tissue Culture Techniques
3.
Am J Physiol Renal Physiol ; 297(2): F272-81, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19515812

ABSTRACT

Both hemorrhagic shock and endotoxemia induce a pronounced vascular activation in the kidney which coincides with albuminuria and glomerular barrier dysfunction. We hypothesized that changes in Tie2, a vascular restricted receptor tyrosine kinase shown to control microvascular integrity and endothelial inflammation, underlie this loss of glomerular barrier function. In healthy murine and human kidney, Tie2 is heterogeneously expressed in all microvascular beds, although to different extents. In mice subjected to hemorrhagic and septic shock, Tie2 mRNA and protein were rapidly, and temporarily, lost from the renal microvasculature, and normalized within 24 h after initiation of the shock insult. The loss of Tie2 protein could not be attributed to shedding as both in mice and healthy volunteers subjected to endotoxemia, sTie2 levels in the systemic circulation did not change. In an attempt to identify the molecular control of Tie2, we activated glomerular endothelial cell cultures and human kidney slices in vitro with LPS or TNF-alpha, but did not observe a change in Tie2 mRNA levels. In parallel to the loss of Tie2 in vivo, an overt influx of neutrophils in the glomerular compartment, which coincided with proteinuria, was seen. As neutrophil-endothelial cell interactions may play a role in endothelial adaptation to shock, and these effects cannot be mimicked in vitro, we depleted neutrophils before shock induction. While this neutrophil depletion abolished proteinuria, Tie2 was not rescued, implying that Tie2 may not be a major factor controlling maintenance of the glomerular filtration barrier in this model.


Subject(s)
Capillary Permeability , Endothelium, Vascular/metabolism , Glomerular Filtration Rate , Kidney/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2/metabolism , Shock, Hemorrhagic/metabolism , Shock, Septic/metabolism , Aged , Albuminuria/metabolism , Albuminuria/physiopathology , Animals , Cell Line , Disease Models, Animal , Down-Regulation , Endothelium, Vascular/physiopathology , Humans , In Vitro Techniques , Kidney/blood supply , Kidney/physiopathology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Middle Aged , Neutrophils/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2/genetics , Shock, Hemorrhagic/physiopathology , Shock, Septic/chemically induced , Shock, Septic/physiopathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
4.
Am J Physiol Renal Physiol ; 294(3): F554-61, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18160627

ABSTRACT

E-selectin-directed targeted drug delivery was analyzed in anti-glomerular basement membrane glomerulonephritis. Liposomes conjugated with anti-E-selectin antibodies (Ab(Esel) liposomes) were internalized by activated endothelial cells in vitro through E-selectin-mediated endocytosis. At the onset of glomerulonephritis in mice, E-selectin was expressed on glomerular endothelial cells, which resulted in homing of Ab(Esel) liposomes to glomeruli after intravenous administration. Accumulation of Ab(Esel) liposomes in the kidney was 3.6 times higher than nontargeted IgG liposomes, whereas the accumulation of both liposomes in the clearance organs liver and spleen and in heart and lungs was comparable. In glomeruli, the Ab(Esel) liposomes colocalized with the endothelial cell marker CD31. Quantitative RT-PCR analysis of laser-microdissected arterioles, glomeruli, and postcapillary venules demonstrated that targeted delivery of dexamethasone by Ab(Esel) liposomes reduced glomerular endothelial expression of P-selectin, E-selectin, and vascular cell adhesion molecule-1 by 60-70%. The expression of these genes was not modulated in endothelial cells in nontargeted renal microvasculatures. Decrease of glomerular endothelial activation at disease onset was followed by reduced albuminuria at day 7. This study demonstrates the potential of vascular bed-specific drug delivery aimed at disease-induced epitopes on the microvascular endothelial cells as a therapeutic strategy for glomerulonephritis.


