1 Pulmonary Critical Care Division, Department of Medicine, 2 Department of Pathology, 4 Institute of Environmental Medicine, and 5 Department of Pharmacology, University of Pennsylvania Medical Center, Philadelphia 19104; and 3 Department of Pathology, Jefferson University, Philadelphia, Pennsylvania 19107
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Vascular immunotargeting is a novel approach for
site-selective drug delivery to endothelium. To validate the strategy,
we conjugated glucose oxidase (GOX) via streptavidin with antibodies to
the endothelial cell surface antigen platelet endothelial cell adhesion
molecule (PECAM). Previous work documented that 1)
anti-PECAM-streptavidin carrier accumulates in the lungs after
intravenous injection in animals and 2) anti-PECAM-GOX binds
to, enters, and kills endothelium via intracellular
H2O2 generation in cell culture. In the present work, we studied the targeting and effect of anti-PECAM-GOX in animals.
Anti-PECAM-GOX, but not IgG-GOX, accumulated in the isolated rat lungs,
produced H2O2, and caused endothelial injury
manifested by a fourfold elevation of angiotensin-converting enzyme
activity in the perfusate. In intact mice, anti-PECAM-GOX accumulated
in the lungs (27 ± 9 vs. 2.4 ± 0.3% injected dose/g for IgG-GOX) and caused severe lung injury and 95% lethality within hours after intravenous injection. Endothelial disruption and blebbing, elevated lung wet-to-dry ratio, and interstitial and alveolar edema indicated that anti-PECAM-GOX damaged pulmonary endothelium. The vascular injury
in the lungs was associated with positive immunostaining for
iPF2-III isoprostane, a marker for
oxidative stress. In contrast, IgG-GOX caused a minor lung injury and
little (5%) lethality. Anti-PECAM conjugated with inert proteins
induced no death or lung injury. None of the conjugates caused major
injury to other internal organs. These results indicate that an
immunotargeting strategy can deliver an active enzyme to selected
target cells in intact animals. Anti-PECAM-GOX provides a novel model
of oxidative injury to the pulmonary endothelium in vivo.
drug delivery; oxidative stress; CD31; platelet endothelial cell adhesion molecule-1; acute lung injury; hydrogen peroxide; isoprostane
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
CONJUGATION (either chemically or genetically) of effector compounds with carrier antibodies directed against endothelial surface antigens promises a novel strategy for site-specific delivery of drugs to endothelium (vascular immunotargeting) (2, 17, 25, 28, 40, 42). The pulmonary vasculature contains roughly one-third of the endothelial cells in the body, receives all the cardiac blood output, and, therefore, represents a privileged target organ. Antibodies directed against endothelial antigens accumulate in the lungs after intravenous injection in animals and permit pulmonary accumulation of the radiolabeled conjugated enzymes (2, 8, 19, 26, 27, 29, 33). However, neither the functional activity of the targeted enzymes nor the consequences of the targeting of antibody-enzyme conjugates to the pulmonary vasculature has been documented in intact animals. Demonstration of delivery of an active enzyme to pulmonary endothelium would, therefore, provide the final data needed to validate the strategy.
To provide this proof of principle for immunotargeting to the pulmonary endothelium, we conjugated glucose oxidase (GOX) with an antibody directed against a highly expressed endothelial surface antigen. GOX is a relatively stable enzyme that generates H2O2 from glucose, inducing oxidative stress in cells and tissues (31, 35). For example, GOX conjugated with collagen antibody caused skin lesions after intradermal injection in rats (24). GOX conjugated with antibodies to endothelial antigens caused oxidative injury in endothelial cell culture (22, 23). 125I-GOX conjugated with monoclonal antibody against angiotensin-converting enzyme (ACE) displayed preferential accumulation in the rat lungs after intravenous injection (29). However, neither functional activity of antibody-conjugated GOX in the lung nor potential manifestation of this activity has been characterized in vivo.
In the present study, to deliver GOX to endothelium in vivo, we used antibodies to platelet endothelial cell adhesion molecule (PECAM)-1 (anti-PECAM). PECAM-1 or CD31 is highly expressed on pulmonary vascular endothelium, whereas platelets and leukocytes express this antigen at much lower levels (9, 32). In a recent study, we found that a biotinylated anti-PECAM-streptavidin carrier bound to and entered endothelium in cell culture and accumulated preferentially in the lungs after intravenous injection in intact animals (28). Furthermore, biotinylated GOX conjugated with an anti-PECAM-streptavidin carrier (i.e., anti-PECAM-GOX) bound specifically to endothelial cells in culture, entered the target cells, and produced H2O2 intracellularly leading to cellular oxidative stress and death (15).
Based on these results, we postulated that intravenously injected anti-PECAM-GOX would 1) accumulate in the pulmonary endothelium, 2) generate H2O2, 3) induce oxidative stress in endothelium, and 4) cause vascular pulmonary injury. In this paper, we describe the pulmonary targeting of anti-PECAM-GOX in isolated perfused rat lungs and in intact mice. We report that systemic administration of anti-PECAM-GOX provides organ-selective targeting of an active enzyme that results in oxidative injury to the pulmonary vasculature, thus validating a novel drug delivery strategy.
