Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Juan-Carlos Murciano is active.

Publication


Featured researches published by Juan-Carlos Murciano.


Nature Biotechnology | 2003

Prophylactic fibrinolysis through selective dissolution of nascent clots by tPA-carrying erythrocytes

Juan-Carlos Murciano; Sandra Medinilla; Donald Eslin; Elena N. Atochina; Douglas B. Cines; Vladimir R. Muzykantov

A fibrinolytic agent consisting of a tissue-type plasminogen activator (tPA) coupled to the surface of red blood cells (RBCs) can dissolve nascent clots from within the clot, in a Trojan horse–like strategy, while having minimal effects on preexisting hemostatic clots or extravascular tissue. After intravenous injection, the fibrinolytic activity of RBC-tPA persisted in the bloodstream at least tenfold longer than did that of free tPA. In a model of venous thrombosis induced by intravenously injected fibrin microemboli aggregating in pulmonary vasculature, soluble tPA lysed pulmonary clots lodged before but not after tPA injection, whereas the converse was true for RBC-tPA. Free tPA failed to lyse occlusive carotid thrombosis whether injected before or after vascular trauma, whereas RBC-tPA circulating before, but not injected after, thrombus formation restored blood flow. This RBC-based drug delivery strategy alters the fibrinolytic profile of tPA, permitting prophylactic fibrinolysis.


Blood | 2010

Sustained thromboprophylaxis mediated by an RBC-targeted pro-urokinase zymogen activated at the site of clot formation

Sergei Zaitsev; Dirk Spitzer; Juan-Carlos Murciano; Bi-Sen Ding; Samira Tliba; M. Anna Kowalska; Oscar A. Marcos-Contreras; Alice Kuo; Victoria Stepanova; John P. Atkinson; Mortimer Poncz; Douglas B. Cines; Vladimir R. Muzykantov

Plasminogen activators (PAs) are used to treat life-threatening thrombosis, but not for thromboprophylaxis because of rapid clearance, risk of bleeding, and central nervous system (CNS) toxicity. We describe a novel strategy that may help to overcome these limitations by targeting a thrombin-activated PA pro-drug to circulating red blood cells (RBCs). We fused a single chain antibody (scFv Ter-119) that binds to mouse glycophorin A (GPA) with a variant human single-chain low molecular weight urokinase construct that can be activated selectively by thrombin (scFv/uPA-T). scFv/uPA-T bound specifically to mouse RBCs without altering their biocompatibility and retained its zymogenic properties until converted by thrombin into an active 2-chain molecule. As a result, RBC-bound scFv/uPA-T caused thrombin-induced fibrinolysis. One hour and 48 hours after intravenous (IV) injection in mice, approximately 70% and approximately 35% of scFv/uPA-T was retained in the blood, respectively, and approximately 95% of the circulating scFv/uPA-T remained bound to RBCs. A single IV injection of scFv/uPA-T provided effective prophylaxis against arterial and venous thrombosis for up to 24 hours. Thus, prophylactic delivery of RBC-targeted PA pro-drugs activated selectively at the site of clot formation represents a new approach to prevent thrombosis in clinical settings where the risk of clotting is high.


Circulation | 2008

Cerebrovascular Thromboprophylaxis in Mice by Erythrocyte-Coupled Tissue-Type Plasminogen Activator

Kristina Danielyan; Kumkum Ganguly; Bi-Sen Ding; Dmitriy N. Atochin; Sergei Zaitsev; Juan-Carlos Murciano; Paul L. Huang; Scott E. Kasner; Douglas B. Cines; Vladimir R. Muzykantov

Background— Cerebrovascular thrombosis is a major source of morbidity and mortality after surgery, but thromboprophylaxis in this setting is limited because of the formidable risk of perioperative bleeding. Thrombolytics (eg, tissue-type plasminogen activator [tPA]) cannot be used prophylactically in this high-risk setting because of their short duration of action and risk of causing hemorrhage and central nervous system damage. We found that coupling tPA to carrier red blood cells (RBCs) prolongs and localizes tPA activity within the bloodstream and converts it into a thromboprophylactic agent, RBC/tPA. To evaluate the utility of this new approach for preventing cerebrovascular thrombosis, we examined the effect of RBC/tPA in animal models of cerebrovascular thromboembolism and ischemia. Methods and Results— Preformed fibrin microemboli were injected into the middle carotid artery of mice, occluding downstream perfusion and causing severe infarction and 50% mortality within 48 hours. Preinjected RBC/tPA rapidly lysed nascent cerebral thromboemboli, providing rapid, durable reperfusion and reducing morbidity and mortality. These beneficial effects were not achieved by preinjection of tPA, even at a 10-fold higher dose, which increased mortality from 50% to 90% by 10 hours after embolization. RBC/tPA injected 10 minutes after tail amputation to simulate postsurgical hemostasis did not cause bleeding from the wound, whereas soluble tPA caused profuse bleeding. RBC/tPA neither aggravated brain damage caused by focal ischemia in a filament model of middle carotid artery occlusion nor caused postthrombotic hemorrhage in hypertensive rats. Conclusions— These results suggest a potential RBC/tPA utility as thromboprophylaxis in patients who are at risk for acute cerebrovascular thromboembolism.