Subject(s)
Anti-Glomerular Basement Membrane Disease/drug therapy , Anti-Inflammatory Agents/administration & dosage , Dexamethasone/administration & dosage , E-Selectin/immunology , Animals , Anti-Glomerular Basement Membrane Disease/metabolism , Anti-Inflammatory Agents/pharmacokinetics , Antibodies/metabolism , Cell Line , Dexamethasone/pharmacokinetics , Drug Delivery Systems , E-Selectin/metabolism , Endothelial Cells/metabolism , Female , Gene Expression/drug effects , Immunoglobulin G/metabolism , Injections, Intravenous , Kidney/blood supply , Kidney Glomerulus/metabolism , Liposomes , Mice , Up-Regulation
5.
Mol Pharmacol ; 72(1): 121-31, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17452496

ABSTRACT

Glomerulonephritis represents a group of renal diseases with glomerular inflammation as a common pathologic finding. Because of the underlying immunologic character of these disorders, they are frequently treated with glucocorticoids and cytotoxic immunosuppressive agents. Although effective, use of these compounds has limitations as a result of toxicity and systemic side effects. In the current study, we tested the hypothesis that targeted delivery of dexamethasone (dexa) by immunoliposomes to activated glomerular endothelium decreases renal injury but prevents its systemic side effects. E-selectin was chosen as a target molecule based on its disease-specific expression on activated glomerular endothelium in a mouse anti-glomerular basement membrane glomerulonephritis. Site-selective delivery of Ab(Esel) liposome-encapsulated dexamethasone strongly reduced glomerular proinflammatory gene expression without affecting blood glucose levels, a severe side effect of administration of free dexamethasone. Dexa-Ab(Esel) liposomes reduced renal injury as shown by a reduction of blood urea nitrogen levels, decreased glomerular crescent formation, and down-regulation of disease-associated genes. Immunoliposomal drug delivery to glomerular endothelium presents a powerful new strategy for treatment of glomerulonephritis to sustain efficacy and prevent side effects of potent anti-inflammatory drugs.


Subject(s)
Dexamethasone/administration & dosage , E-Selectin/immunology , Endothelium, Vascular/drug effects , Glomerulonephritis/drug therapy , Immunoglobulin G/administration & dosage , Kidney Glomerulus/drug effects , Animals , Disease Progression , Female , Glomerular Basement Membrane/immunology , Liposomes , Mice , Mice, Inbred C57BL , Vascular Cell Adhesion Molecule-1/biosynthesis
6.
Microcirculation ; 13(5): 397-409, 2006.
Article in English | MEDLINE | ID: mdl-16815825

ABSTRACT

OBJECTIVES: The essential role of erythrocytes as oxygen carriers is historically well established, but their function to aggregate and the consequences on the microcirculation is under debate. The pathogenic potential of low erythrocyte aggregation could be important for patients undergoing on-pump cardipopulmonary bypass. These patients are severely hemodiluted due to preoperative isovolemic hemodilution (IHD), circuit priming, and large fluid infusions perioperatively. Considering the vascular endothelium sensitivity to variations in blood rheology, the authors hypothesize that low erythrocyte aggregation will be responsible for activation of vascular endothelium during acute IHD. METHODS: Acute IHD (30 mL/kg exchange transfusion with colloid solutions) was induced in an "aggregating species"(pigs, n = 15). The hypoxic oxidative stress (plasma malondialdehyde, ex vivo oxygen radicals production in heart, lung, kidney, liver, and ileum tissue biopsies), erythrocyte aggregation (LORCA), and endothelial activation (real-time quantitative RT-PCR on von Willebrand factor (vWF), E- and P-selectins, endothelial nitric oxide synthase gene-expression in tissue biopsies) were investigated. RESULTS: The production of superoxide and hydroxyl radicals, measured as H2O2 generation, was similar at all times in sham-operated and hemodiluted animals, proving a maintained oxygen delivery to tissues. Acute IHD was followed by a dramatic drop in erythrocyte aggregation and immediate prothrombotic (significant vWF mRNA upregulation in heart, lungs, kidney, liver, ileum) and proinflammatory (significant E- and P-selectins mRNA upregulation in lungs and ileum) endothelial activation. Low erythrocyte aggregation was significantly correlated with increased mRNA-expression of vWF (heart, liver, ileum) and P-selectin (lungs, ileum, and heart). CONCLUSIONS: These results suggest that low erythrocyte aggregation might trigger endothelium-dependent thrombogenic and proinflammatory response during acute isovolemic hemodilution.