![]() |
MATERIALS AND METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The following materials were used in the study: IODOGEN from Pierce
(Rockford, IL); Na125I from Amersham (Arlington Heights,
IL), fatty acid-free BSA from Boehringer Mannheim (Indianapolis, IN);
dimethyl formamide, 30% aqueous solution of
H2O2, mouse IgG, biotinylated glucose oxidase (b-GOX), biotinylated -galactosidase, biotinylated ferritin, and
components of buffer solutions from Sigma (St. Louis, MO); O-phthalaldehyde and Z-Phe-His-Leu from Serva
(Heidelberg, Germany); bovine liver catalase (20,000 U/mg) from Fluka
(Ronkonkoma, NY); and streptavidin (SA) and 6-biotinylaminocaproic acid
N-hydroxysuccinimide ester (BxNHS) from Calbiochem (San Diego,
CA). Protein concentration was determined by Bio-Rad microassay kit
(Hercules, CA). A monoclonal antibody to human PECAM-1 (MAb 62, a mouse
monoclonal IgG reacting with the first Ig-like loop of human and rat
PECAM-1) was kindly provided by Dr. Marian Nakada (Centocor, Malvern,
PA). Monoclonal antibody MAb 390 is a monoclonal antibody produced in
rat and reacting with murine PECAM-1 (45). A polyclonal rabbit
antiserum directed against isoprostane, iPF2
-III, was a
generous gift from Dr. Jacques Maclouf (INSERM Unit 398, Institute
Circulation-Lariboisiere, Hopital Lariboisiere, Paris, France).
Conjugation of glucose oxidase to anti-PECAM. Immunoglobulins were biotinylated at 10-fold molar excess of biotinylating reagent BxNHS as described previously (24, 26). In the following text, biotinylated proteins will be designated as b-IgG, b-MAb, or b-GOX. Biotinylated GOX was labeled with 125I using IODOGEN-coated tubes, according to the manufacturer's recommendation. To construct the trimolecular heteropolymer complex, b-anti-PECAM-SA-b-GOX or b-IgG-SA-b-GOX, as well as conjugates with other biotinylated enzymes (b-catalase), we used a two-step procedure established in our laboratory (24). Briefly, SA and b-GOX were mixed at a molar ratio of SA to b-GOX = 5 and incubated for 1 h on ice. This ratio is optimal for conjugation of SA-b-GOX complex with biotinylated immunoglobulins. The complex was then incubated with b-anti-PECAM or b-IgG to form b-anti-PECAM-SA-b-GOX conjugate or its nonimmune counterpart b-IgG-SA-b-GOX. These conjugates are indicated as anti-PECAM-GOX and IgG-GOX in the text. Enzymatic activity of GOX conjugated with either the immune or the nonimmune carrier did not differ from that of the initial preparation of biotinylated GOX (~100 U/mg).
Isolated perfused rat lungs. Isolated perfused rat lungs (IPL) were prepared as described (3). Briefly, male Sprague-Dawley rats (170-200 g) were anesthetized, the trachea was cannulated, and the lungs were ventilated with a humidified gas mixture containing 5% CO2-95% air. Ventilation was achieved with an SAR-830 rodent ventilator (CWE) at 60 cycles/min, a tidal volume of 2 ml, and 2 cmH2O end-expiratory pressure. After thoracotomy, the main pulmonary artery was cannulated through the transected heart. The lungs were then excised and transferred to the water-jacketed perfusion chamber maintained at 37°C. There was no interruption of ventilation during this transfer process, and interruption of lung perfusion was <5 s. Perfusion through the artery was maintained by a peristaltic pump at a constant flow rate of 10 ml/min. The perfusate (45 ml) was Krebs-Ringer buffer (KRB), pH 7.4, containing 10 mM glucose and 3% fatty acid-free BSA (KRB-BSA solution). The perfusate was filtered through a 0.4-µm filter before perfusion to eliminate particulates.
Determination of the pulmonary uptake of the conjugates in IPL. Perfusion of radiolabeled GOX conjugates was performed as previously described (2). Briefly, 1 µg of anti-PECAM-125I-GOX or IgG-125I-GOX was added to the perfusate and allowed to circulate for 1 h at 37°C. Nonbound conjugates were eliminated by 5-min nonrecirculating perfusion with conjugate-free buffer. Radioactivity in the lung tissue was determined in a gamma counter and calculated as a percent of the injected dose per gram of lung tissue (%ID/g).
Determination of ACE activity in the perfusates. ACE activity in the perfusates was measured by the rate of generation of His-Leu formed from the ACE substrate Z-Phe-His-Leu using a fluorometric assay (12). Ten microliters of the perfusate were added to 200 µl of 50 mM Tris · HCl-0.15 M NaCl, pH 8.3, buffer containing 0.5 mM substrate. Samples of perfusate were incubated at 37°C for 120 min, then the reaction was terminated by the addition of 1.5 ml of 0.28 N NaOH. O-phthalaldehyde (1 mg in 100 µl of methanol) was added for 10 min before the reaction was stopped by 2 N HCl. His-Leu was measured with a fluorescence spectrophotometer at an excitation wavelength of 363 nm and an emission wavelength of 500 nm. Results were calculated as milliunits of ACE activity per total perfusate (45 ml), where 1 mU represents the generation of 1 nmol His-Leu/min.