The FASEB Journal | 2001

Cell-selective intracellular delivery of a foreign enzyme to endothelium in vivo using vascular immunotargeting

Arnaud Scherpereel; Rainer Wiewrodt; Melpo Christofidou-Solomidou; Radj Gervais; Juan-Carlos Murciano; Steven M. Albelda; Vladimir R. Muzykantov

Vascular immunotargeting, the administration of drugs conjugated with antibodies to endothelial surface antigens, has the potential for drug delivery to the endothelium. Our previous cell culture studies showed that biotinylated antibodies to PECAM‐1 (a highly expressed endothelial surface antigen) coupled with streptavidin (SA, a cross‐linking protein that facilitates anti‐PECAM internalization and targeting) may provide a carrier for the intracellular delivery of therapeutic enzymes. This paper describes the PECAM‐directed vascular immunotargeting of a reporter enzyme (β‐galactosidase, β‐Gal) in intact animals. Intravenous injection of [125I]SA‐β‐Gal conjugated with either anti‐PECAM or IgG led to a high 125I uptake in liver and spleen, yet β‐Gal activity in these organs rapidly declined to the background levels, suggesting rapid degradation of the conjugates. In contrast, anti‐PECAM/[125I]SA‐β‐Gal, but not IgG/[125I]SA‐β‐Gal, accumulated in the lungs (36.0±1.3 vs. 3.9±0.6% injected dose/g) and induced a marked elevation of β‐Gal activity in the lung tissue persisting for up to 8 h after injection (10‐fold elevation 4 h postinjection). Using histochemical detection, the β‐Gal activity in the lungs was detected in the endothelial cells of capillaries and large vessels. The anti‐PECAM carrier also provided 125I uptake and β‐Gal activity in the renal glomeruli. Predominant intracellular localization of anti‐PECAM/SA‐β‐Gal was documented in the PECAM‐expressing cells in culture by confocal microscopy and in the pulmonary endothelium by electron microscopy. Therefore, vascular immunotargeting is a feasible strategy for cell‐selective, intracellular delivery of an active foreign enzyme to endothelial cells in vivo, and thus may be potentially useful for the treatment of acute pulmonary or vascular diseases.—Scherpereel, A., Wiewrodt, R., Christofidou‐Solomidou, M., Gervais, R., Murciano, J.‐C., Albelda, S. M., Muzykantov, V. R. Cell‐selective intracellular delivery of a foreign enzyme to endothelium in vivo using vascular immunotargeting. FASEB J. 15, 416‐426 (2001).


American Journal of Pathology | 2002

Vascular Immunotargeting of Glucose Oxidase to the Endothelial Antigens Induces Distinct Forms of Oxidant Acute Lung Injury: Targeting to Thrombomodulin, But Not to PECAM-1, Causes Pulmonary Thrombosis and Neutrophil Transmigration

Melpo Christofidou-Solomidou; Stephen J. Kennel; Arnaud Scherpereel; Rainer Wiewrodt; Charalambos C. Solomides; Giuseppe G. Pietra; Juan-Carlos Murciano; Sayed A. Shah; Harry Ischiropoulos; Steven M. Albelda; Vladimir R. Muzykantov

Oxidative endothelial stress, leukocyte transmigration, and pulmonary thrombosis are important pathological factors in acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Vascular immunotargeting of the H(2)O(2)-generating enzyme glucose oxidase (GOX) to the pulmonary endothelium causes an acute oxidative lung injury in mice.(1) In the present study we compared the pulmonary thrombosis and leukocyte transmigration caused by GOX targeting to the endothelial antigens platelet-endothelial cell adhesion molecule (PECAM) and thrombomodulin (TM). Both anti-PECAM and anti-TM delivered similar amounts of (125)I-GOX to the lungs and caused a dose-dependent, tissue-selective lung injury manifested within 2 to 4 hours by high lethality, vascular congestion, polymorphonuclear neutrophil (PMN) sequestration in the pulmonary vasculature, severe pulmonary edema, and tissue oxidation, yet at an equal dose, anti-TM/GOX inflicted more severe lung injury than anti-PECAM/GOX. Moreover, anti-TM/GOX-induced injury was accompanied by PMN transmigration in the alveolar space, whereas anti-PECAM/GOX-induced injury was accompanied by PMN degranulation within vascular lumen without PMN transmigration, likely because of PECAM blockage. Anti-TM/GOX caused markedly more severe pulmonary thrombosis than anti-PECAM/GOX, likely because of TM inhibition. These results indicate that blocking of specific endothelial antigens by GOX immunotargeting modulates important pathological features of the lung injury initiated by local generation of H(2)O(2) and that this approach provides specific and robust models of diverse variants of human ALI/ARDS in mice. In particular, anti-TM/GOX causes lung injury combining oxidative, prothrombotic, and inflammatory components characteristic of the complex pathological picture seen in human ALI/ARDS.