Subject(s)
Endothelium, Vascular/physiopathology , Erythrocyte Aggregation , Hemodilution/adverse effects , Inflammation/etiology , Animals , Blood Coagulation , Blood Pressure , Blood Volume , Coronary Artery Bypass/adverse effects , E-Selectin/genetics , Heart Rate , Hemorheology , Humans , Malondialdehyde/blood , Models, Animal , Oxidative Stress , P-Selectin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
7.
Am J Physiol Cell Physiol ; 289(5): C1229-39, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15972838

ABSTRACT

Endothelial cells actively participate in inflammatory events by regulating leukocyte recruitment via the expression of inflammatory genes such as E-selectin, VCAM-1, ICAM-1, IL-6, IL-8, and cyclooxygenase (COX)-2. In this study we showed by real-time RT-PCR that activation of human umbilical vein endothelial cells (HUVEC) by TNF-alpha and IL-1beta differentially affected the expression of these inflammatory genes. Combined treatment with TNF-alpha and IL-1beta resulted in nonadditive, additive, and even synergistic induction of expression of VCAM-1, IL-8, and IL-6, respectively. Overexpression of dominant-negative inhibitor kappaB protein blocking NF-kappaB signaling confirmed a major role of this pathway in controlling both TNF-alpha- and IL-1beta-induced expression of most of the genes studied. Although dexamethasone exerted limited effects at 1 muM, the thioredoxin inhibitor MOL-294, which regulates the redox state of NF-kappaB, mainly inhibited adhesion molecule expression. Its most pronounced effect was seen on VCAM-1 mRNA levels, especially in IL-1beta-activated endothelium. One micromolar RWJ-67657, an inhibitor of p38 MAPK activity, diminished TNF-alpha- and IL-1beta-induced expression of IL-6, IL-8, and E-selectin but had little effect on VCAM-1 and ICAM-1. Combined treatment of HUVEC with MOL-294 and RWJ-67657 resulted in significant blocking of the expression of E-selectin, IL-6, IL-8, and COX-2. The inhibitory effects were much stronger than those observed with single drug treatment. Application of combinations of drugs that affect multiple targets in activated endothelial cells may therefore be considered as a potential new therapeutic strategy to inhibit inflammatory disease activity.


Subject(s)
Endothelial Cells/physiology , Interleukin-1/physiology , NF-kappa B/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Cell Adhesion Molecules/biosynthesis , Cyclooxygenase 2 , Dexamethasone/pharmacology , E-Selectin/biosynthesis , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , I-kappa B Proteins/metabolism , Imidazoles/pharmacology , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Membrane Proteins , Proline/analogs & derivatives , Proline/pharmacology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Pyridazines/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Thiocarbamates/pharmacology , Triazoles/pharmacology
8.
J Soc Gynecol Investig ; 12(2): 98-106, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15695104

ABSTRACT

OBJECTIVE: Systemic endothelial dysfunction is a central feature in the pathophysiology of preeclampsia. Its cell biologic and molecular basis is poorly understood. One leading hypothesis argues that endothelial dysfunction is caused by (at present largely unknown) circulating factors released from the ischemic placenta. This study investigated the effects of plasma factors of severe, early-onset preeclamptic women versus healthy pregnant women on endothelial gene expression in vitro. METHODS: Plasma samples were taken from eight severe early-onset preeclamptic women and eight matched pregnant control women. Primary human umbilical vein endothelial cell (HUVEC) and human glomerular microvascular endothelial cell (hGMEC) cultures were incubated with 20% (vol/vol) plasma for 4, 12, and 24 hours. Identical amounts of RNA isolated from HUVEC from three preeclamptic and three control samples were pooled for each time point, and subsequently hybridized on human 60-mer oligonucleotide microarrays containing 17,000 genes. Gene expression levels of vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), interleukin-8 (IL-8), and interleukin-6 (IL-6) in HUVEC and hGMEC were quantified using real-time reverse transcription polymerase chain reaction (RT-PCR). RESULTS: Microarray analyses of individual genes identified no genes that were up- or down-regulated more than 2.7-fold, and analyses of gene ontologies showed no gene ontology significantly up- or down-regulated in HUVEC by preeclamptic plasma. IL-8 gene expression was modestly induced by preeclamptic plasma after 4, 12, and 24 hours of HUVEC and hGMEC incubation, as identified by real-time RT-PCR. The other genes analyzed did not show altered regulation by preeclamptic plasma factors. CONCLUSIONS: In vitro, plasma from preeclamptic patients does not substantially alter endothelial gene expression profile. Only modest induction of IL-8 gene expression was observed. These results indicate that mechanisms other than soluble plasma constituents are likely involved in systemic endothelial cell activation in preeclampsia.


Subject(s)
Endothelium, Vascular/physiology , Intercellular Adhesion Molecule-1/blood , Interleukin-6/blood , Interleukin-8/blood , Pre-Eclampsia/blood , Vascular Cell Adhesion Molecule-1/blood , Adult , Antigens/metabolism , C-Reactive Protein/metabolism , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Profiling , Humans , Infant, Newborn , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Interleukin-8/biosynthesis , Interleukin-8/genetics , Oligonucleotide Array Sequence Analysis , Pre-Eclampsia/genetics , Pregnancy , Pregnancy Trimester, Second , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , von Willebrand Factor/immunology
9.
Curr Vasc Pharmacol ; 3(1): 11-39, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15638780

ABSTRACT

In chronic inflammatory conditions, endothelial cells actively recruit immune cells from the circulation into the underlying tissue and participate in angiogenesis to support the continuous demand for oxygen and nutrients. They do so in response to activation by cytokines and growth factors such as tumour necrosis factor alpha (TNFalpha), interleukin-1 (IL-1), vascular endothelial growth factor (VEGF), and fibroblast growth factors (FGFs). Receptor triggering initiates intracellular signal transduction leading to activation of nuclear factor kappaB (NFkappaB), mitogen activated protein kinase (MAPK) activity, and nitric oxide and reactive oxygen species production, among others. As a result, adhesion molecules, cytokines and chemokines, and a variety of other genes are being expressed that mediate and control the inflammatory process. In recent years, different classes of drugs have been developed that interfere with selected enzymes involved in the intracellular signalling cascades. In endothelial cell cultures, they exert potent inhibitory effects on the expression of genes, while several studies also report on in vivo effectiveness to confine the inflammatory responses. To prevent undesired toxicity and to improve drug behaviour and efficacy, drug carrier systems have been developed that selectively deliver the therapeutics into the activated endothelial cells. The above subjects are recapitulated to give an overview on the status of development of endothelial cell directed therapeutic strategies to pharmacologically interfere with chronic inflammatory diseases.


Subject(s)
Drug Delivery Systems/methods , Endothelial Cells/metabolism , Inflammation/metabolism , Mechanotransduction, Cellular/drug effects , Animals , Cells, Cultured , Chronic Disease , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Humans , Inflammation/etiology , Inflammation/pathology , Mechanotransduction, Cellular/genetics , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
10.
Mol Pharmacol ; 67(3): 883-90, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15598973

ABSTRACT

We prepared polyethylene glycol (PEG)-stabilized antisense oligonucleotide (ODN)/lipid particles from a lipid mixture including the positively charged amphiphile 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and anti-intercellular adhesion molecule 1 (ICAM-1) antisense ODN by an extrusion method in the presence of 40% ethanol. These particles were targeted to scavenger receptors on liver endothelial cells by means of covalently coupled polyanionized albumin. Two types of such targeted particles were prepared, one with the albumin coupled to a maleimide group attached to the particle's lipid bilayer and the other with the protein coupled to a maleimide group attached at the distal end of added bilayer-anchored PEG chains. Upon intravenous injection, the ODN particles with bilayer-coupled albumin were cleared from the blood circulation at the same low rate as untargeted particles (<5% in 30 min). By contrast, the distal-end coupled particles were very rapidly cleared from the blood and preferentially taken up by the endothelial cells of the hepatic sinusoid (55% of injected dose after 30 min). Despite this substantial endothelial targeting, no consistent inhibition of ICAM-1 expression could be demonstrated in this cell type, either in vivo or in vitro. However, in J774 cells that also express scavenger receptors and ICAM-1, significant down-regulation of ICAM-1 mRNA was achieved with distal-end targeted lipid particles, as determined with real-time RT-PCR. It is concluded that massive delivery of ODN to cell types that express scavenger receptors can be achieved if lipid particles are provided with negatively charged albumin distally attached to bilayer anchored PEG chains.


Subject(s)
Endothelial Cells/physiology , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/physiology , Liver/cytology , Oligonucleotides, Antisense/pharmacology , Animals , Cell Line , Fatty Acids, Monounsaturated/pharmacology , Kupffer Cells/physiology , Polyethylene Glycols , Quaternary Ammonium Compounds/pharmacology , Rats , Rats, Inbred Strains
11.
Biochem Pharmacol ; 65(10): 1729-39, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12754109

ABSTRACT

To deliver selectively anti-inflammatory agents into activated endothelial cells, drug-targeting conjugates were developed. Dexamethasone (Dexa) was covalently linked to a monoclonal antibody specifically recognizing E-selectin, which is strongly upregulated in endothelial cells at inflammatory sites. In the present study, the pharmacological effects of this Dexa-mouse antihuman E-selectin antibody (H18/7) (Ab(hEsel)) conjugate were investigated and compared to the effects obtained by free Dexa in human umbilical vein endothelial cells. Flow cytometry and ELISA were performed to analyze the levels of cell adhesion molecules (ICAM-1 and VCAM-1) and secreted cytokines (IL-6 and IL-8). The studies were extended by analysis of a complex gene expression pattern, using a cDNA expression array containing 268 genes encoding human cytokines/cytokine-receptors. Fifty genes and 28 genes were upregulated (ratio> or =2) upon incubation of human umbilical vein endothelial cells with TNFalpha for 6 and 24hr, respectively. This gene expression profile was markedly altered when cells were activated with TNFalpha in the presence of Dexa (100 nM) or Dexa-Ab(hEsel) conjugate (10 micro g/mL conjugate corresponding to 100 nM Dexa). Relative and competitive RT-PCR analysis verified downregulation of TNFalpha-mediated expression of CD40L and IL-8 by Dexa and Dexa-Ab(hEsel), respectively. These results indicated a successful internalization and processing of Dexa-Ab(hEsel) in activated endothelial cells, allowing the intracellularly delivered Dexa to exert its pleiotropic anti-inflammatory activity.


Subject(s)
Dexamethasone/pharmacology , Endothelium, Vascular/drug effects , Gene Expression/drug effects , Immunoconjugates/pharmacology , Animals , Biological Transport/drug effects , Cytokines/metabolism , Dexamethasone/administration & dosage , Dexamethasone/chemistry , Drug Delivery Systems , E-Selectin/immunology , Endothelium, Vascular/metabolism , Gene Expression Profiling , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-6/metabolism , Interleukin-8/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesis , Vascular Cell Adhesion Molecule-1/biosynthesis
12.
Pharm Res ; 20(1): 64-72, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12608538

ABSTRACT

PURPOSE: Drug targeting to activated endothelial cells is now being explored as a new approach to interfere with chronic inflammation. This study compares a dexamethasone-anti-E-selectin immunoconjugate (dexa-AbEsel) with anti-E-selectin immunoliposomes (AbEsel-immunoliposomes) that contain dexamethasone, regarding in vitro binding and internalization as well as in vivo accumulation in activated endothelial cells. METHODS: In vitro binding and internalization of dexa-AbEsel and the AbEsel-immunoliposomes into TNFalpha-activated HUVECs was studied using confocal laser scanning microscopy and radiolabeled compounds. Tissue accumulation of both compounds was studied in a murine delayed-type hypersensitivity model using immunohistochemistry. RESULTS AND CONCLUSIONS: Both preparations were selectively internalized by activated endothelial cells. Dexa-AbEsel was internalized by activated HUVECs to a larger extent than the AbEsel-immunoliposomes, although in theory the high drug-loading capacity of the liposomes may enable a larger amount of dexamethasone to be delivered intracellularly. Both dexa-AbEsel and AbEsel-immunoliposomes accumulated in activated endothelial cells in murine inflamed skin. AbEsel-immunoliposomes, but not dexa-AbEsel, were additionally detected in control skin, though to a lesser extent, and in macrophages of the liver and the spleen. Studies on therapeutic effects and side effects in models of chronic inflammation are now necessary to establish pharmacodynamics of dexa-AbEsel and/or AbEsel-immunoliposomes in the treatment of chronic inflammation.