Detection of H2O2 generation in IPL. We used the oxidation-dependent fluorogen 2,7-dichlorofluorescein diacetate (DCF-DA; Molecular Probes, Eugene, OR) to detect generation of H2O2 in the lung tissue (1). This probe readily enters and becomes irreversibly entrapped intracellularly via cleavage by esterases. DCF-DA was perfused concomitantly with 100 µg of anti-PECAM-GOX or IgG-GOX in KRB-BSA for 1 h at 37°C to allow for accumulation of the conjugates and the probe. Glucose-free buffer was used to avoid massive formation of H2O2 in the perfusate by the circulating conjugates. Lungs were perfused for 2 h with glucose-containing KRB-BSA after elimination of nonbound material. DCF fluorescence (excitation 488 nm, emission 530 nm) was determined in the lung homogenates in a spectrofluorimeter and is expressed in arbitrary fluorescent units.
Evaluation of the pulmonary targeting and biodistribution of anti-PECAM-GOX in intact mice. Normal BALB/c mice were injected with 1 µg of anti-PECAM-125I-GOX or IgG-125I-GOX in 100 µl of physiological saline via the tail vein to characterize targeting of the conjugated GOX in vivo. One hour after injection, animals were killed, and the tissues were harvested. Radioactivity in the tissues was measured in a gamma counter. The tissue uptake of 125I-GOX was calculated as a percent of the injected dose per gram of lung tissue.
Assessment of pathological effect(s) caused by anti-PECAM-GOX in mice. To study effects of the conjugates in intact animals, we injected anesthetized BALB/c mice with 25-100 µg of anti-PECAM-GOX or control counterparts (IgG-GOX, nonconjugated anti-PECAM, anti-PECAM-ferritin, or anti-PECAM-catalase) via the tail vein. Surviving animals were killed 4-24 h after injection. Internal organs were inspected, photographed, and processed for cryosectioning, routine histological processing, or electron microscopy.
Histological evaluation of mouse organs. Tissue evaluation after treatment with anti-PECAM-GOX or IgG-GOX was performed on paraffin-embedded tissues. The lungs were instilled with 0.75 ml of buffered Formalin intratracheally with 20-gauge angiocatheters (Fisher) before removal from the animal. After excision, all organs (kidneys, liver, spleen, heart, and lung) were immersed in buffered Formalin overnight and processed for conventional paraffin histology. The sections were stained with hematoxylin and eosin and evaluated by light microscopy.
Immunohistochemical evaluation of mouse tissues.
Immunostaining was performed with 6-µm-thick frozen sections from
optimal cutting temperature-embedded tissues or from 4-µm paraffin
sections. The following antibodies were used: a rabbit polyclonal
antibody directed against iPF2-III, an isoprostane formerly known as 8-epi- or 8-iso-PGF2
(37, 38), and the
monoclonal antibody against GOX obtained from Sigma. Visualization was
achieved by the use of Vectastain kit or by alkaline phosphatase kit
(Vector Laboratories). For conventional light microscopy, the sections
were viewed with an Olympus II photomicroscope and for
immunofluorescence with a Zeiss fluorescent microscope.
Electron-microscopic processing of the lung tissue. This was performed on Epon-embedded tissues as described previously (7). For lung tissue, the trachea was exposed and cannulated with a 20-gauge angiocatheter. The lungs were instilled with cold 0.75 ml Karnowsky's fixative (2% paraformaldelyde and 2.5% glutaraldehyde; Electron Microscopy Sciences) in 0.1 M cacodylate buffer. They were immediately removed from the animal intact, the trachea was tied with a suture, and the entire tissue block was immersed in the same fixative for 30 min on ice. The lungs were then cut with a blade in 1-mm3 pieces, placed in fresh fixative, and left under vacuum (20 mmHg) overnight. After a rinse in cacodylate buffer, followed by a rinse in 0.05 M maleate buffer, pH 5.2, the tissue was postfixed with 1% osmium tetroxide in 0.7 M potassium ferrocyanide for 1 h on ice. En block staining was performed with uranyl acetate for 20 min, followed by dehydration in a graded series of alcohols and embedding in Poly/Bed 812 (Polysciences). Ultrathin sections (gold) were cut with a diamond knife (Diatome) on a Reichert-Jung (Vienna, Austria) ultramicrotome. Sections were collected on uncoated 150-mesh copper grids (Electron Microscopy Sciences) postcontrasted with 20% aqueous uranyl acetate (Amersham) for 3 min followed by aqueous 0.5% lead citrate for 3 min. The sections were viewed on a Hitachi H-600 transmission electron microscope (Nissey Sangyo) at 75 kV.
Statistical analysis. Statistical analysis was performed with a one-way ANOVA. When significant differences were found (P < 0.05), individual comparisons were made with the Bonferroni-Dunn test (Statview 4.0).
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Accumulation and effects of anti-PECAM-GOX in isolated rat lungs. In the first series of experiments, we studied the uptake and effects of anti-PECAM-GOX in isolated rat lungs perfused with blood-free buffer. This approach allowed us to examine the effects of conjugates in a simplified "ex vivo" system without confounding effects due to the conjugate action in other organs or its interactions with blood elements.
Pulmonary uptake of anti-PECAM-125I-GOX in the isolated rat lungs attained 20%ID/g, whereas that of IgG-125I-GOX did not exceed 0.5%ID/g (Fig. 1A). To detect H2O2 generated by anti-PECAM-GOX accumulated in the lung tissue, we used DCF-DA, a probe that forms a fluorescent dye in reaction with H2O2. Fluorescence in the tissue homogenates was markedly higher after perfusion of anti-PECAM-GOX than that after perfusion of IgG-GOX (Fig. 1B). We measured activity of ACE in the perfusate to characterize specifically endothelial injury by the conjugates. Release/shedding of ACE to the perfusate is a sensitive and cell-specific marker of endothelial injury in the lungs (3, 12). Anti-PECAM-GOX caused a marked elevation of ACE activity in the perfusate, whereas IgG-GOX had no effect (Fig. 1C) compared with the level of ACE activity in the perfusates of control lungs perfused with the buffer (shown as a dashed line in Fig. 1C).
|
Biodistribution and pulmonary uptake of anti-PECAM-GOX in intact animals. Based on the positive results obtained in an ex vivo model, we initiated a series of studies in intact animals. A trace amount of anti-PECAM-125I-GOX or IgG-125I-GOX was injected in the tail veins of BALB/c mice to characterize the immunotargeting of anti-PECAM-GOX in vivo. The aims of this study were to 1) compare the carrier, anti-PECAM-streptavidin, with other known carriers and 2) draw a correlation between uptake and the effect of the conjugate in different organs.
One hour after intravenous injection, the blood level of anti-PECAM-125I-GOX was similar to that of IgG-125I-GOX (2.9 ± 0.2 vs. 2.7 ± 0.1%ID/g). In contrast, pulmonary uptake of anti-PECAM-125I-GOX achieved 30%ID/g and was 10 times higher than that of IgG-125I-GOX (Fig. 2). The lung-to-blood ratio was 10.6 ± 1.6 for anti-PECAM-125I-GOX vs. 0.9 ± 0.1 for IgG-125I-GOX.
|
|
|
Effect of systemic administration of anti-PECAM-GOX in intact mice.
In the next series of experiments, we injected mice with 25-100
µg of anti-PECAM-GOX or IgG-GOX to determine the physiological effects of conjugate injections. Mice injected with saline served as
negative controls for this study. Because anti-endothelial antibodies
or conjugates could cause GOX-independent lung injury via complement-
and/or leukocyte-mediated pathways, we also studied the effects of
nonconjugated anti-PECAM as well as anti-PECAM conjugated to inert
proteins (catalase, -galactosidase, or ferritin). The goals of these
experiments were to 1) determine whether intravenous injection
of anti-PECAM-GOX causes lung injury, 2) characterize the
specificity of the injury (by comparing anti-PECAM-GOX with control
counterparts), 3) evaluate tissue selectivity of the injury (by
comparing lung with other organs), and 4) determine the
lethality, kinetics, and dose dependence of the effect of
anti-PECAM-GOX.
|
|
|
|
Characterization of the lung injury caused by anti-PECAM-GOX. The data shown above indicate that lung was the only organ where major differences between anti-PECAM-GOX and IgG-GOX occurred, both in terms of uptake and effect in the tissue. Based on these results, we focused on the lungs harvested from the mice injected with 100 µg of anti-PECAM-GOX or IgG-GOX to characterize more precisely the lung injury. The goals were to determine whether the endothelium was the primary site of anti-PECAM-GOX-mediated injury in vivo and whether the injury was associated with pulmonary oxidative stress.
First, we determined the lung wet-to-dry weight ratio. Anti-PECAM-GOX caused a significant elevation of the lung wet-to-dry weight ratio to 7.7 ± 0.7 (n = 8) vs. 4.5 ± 0.2 (n = 5) in the saline-injected control group (P < 0.05). IgG-GOX caused no elevation of the lung wet-to-dry weight ratio (4.5 ± 0.3, n = 5). Thus anti- PECAM-GOX caused an elevation of vascular permeability, an abnormality consistent with endothelial injury. Second, we used transmission electron microscopy to characterize the lung injury at cellular and subcellular levels. IgG-GOX induced no marked pathological alterations in the lungs (Fig. 8, panels 1-3). In contrast, severe vascular injury was present in the lungs after injection of anti-PECAM-GOX. Although alveolar injury was also noted, the pulmonary microvascular endothelium appeared to be the major target of injury. This conclusion is based on the following findings. First, anti-PECAM-GOX caused massive interstitial and alveolar edema (Fig. 9, panel 1). This finding is consistent with an elevated lung wet-to-dry ratio and implies that the conjugate increases vascular permeability by disruption of endothelial lining (Fig. 9, panel 2). Second, endothelial blebbing (microvesiculation), disruption of mitochondrial cristae, and swelling of their matrix were noted in the endothelium (Fig. 9, panel 3), consistent with oxidative cellular injury (20).
|
|
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Site-selective delivery of effector compounds (such as drugs, oligonucleotides, genes, toxins, or enzymes) to the vascular endothelium represents an important goal (2, 17, 25, 28, 40, 42). Enzymes are good candidates for immunotargeting for use in either experimental or therapeutic settings, since in contrast to other agents, even small amounts of active enzymes may produce an effect via generation of biologically active products when delivered to the target (36). Importantly, enzymes may work immediately after delivery, in contrast to genes, which require significant time for protein synthesis. Thus enzyme immunotargeting may afford a mechanism for effective, rapid, and site-selective interventions in endothelial cell function.
To our knowledge, results documented in this paper represent the first direct experimental evidence that systemic vascular administration of an enzyme conjugated with an antibody to endothelial surface antigen was able to provide site-selective delivery of an active enzyme to the pulmonary endothelium, causing a specific local effect in intact animals. Thus the present study verifies the validity and potential applicability of the strategy of vascular immunotargeting. In addition, this approach provides a novel model of oxidative vascular injury, a key component in pathogenesis of many pulmonary and cardiovascular diseases.
Although a number of endothelial antigens have been proposed for immunotargeting, our study focused on PECAM-1. In terms of the effectiveness (uptake of 30%ID/g), tissue selectivity (lung-to-blood ratio of 10), and immunospecificity (ratio between uptake values of the immune and nonimmune conjugates of 13), pulmonary targeting of anti-PECAM-GOX is similar to that of other radiolabeled compounds conjugated with other antiendothelial carrier antibodies, such as anti-ACE (26, 27) and anti-ICAM-1 (2). Immunotargeting to PECAM, however, offers potential advantages over other targeting systems. First, endothelial cells possess extremely high binding capacity for PECAM antibodies, and PECAM expression is not suppressed upon inflammation (32). Second, we have shown that enzymes conjugated with anti-PECAM enter endothelial cells, escape rapid intracellular degradation, and are enzymatically active (15, 28). Third, PECAM antibodies have been shown to inhibit transendothelial migration of leukocytes and thus may be advantageous when used in an anti-inflammatory therapeutic context (43). Therefore, PECAM-directed immunotargeting may be useful for the intracellular delivery of enzymes such as catalase or superoxide dismutase (for antioxidant protection), antisense oligonucleotides (for gene-selective suppression of protein synthesis), or plasmids (for specific stimulation of protein synthesis in the endothelial cells).
An important issue in immunotargeting relates to selectivity of delivery. Anti-PECAM-GOX caused primarily lung damage, consistent with the high pulmonary uptake of the conjugate. Injury was minimal in other organs (i.e., heart), consistent with the low tissue uptake of the conjugate in these organs. Similarly, the results of the survival study (Fig. 5) demonstrated that the injection of anti-PECAM-GOX is lethal, whereas IgG-GOX is well tolerated. However, the results of the biodistribution of anti-PECAM-125I-GOX and IgG-125I-GOX (Table 1) demonstrate that liver and spleen collect equally large doses of GOX after injection of either conjugate. This high hepatic and splenic uptake is similar to that observed with radiolabeled catalase, superoxide dismutase, or urokinase conjugated with carrier antibodies directed against PECAM, ACE, or intracellular adhesion molecule-1, as well as with their nonimmune counterparts (2, 26, 27). Interestingly, although subclinical damage may have occurred, data from postmortem gross examination, light microscopy, and electron microscopy showed no significant injury in liver and spleen despite a very high uptake of the conjugates. This high hepatic and splenic uptake of IgG-GOX taken together with a low lethality of mice injected with IgG-GOX indicates that the liver and spleen are resistant to GOX. This is likely due to the fact that most of the uptake in these organs is probably by Fc receptor-bearing macrophages, Kuppfer cells, or other phagocytic cells. The phagocytes may degrade the conjugates and/or be resistant to oxidants. Accordingly, we view our results as showing lung "selective" (rather than "specific") delivery of an active enzyme, GOX.
We are currently performing studies with anti-PECAM--galactosidase
conjugate, which causes no injury in animals but allows one to trace
directly the enzymatic activity of the conjugates in vivo (A. Sherperel, R. Wiewrodt, M. Christofidou-Solomidou, R. Gervais, S. Albelda, and V. Muzykantov, unpublished data). We detected significant
uptake and activity of
-galactosidase in the liver 1 h after
injection of either IgG-
-galactosidase or
anti-PECAM-
-galactosidase. Interestingly,
-galactosidase activity
in the liver declined extremely rapidly and was indistinguishable from
the background level as soon as 4 h after injection. In sharp contrast,
4 h after injection of anti-PECAM-
-galactosidase, the enzyme
activity in the lungs declined by only 30% and was still 10 times
higher than the background level. These new data show that the
anti-PECAM-
-galactosidase conjugate undergoes extremely rapid
degradation in the liver (probably within Kuppfer cells), but not in
the lungs. Therefore, we believe that the lack of anti-PECAM-GOX hepatotoxicity can be explained by different metabolism/resistance of
the anti-PECAM-GOX in the lung and liver.
In this study, we used anti-PECAM antibodies to target a
H2O2-generating enzyme, GOX.
H2O2 forms the strong oxidants hydroxyl anion
(in reactions with metals or with superoxide anion) and hypochlorous
acid (in reaction catalyzed by myeloperoxidase), which cause oxidative
stress in the cells and tissues (16). GOX has been used in previous in
vitro and in vivo studies, where local administration of enzyme led to
oxidant generation (21, 31, 35). The morphological characteristics of
the lung injury caused by anti-PECAM-GOX (endothelial blebbing and
disruption), elevation of the pulmonary vascular permeability,
and release of ACE imply that the pulmonary endothelium is the primary
site of injury. Elevated fluorescence of a
H2O2-sensitive probe and positive
immunostaining for iPF2-III isoprostane in the lung tissue indicate that oxidative injury to endothelium caused by H2O2 is likely a mechanism for the injury.
In addition to demonstrating the validity of vascular immunotargeting, anti-PECAM-GOX offers a new model of endothelial oxidative stress. Endothelial oxidative injury has been implicated in many types of lung pathology including ischemia-reperfusion injury, postcardiopulmonary bypass lung injury, sepsis, and the adult respiratory distress syndrome (1, 6, 14, 41). Studies in cell cultures, in isolated organs, and in vivo indicate that H2O2 is one of the key reactive oxygen species involved both in cell signaling, host defense, and oxidative injury in inflammation, infarction, and vascular injury (11, 16, 30). Endothelial oxidative stress caused by either extracellular reactive oxygen species (e.g., released from activated neutrophils) or intracellular reactive oxygen species (e.g., generated within endothelium upon ischemia-reperfusion) plays an important role in pathogenesis of cardiovascular and lung disease conditions (5, 6, 11, 18). Anti-PECAM-GOX could be useful to address mechanisms of pulmonary and vascular oxidative injury caused by or associated with a site-specific intracellular generation of the well-defined oxidant H2O2 in models ranging from cell culture to experiments in laboratory animals. Animal models based on GOX immunotargeting may help to dissect the primary (initiating) role of reactive oxygen species generated in endothelium and secondary (augmenting) role of oxidants released by leukocytes attracted to the site of the vascular injury. This area is under active investigation in our laboratory.
In conclusion, our results provide direct experimental evidence that vascular immunotargeting of enzymes conjugated with antibodies directed against surface endothelial antigens provides a mechanism for site-selective interventions in endothelial functions. Immunotargeting of glucose oxidase may be useful for investigation of the oxidative vascular injury and for tumor eradication. Conjugation of other potentially therapeutic compounds (such as antioxidant enzymes) with anti-endothelial antibodies could be useful in the treatment of a variety of pulmonary and vascular diseases caused by or associated with endothelial cell dysfunction.
![]() |
ACKNOWLEDGEMENTS |
---|
We thank Mildred Daise for antibody purification and Melanie Minda for
help with the electron microscopy studies. We thank Dr. Marian Nakada
for MAb 62, reacting with rat PECAM-1. We greatly appreciate a generous
gift of the antibody directed against iPF2-III isoprostane provided by Dr. Jacques Maclouf and with this paper honor
his memory. We thank Drs. Aron Fisher, Harry Ischiropoulos, and Andrew
Gow for discussions, reading of the manuscript, and comments.
![]() |
FOOTNOTES |
---|
This study was supported by National Heart, Lung, and Blood Institute Specialized Center of Research in Acute Lung Injury Grant HL-60290 (Project 4) and by American Heart Association Established Investigator Grant 9640204N (to V. R. Muzykantov).
The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. §1734 solely to indicate this fact.
Address for reprint requests and other correspondence: V. R. Muzykantov, Institute of Environmental Medicine, Univ. of Pennsylvania Medical Center, 1 John Morgan Bldg., 36th St. and Hamilton Walk, Philadelphia, PA 19104-6068 (E-mail: muzykant{at}mail.med.upenn.edu).
Received 8 March 1999; accepted in final form 1 November 1999.
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1.
Al-Mehdi, A,
Shuman H,
and
Fisher AB.
Fluorescence microphotography of oxidative stress in lung ischemia-reperfusion.
Lab Invest
70:
579-587,
1994[ISI][Medline].
2.
Atochina, EN,
Balyasnikova IV,
Danilov SM,
Granger DN,
Fisher AB,
and
Muzykantov VR.
Immunotargeting of catalase to ACE or ICAM-1 protects perfused rat lungs against oxidative stress.
Am J Physiol Lung Cell Mol Physiol
275:
L806-L817,
1998
3.
Atochina, EN,
Muzykantov VR,
Al-Mehdi AB,
Danilov SM,
and
Fisher AB.
Normoxic lungs ischemia/reperfusion accelerates shedding of angiotensin converting enzyme from the pulmonary endothelium.