Journal of Pharmacology and Experimental Therapeutics | 2010

Targeting of a mutant plasminogen activator to circulating red blood cells for prophylactic fibrinolysis.

Sergei Zaitsev; Dirk Spitzer; Juan-Carlos Murciano; Bi-Sen Ding; Samira Tliba; M. Anna Kowalska; Khalil Bdeir; Alice Kuo; Victoria Stepanova; John P. Atkinson; Mortimer Poncz; Douglas B. Cines; Vladimir R. Muzykantov

Chemical coupling to carrier red blood cells (RBCs) converts tissue type plasminogen activator (tPA) from a problematic therapeutic into a safe agent for thromboprophylaxis. The goal of this study was to develop a more clinically relevant recombinant biotherapeutic by fusing a mutant tPA with a single-chain antibody fragment (scFv) with specificity for glycophorin A (GPA) on mouse RBCs. The fusion construct (anti-GPA scFv/PA) bound specifically to mouse but not human RBCs and activated plasminogen; this led to rapid and stable attachment of up to 30,000 copies of anti-GPA scFv/PA per mouse RBC that were thereby endowed with high fibrinolytic activity. Binding of anti-GPA scFv/PA neither caused RBC aggregation, hemolysis, uptake in capillary-rich lungs or in the reticuloendothelial system nor otherwise altered the circulation of RBCs. Over 40% of labeled anti-GPA scFv/PA injected in mice bound to RBC, which markedly prolonged its intravascular circulation and fibrinolytic activity compared with its nontargeted PA counterpart, anti-GPA scFv/PA, but not its nontargeted PA analog, prevented thrombotic occlusion in FeCl3 models of vascular injury. These results provide proof-of-principle for the development of a recombinant PA variant that binds to circulating RBC and provides thromboprophylaxis by use of a clinically relevant approach.


Stroke | 2004

Mouse Model of Microembolic Stroke and Reperfusion

Dmitriy N. Atochin; Juan-Carlos Murciano; Yasemin Gursoy-Ozdemir; Tatiana Krasik; F. Noda; Cenk Ayata; Andrew K. Dunn; Michael A. Moskowitz; Paul L. Huang; Vladimir R. Muzykantov

Background and Purpose— To test the role of fibrinolysis in stroke, we used a mouse model in which preformed 2.5- to 3-μm-diameter fibrin microemboli are injected into the cerebral circulation. The microemboli lodge in the downstream precapillary vasculature and are susceptible to fibrinolysis. Methods— We injected various doses of microemboli into the internal carotid artery in mice and characterized their distribution, effects on cerebral blood flow, neurological deficit, infarct area, and spontaneous dissolution. By comparing wild-type and tissue plasminogen activator (tPA) knockout (tPA−/−) mice, we analyzed the role of endogenous tPA in acute thrombotic stroke. Results— Microemboli cause dose-dependent brain injury. Although moderate doses of microemboli are followed by spontaneous reperfusion, they result in reproducible injury. Gene knockout of tPA markedly delays dissolution of cerebral emboli and restoration of blood flow and aggravates ischemic thrombotic infarction in the brain. Conclusions— We describe a microembolic model of stroke, in which degree of injury can be controlled by the dose of microemboli injected. Unlike vessel occlusion models, this model can be modulated to allow spontaneous fibrinolysis. Application to tPA−/− mice supports a key role of endogenous tPA in restoring cerebral blood flow and limiting infarct size after thrombosis.


Journal of Pharmacology and Experimental Therapeutics | 2007

The Glycocalyx Protects Erythrocyte-Bound Tissue-Type Plasminogen Activator from Enzymatic Inhibition

Kumkum Ganguly; Juan-Carlos Murciano; Randal Westrick; John Leferovich; Douglas B. Cines; Vladimir R. Muzykantov