Subject(s)
Drug Delivery Systems/methods , E-Selectin/administration & dosage , Endothelium, Vascular/drug effects , Immunoconjugates/administration & dosage , Inflammation/drug therapy , Animals , Cells, Cultured , E-Selectin/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Humans , Immunoconjugates/metabolism , Inflammation/immunology , Inflammation/pathology , Liposomes , Male , Mice , Mice, Inbred BALB C
13.
Pharm Res ; 19(11): 1730-5, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12458680

ABSTRACT

PURPOSE: For selective inhibition of endothelial cell activation in chronic inflammation, we have developed a dexamethasone-anti-E-selectin immunoconjugate. The present study was performed to evaluate the cellular handling of this immunoconjugate by activated primary endothelial cells and to compare its drug delivery capacity with free dexamethasone. METHODS: The binding, uptake, and degradation of 125I-radiolabeled dexamethasone-anti-E-selectin immunoconjugate by TNFalpha-activated endothelial cells were studied for different time periods and at different concentrations, as well as in the presence of inhibitors for E-selectin binding and lysosomal degradation. Its drug delivery capacity was compared with the uptake of unconjugated 3H-labeled dexamethasone. RESULTS: The immunoconjugate was internalized by E-selectin expressing activated endothelial cells and degraded in the lysosomal compartment. The receptor-mediated binding and uptake was saturable, implying a maximal attainable intracellular concentration of the drug. In contrast, free dexamethasone entered both resting and activated endothelial cells by passive diffusion. CONCLUSIONS: The dexamethasone-anti-E-selectin immunoconjugate is capable of selective delivering the coupled drug into activated endothelial cells. This targeting concept enables disease-induced drug delivery in which intracellular concentrations can be reached comparable with those obtained after incubation with 3 FM dexamethasone.


Subject(s)
Dexamethasone/pharmacokinetics , E-Selectin/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Immunoconjugates/pharmacokinetics , Cells, Cultured , Chemistry, Pharmaceutical , Humans
14.
Biotechnol Annu Rev ; 8: 133-65, 2002.
Article in English | MEDLINE | ID: mdl-12436918

ABSTRACT

Endothelial cells play a pathological role in cancer and chronic inflammation and are therefore attractive targets for therapeutic intervention. This review focuses on endothelial cell specific drug targeting strategies for the treatment of these diseases. The cellular and molecular processes involved in the activation of endothelial cells in angiogenesis and inflammation will be reviewed. Various target epitopes expressed by activated endothelium suitable for targeting purposes, design and development of drug-carrier complexes, drugs of interest which might interfere with endothelial cell activation, as well as in vitro and in vivo experimental approaches to study (intra) cellular drug delivery will be discussed.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Inflammation/drug therapy , Neoplasms/drug therapy , Chronic Disease , Drug Carriers , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Humans , Inflammation/immunology , Inflammation/physiopathology , Neoplasms/physiopathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology
15.
Bioconjug Chem ; 13(1): 128-35, 2002.
Article in English | MEDLINE | ID: mdl-11792188

ABSTRACT

Tumor blood vessels can be selectively targeted by RGD-peptides that bind to alpha(v)beta(3) integrin on angiogenic endothelial cells. By inhibiting the binding of these integrins to its natural ligands, RGD-peptides can serve as antiangiogenic therapeutics. We have prepared multivalent derivatives of the cyclic RGD-peptide c(RGDfK) by covalent attachment of the peptide to side chain amino groups of a protein. These RGDpep-protein conjugates inhibited alpha(v)beta(3)-mediated endothelial cell adhesion in vitro, while conjugates prepared with a control RAD-peptide showed no activity. Radiobinding and displacement studies with endothelial cells demonstrated an increased affinity of the RGDpep-protein conjugates compared to the free peptide, with IC(50) values ranging from 23 to 0.6 nM, depending on the amount of coupled RGDpep per protein. Compared to the parental RGD-peptide and the related RGD-peptide ligand c(RGDfV), the RGDpep-protein conjugates showed a considerable increase in affinity (IC(50) parent RGDpep: 818 nM; IC(50) c(RGDfV): 158 nM). We conclude that the conjugation of RGD-peptides to a protein, resulting in products that can bind multivalently, is a powerful approach to increase the affinity of peptide ligands for alpha(v)beta(3)/alpha(v)beta(5) integrins.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , Oligopeptides/chemistry , Proteins/chemistry , Proteins/pharmacology , Receptors, Vitronectin/drug effects , Cell Adhesion/drug effects , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Immunoglobulin G/chemistry , Peptides/chemistry
16.
J Immunol ; 168(2): 883-9, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11777986

ABSTRACT

In chronic inflammatory diseases, the endothelium is an attractive target for pharmacological intervention because it plays an important role in leukocyte recruitment. Hence, inhibition of endothelial cell activation and consequent leukocyte infiltration may improve therapeutic outcome in these diseases. We report on a drug targeting strategy for the selective delivery of the anti-inflammatory drug dexamethasone to activated endothelial cells, using an E-selectin-directed drug-Ab conjugate. Dexamethasone was covalently attached to an anti-E-selectin Ab, resulting in the so-called dexamethasone-anti-E-selectin conjugate. Binding of the conjugate to E-selectin was studied using surface plasmon resonance and immunohistochemistry. Furthermore, internalization of the conjugate was studied using confocal laser scanning microscopy and immuno-transmission electron microscopy. It was demonstrated that the dexamethasone-anti-E-selectin conjugate, like the unmodified anti-E-selectin Ab, selectively bound to TNF-alpha-stimulated endothelial cells and not to resting endothelial cells. After binding, the conjugate was internalized and routed to multivesicular bodies, which is a lysosome-related cellular compartment. After intracellular degradation, pharmacologically active dexamethasone was released, as shown in endothelial cells that were transfected with a glucocorticoid-responsive reporter gene. Furthermore, intracellularly delivered dexamethasone was able to down-regulate the proinflammatory gene IL-8. In conclusion, this study demonstrates the possibility to selectively deliver the anti-inflammatory drug dexamethasone into activated endothelial cells, using an anti-E-selectin Ab as a carrier molecule.


Subject(s)
Dexamethasone/pharmacology , E-Selectin/pharmacology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Immunoconjugates/pharmacology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Antibodies/chemistry , Antibodies/metabolism , Antibodies/pharmacology , Binding Sites, Antibody , Cells, Cultured , Dexamethasone/immunology , Dexamethasone/metabolism , Down-Regulation/immunology , E-Selectin/chemistry , E-Selectin/metabolism , Endothelium, Vascular/cytology , Genes, Reporter , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/biosynthesis , Surface Plasmon Resonance
17.
Microbiology (Reading) ; 141(6): 1281-1288, 1995 Jun.
Article in English | MEDLINE | ID: mdl-33799318

ABSTRACT

The Schizophyllum commune SC3 gene, which encodes a hydrophobin that coats aerial hyphae, is expressed in both monokaryons and dikaryons. The dikaryons were formed by mating two monokaryons with different MATA and MATB genes, leading to activation of the MATA- and MATB-controlled pathways (MATA-on and MATB-on). In contrast to the monokaryons, the dikaryons also expressed other hydrophobin genes (SC1, SC4) as well as a gene (SC7) encoding a hydrophilic wall protein. None of these four genes was expressed in MATA-off MATB-on mycelia, indicating that MATB-on represses SC3 and that both MATA-on and MATB-on are required for activation of SC1, SC4 and SC7. In fruiting dikaryons, immunolabelling revealed that SC3p was produced by aerial hyphae but not by hyphae that constitute the fruit-body tissue. In contrast to aerial hyphae, the latter produced dikaryon-specific transcripts and secreted SC7p into the extracellular matrix of the tissue. This suggests that in the aerial hyphae of the dikaryon the MATB-on pathway was not effective (MATB-off). We observed that in these aerial hyphae the two nuclei were wider apart than in a typical dikaryon. Although other explanations are not ruled out, we tentatively propose that effective interaction of different MATB genes requires proximity of the two nuclei containing these genes, and that disruption of this binucleate state represents a novel mechanism of gene control for spatial cell differentiation in the secondary mycelium.

SELECTION OF CITATIONS
SEARCH DETAIL
...