Am J Respir Crit Care Med
156:
1114-1119,
1997
4.
Becker, PM,
Sanders SP,
and
Christman BW.
F2-isoprostane generation in isolated ferret lungs after oxidant injury or ventilated ischemia.
Free Radic Biol Med
25:
703-711,
1998[ISI][Medline].
5.
Brigham, K,
and
Meyrick B.
Endotoxin and lung injury.
Am Rev Respir Dis
133:
913-927,
1986[ISI][Medline].
6.
Chabot, F,
Mitchell JA,
Gutteridge JM,
and
Evans TW.
Reactive oxygen species in acute lung injury.
Eur Respir J
11:
745-757,
1998
7.
Christofidou-Solomidou, M,
Longley J,
Whitaker-Menezes D,
Albelda SM,
and
Murphy GF.
Human skin/SCID mouse chimeras as an in vivo model to study human cutaneous mastocytosis.
J Invest Dermatol
109:
102-107,
1997[Abstract].
8.
Danilov, S,
Muzykantov V,
Martynov A,
Atochina E,
Sakharov I,
Trakht I,
and
Smirnov V.
Lung is a target organ for monoclonal antibody to ACE.
Lab Invest
64:
118-124,
1991[ISI][Medline].
9.
DeLisser, HM,
Newman PJ,
and
Albelda SM.
Molecular and functional aspects of PECAM-1/CD31.
Immunol Today
15:
490-495,
1994[ISI][Medline].
10.
Fisher, AB,
Dodia C,
Tan Z,
Ayene I,
and
Eckenhoff RG.
Oxygen-dependent lipid peroxidation during lung ischemia.
J Clin Invest
88:
674-679,
1991[ISI][Medline].
11.
Freeman, B,
and
Crapo J.
Free radicals and tissue injury.
Lab Invest
47:
412-426,
1987[ISI][Medline].
12.
Friedland, J,
and
Silverstein E.
A sensitive fluorimetric assay for serum ACE.
Am J Clin Pathol
66:
416-424,
1976[ISI][Medline].
13.
Fulkerson, WJ,
MacIntyre N,
Stamler J,
and
Crapo JD.
Pathogenesis and treatment of the acute respiratory distress syndrome.
Arch Intern Med
156:
29-38,
1996[Abstract].
14.
Gibbons, G,
and
Dzau V.
Molecular therapies for vascular diseases.
Science
272:
689-693,
1996[Abstract].
15.
Gow, A,
Branco F,
Christofidou-Solomidou M,
Schultz L,
Albelda S,
and
Muzykantov V.
Immunotargeting of glucose oxidase: intracellular production of H2O2 and endothelial oxidative stress.
Am J Physiol Lung Cell Mol Physiol
277:
L271-L281,
1999
16.
Halliwell, B,
and
Gutteridge J.
Oxygen free radicals and iron in relation to biology and medicine: some problems and concepts.
Arch Biochem Biophys
246:
501-514,
1986[ISI][Medline].
17.
Huang, X,
Molema G,
King S,
Watkins L,
Edgington TS,
and
Thorpe PE.
Tumor infarction in mice by antibody-directed targeting of tissue factor to tumor vasculature.
Science
275:
547-550,
1997
18.
Kehrer, JP.
Free radicals as mediators of tissue injury and disease.
Crit Rev Toxicol
23:
21-48,
1993[ISI][Medline].
19.
Kennel, S,
Lee R,
Bultman S,
and
Kabalka G.
Rat monoclonal antibody distribution in mice: an epitope inside the lung vascular space mediates very efficient localization.
Int J Rad Appl Instrum B
17:
193-200,
1990[Medline].
20.
McIntyre, TM,
Modur V,
Prescott SM,
and
Zimmerman GA.
Molecular mechanisms of early inflammation.
Thromb Hemostasis
78:
302-305,
1997[ISI][Medline].
21.
Muzykantov, V,
Peklo M,
and
Printzeva O.
Local tissue injury induced by glucose oxidase conjugated with anti-collagen antibody.
J Immunol Methods
119:
65-73,
1989[ISI][Medline].
22.
Muzykantov, V,
Sakharov D,
Sinitsyn V,
Goncharov N,
and
Danilov S.
Specific killing of human endothelial cells by antibody-conjugated glucose oxidase.
Anal Biochem
169:
383-389,
1988[ISI][Medline].
23.
Muzykantov, V,
Trubetskaja O,
Puchnina E,
Sakharov D,
and
Domogatsky S.
Cytotoxicity of glucose oxidase conjugated with antibodies to target cells: efficiency of killing depends on the conjugate internalization.
Biochim Biophys Acta
1053:
27-31,
1990[ISI][Medline].
24.
Muzykantov, VR.
Conjugation of catalase to a carrier antibody via streptavidin-biotin cross-linker.
Biotechnol Appl Biochem
26:
103-109,
1997[ISI][Medline].
25.
Muzykantov, VR.
Immunotargeting of drugs to the pulmonary vascular endothelium as a therapeutic strategy.
Pathophysiology
5:
15-33,
1998.
26.
Muzykantov, VR,
Atochina E,
Ischiropoulos H,
Danilov S,
and
Fisher AB.
Immunotargeting of antioxidant enzymes to the pulmonary endothelium.
Proc Natl Acad Sci USA
93:
5213-5218,
1996
27.
Muzykantov, VR,
Barnathan ES,
Atochina E,
Kuo A,
Danilov S,
and
Fisher AB.
Targeting of antibody-conjugated plasminogen activators to the pulmonary vasculature.
J Pharmacol Exp Ther
279:
1026-1034,
1996[Abstract].
28.
Muzykantov, VR,
Christofidou-Solomidou M,
Balyasnikova I,
Harshaw D,
Schultz L,
Fisher A,
and
Albelda S.
Streptavidin facilitates internalization and pulmonary uptake of an anti-endothelial cell antibody (PECAM-1): a strategy for vascular immunotargeting of drugs.
Proc Natl Acad Sci USA
96:
2379-2384,
1999
29.
Muzykantov, VR,
Martynov AV,
Puchina EA,
and
Danilov SM.
In vivo administration of glucose oxidase conjugated with monoclonal antibodies to angiotensin-converting enzyme.
Am Rev Respir Dis
139:
1464-1473,
1989[ISI][Medline].
30.
Natarajan, V.
Oxidants and signal transduction in vascular endothelium.
J Lab Clin Med
125:
26-37,
1995[ISI][Medline].
31.
Nathan, C,
and
Cohn Z.
Antitumor effects of hydrogen peroxide in vivo.
J Exp Med
154:
1539-1553,
1981
32.
Newman, PJ.
The biology of PECAM.
J Clin Invest
99:
3-7,
1997
33.
Panes, J,
Perry M,
Anderson D,
Muzykantov V,
Garden D,
Miyasaka M,
and
Granger D.
Portal hypertension enhances endotoxin-induced ICAM-1 up- regulation in the rat.
Gastroenterology
110:
866-874,
1996[ISI][Medline].
34.
Patrono, C,
and
FitzGerald G.
Isoprostanes: potential markers of oxidant stress in atherothrombotic disease.
Arterioscler Thromb Vasc Biol
17:
2309-2315,
1997
35.
Philpott, GW,
Kulczycky A,
Grass EH,
and
Parker CW.
Selective binding and cytotoxicity of rat basophyllic leukemia cells with immunoglobulin E-biotin and avidin-glucose oxidase conjugates.
J Immunol
125:
1201-1208,
1980
36.
Poznansky, MJ,
and
Juliano RL.
Biological approaches to the controlled delivery of drugs: a critical review.
Pharmacol Rev
36:
277-334,
1984[ISI][Medline].
37.
Pratico, D,
Iuliano L,
Mauriello A,
Spagnoli L,
Lawson JA,
Maclouf J,
Violi F,
and
FitzGerald GA.
Localization of distinct F-2-isoprostanes in human atherosclerotic lesions.
J Clin Invest
100:
2028-2034,
1997
38.
Pratico, D,
Tangilla R,
Rader D,
Rokach J,
and
FitzGerald GA.
Vitamin E suppresses isoprostane generation and reduced atherosclerosis.
Nat Med
4:
1189-1192,
1992.
39.
Rokach, J,
Khanapure SP,
Hwang S-W,
Adiyaman M,
Lawson JA,
and
FitzGerald GA.
Nomenclature of isoprostanes: a proposal.
Prostaglandins
54:
853-873,
1997[ISI][Medline].
40.
Schnitzer, JE.
Vascular targeting as a strategy for cancer therapy.
N Engl J Med
339:
472-474,
1998
41.
Thomas, JR,
Harlan JM,
Rice CL,
and
Winn RK.
Role of leukocyte CD11/CD18 complex in endotoxic and septic shock in rabbits.
J Appl Physiol
73:
1510-1516,
1992
42.
Trubetskoy, V,
Torchilin V,
Kennel S,
and
Huang L.
Cationic liposomes enhance targeted delivery and expression of exogenous DNA mediated by N-terminal modified poly-(L-lysine)-antibody conjugate in mouse lung endothelial cells.
Biochim Biophys Acta
1131:
311-313,
1992[ISI][Medline].
43.
Vaporciyan, AA,
Delisser HM,
Yan H-C,
Mendiguren I,
Thom SR,
Jones ML,
Ward PA,
and
Albelda SM.
Involvement of PECAM-1 in neutrophil recruitment in vivo.
Science
262:
1580-1582,
1993[ISI][Medline].
44.
Wang, Z,
Ciabattoni G,
Creminon C,
Lawson J,
FitzGerald GA,
Patrono C,
and
Maclouf J.
Immunological characterization of urinary 8-epi-prostaglandin PGF2 excretion in man.
J Pharmacol Exp Ther
275:
94-100,
1995[Abstract].
45.
Yan, H,
Baldwin S,
Sun J,
Buck C,
Albelda SM,
and
DeLisser M.
Alternative splicing of a specific cytoplasmic exon alters the binding characteristics of mouse PECAM-1.
J Biol Chem
270:
23672-23680,
1995