Coupling tissue-type plasminogen activator (tPA) to carrier red blood cells (RBC) prolongs its intravascular life span and permits its use for thromboprophylaxis. Here, we studied the susceptibility of RBC/tPA to PA inhibitors including plasminogen activator inhibitor-1 (PAI-1) that constrain its activity and may reduce the duration of its effect. Despite lesser spatial and diffusional limitations, soluble tPA was far less effective than RBC/tPA in dissolving clots formed in vitro from blood of wild-type (WT) mice (40 versus 80% lysis at equal doses of tPA). Furthermore, after i.v. injection, soluble tPA lost activity faster in transgenic mice expressing a high level of PAI-1 than in WT mice, whereas the activity of RBC/tPA was unaffected. PAI-1 inactivated soluble tPA at equimolar ratios in vitro, but it had no effect on the amidolytic or fibrinolytic activity of RBC/tPA. RBC/tPA was also more resistant than soluble tPA to in vitro inhibition by other serpins (α2-macroglobulin and α1-antitrypsin) and pathologically high levels of glucose. However, coupling to RBC did not protect a truncated tPA mutant, Retavase, from plasma inhibitors. Chemical removal of the RBC glycocalyx negated tPA protection from inhibitors: tPA coupled to glycocalyx-stripped RBC bound twice as much 125I-PAI-1 as did tPA coupled to naive RBC, and susceptibility of the bound tPA to inhibition by PAI-1 was restored. Thus, the RBC glycocalyx protects RBC-coupled tPA against inhibition. Resistance to high levels of inhibitors in vivo contributes to the potential utility of RBC/tPA for thromboprophylaxis.


Journal of Controlled Release | 2009

Soluble urokinase receptor conjugated to carrier red blood cells binds latent pro-urokinase and alters its functional profile.

Juan-Carlos Murciano; Abd Al-Roof Higazi; Douglas B. Cines; Vladimir R. Muzykantov

Coupling plasminogen activators to carrier red blood cells (RBC) prolongs their life-time in the circulation and restricts extravascular side effects, thereby allowing their utility for short-term thromboprophylaxis. Unlike constitutively active plasminogen activators, single chain urokinase plasminogen activator (scuPA) is activated by plasmin proteolysis or binding to its receptor, uPAR. In this study we conjugated recombinant soluble uPAR (suPAR) to rat RBC, forming RBC/suPAR complex. RBC carrying suPAR circulated in rats similarly to naïve RBC and markedly prolonged the circulation time of suPAR. RBC/suPAR carrying approximately 3x10(4) suPAR molecules per RBC specifically bound up to 2x10(4) molecules of scuPA, retained approximately 75% of scuPA-binding capacity after circulation in rats and markedly altered the functional profile of bound scuPA. RBC carrying directly conjugated scuPA adhered to endothelial cells, while showing no appreciable fibrinolytic activity. In contrast, RBC/suPAR loaded with scuPA did not exhibit increased adhesion to endothelium, while effectively dissolving fibrin clots. This molecular design, capitalizing on unique biological features of the interaction of scuPA with its receptor, provides a promising modality to deliver a pro-drug for prevention of thrombosis.


Journal of Pharmacology and Experimental Therapeutics | 2005

Fibrin Affinity of Erythrocyte-Coupled Tissue-Type Plasminogen Activators Endures Hemodynamic Forces and Enhances Fibrinolysis in Vivo

Kumkum Ganguly; Mukul S. Goel; Tatyana Krasik; Khalil Bdeir; Scott L. Diamond; Douglas B. Cines; Vladimir R. Muzykantov; Juan-Carlos Murciano

Plasminogen activators (PAs; e.g., tissue-type, tPA) coupled to red blood cells (RBCs) display in vivo features useful for thromboprophylaxis: prolonged circulation, minimal extravasation, and preferential lysis of nascent versus preexisting clots. Yet, factors controlling the activity of RBC-bound PAs in vivo are not defined and may not mirror the profile of soluble PAs. We tested the role of RBC/PA binding to fibrin in fibrinolysis. RBC/tPA and RBC/tPA variant with low fibrin affinity (rPA) bound to and lysed plasminogen-containing fibrin clots in vitro comparably. In contrast, when coinjected in mice with fibrin emboli lodging in pulmonary vasculature, only RBC/tPA accumulated in lungs, which resulted in a more extensive fibrinolysis versus RBC/rPA (p < 0.01). Reconciling this apparent divergence between in vitro and in vivo behaviors, RBC/tPA, but not RBC/rPA perfused over fibrin in vitro at physiological shear stress bound to fibrin clots and caused greater fibrinolysis versus RBC/rPA (p < 0.001). These results indicate that because of high fibrin affinity, RBC/tPA binding to clots endures hemodynamic stress, which enhances fibrinolysis. Behavior of RBC/PAs under hemodynamic pressure is an important predictor of their performance in vivo.

Collaboration


Dive into the Juan-Carlos Murciano's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas B. Cines

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kumkum Ganguly

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Khalil Bdeir

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Steven M. Albelda

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bi-Sen Ding

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Sergei Zaitsev

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Alice Kuo

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

M. Anna Kowalska

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge