Network


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

Hotspot


Dive into the research topics where Marian T. Nakada is active.

Publication


Featured researches published by Marian T. Nakada.


Journal of Immunology | 2000

Antibodies Against the First Ig-Like Domain of Human Platelet Endothelial Cell Adhesion Molecule-1 (PECAM-1) That Inhibit PECAM-1-Dependent Homophilic Adhesion Block In Vivo Neutrophil Recruitment

Marian T. Nakada; Kunjlata M. Amin; Melpo Christofidou-Solomidou; Christopher D. O’Brien; Jing Sun; Indira Gurubhagavatula; George A. Heavner; Alexander H. Taylor; Cathy Paddock; Qi-Hong Sun; James L. Zehnder; Peter J. Newman; Steven M. Albelda; Horace M. DeLisser

Platelet endothelial cell adhesion molecule (PECAM-1), a member of the Ig superfamily, is found on endothelial cells and neutrophils and has been shown to be involved in the migration of leukocytes across the endothelium. Adhesion is mediated, at least in part, through binding interactions involving its first N-terminal Ig-like domain, but it is still unclear which sequences in this domain are required for in vivo function. Therefore, to identify functionally important regions of the first Ig-like domain of PECAM-1 that are required for the participation of PECAM-1 in in vivo neutrophil recruitment, a panel of mAbs against this region of PECAM-1 was generated and characterized in in vitro adhesion assays and in an in vivo model of cutaneous inflammation. It was observed that mAbs that disrupted PECAM-1-dependent homophilic adhesion in an L cell aggregation assay also blocked TNF-α-induced intradermal accumulation of neutrophils in a transmigration model using human skin transplanted onto SCID mice. Localization of the epitopes of these Abs indicated that these function-blocking Abs mapped to specific regions on either face of domain 1. This suggests that these regions of the first Ig-like domain may contain or be close to binding sites involved in PECAM-1-dependent homophilic adhesion, and thus may represent potential targets for the development of antiinflammatory reagents.


Journal of Immunology | 2000

Divergent effects of platelet-endothelial cell adhesion molecule-1 and beta 3 integrin blockade on leukocyte transmigration in vivo.

Richard Thompson; Matthew W. Wakelin; Karen Y. Larbi; Ann Dewar; George Asimakopoulos; Michael A. Horton; Marian T. Nakada; Sussan Nourshargh

The final stage in the migration of leukocytes to sites of inflammation involves movement of leukocytes through the endothelial cell layer and the perivascular basement membrane. Both platelet-endothelial cell adhesion molecule-1 (PECAM-1/CD31) and the integrin αvβ3 have been implicated in this process, and in vitro studies have identified αvβ3 as a heterotypic ligand for PECAM-1. In the present study we have addressed the roles of these molecules by investigating and comparing the effects of PECAM-1 and αvβ3 blockade on leukocyte migration in vivo. For this purpose we have examined the effects of neutralizing Abs directed against PECAM-1 (domain 1-specific, mAb 37) and β3 integrins (mAbs 7E3 and F11) on leukocyte responses in the mesenteric microcirculation of anesthetized rats using intravital microscopy. The anti-PECAM-1 mAb suppressed leukocyte extravasation, but not leukocyte rolling or firm adhesion, elicited by IL-1β in a dose-dependent manner (e.g., 67% inhibition at 10 mg/kg 37 Fab), but had no effect on FMLP-induced leukocyte responses. Analysis by electron microscopy suggested that this suppression was due to an inhibition of neutrophil migration through the endothelial cell barrier. By contrast, both anti-β3 integrin mAbs, 7E3 F(ab′)2 (5 mg/kg) and F11 F(ab′)2 (5 mg/kg), selectively reduced leukocyte extravasation induced by FMLP (38 and 46%, respectively), but neither mAb had an effect on IL-1β-induced leukocyte responses. These findings indicate roles for both PECAM-1 and β3 integrins in leukocyte extravasation, but do not support the concept that these molecules act as counter-receptors in mediating leukocyte transmigration.


Angiogenesis | 1999

Contortrostatin, a dimeric disintegrin from Agkistrodon contortrix contortrix, inhibits angiogenesis

Qing Zhou; Marian T. Nakada; Catherine Arnold; Kate Y. Shieh; S Francis MarklandJr.

Contortrostatin, a 13.5 kDa disulfide-linked homodimeric polypeptide possessing an Arg–Gly–Asp sequence, was isolated from venom of the southern copperhead snake. Daily injection of contortrostatin into the primary tumor of human breast cancer MDA-MB-435 carried in nude mice significantly inhibited tumor growth and neovascularization of the tumor tissue. On the chick embryo chorioallantoic membrane, contortrostatin inhibited angiogenesis induced by MDA-MB-435 cells, basic fibroblast growth factor, and vascular endothelial growth factor. In addition, contortrostatin effectively blocked adhesion of human umbilical vein endothelial cells (HUVEC) to immobilized vitronectin and significantly inhibited invasion of HUVEC through a Matrigel barrier. Competitive binding assays and adhesion assays with different integrin antibodies suggested that integrin αvβ3 is a binding site for contortrostatin on vascular endothelial cells. Detachment of HUVEC from vitronectin by contortrostatin induced apoptosis. HUVEC adhered and spread well on immobilized contortrostatin without undergoing apoptosis, suggesting that it is the inhibition of adhesion and spreading of HUVEC on extracellular matrix proteins, rather than binding of contortrostatin to integrins per se, that triggers apoptosis. We conclude that contortrostatin binds to αvβ3, and interferes with the anchorage-dependent survival mechanism of the vascular endothelial cells, and the mobility of the cells. The consequent suppression of angiogenesis is an important component of the antineoplastic activity of contortrostatin.


The FASEB Journal | 2011

PECAM-targeted delivery of SOD inhibits endothelial inflammatory response

Vladimir V. Shuvaev; Jingyan Han; Kevin Yu; Shaohui Huang; Brian J. Hawkins; Muniswamy Madesh; Marian T. Nakada; Vladimir R. Muzykantov

Elevated generation of reactive oxygen species (ROS) by endothelial enzymes, including NADPH‐oxidase, is implicated in vascular oxidative stress and endothelial proinflammatory activation involving exposure of vascular cell adhesion molecule‐1 (VCAM‐1). Catalase and superoxide dismutase (SOD) conjugated with antibodies to platelet/endothelial cell adhesion molecule 1 (PECAM‐1) bind specifically to endothelium and inhibit effects of corresponding ROS, H2O2, and superoxide anion. In this study, anti‐PECAM/SOD, but not anti‐PECAM/catalase or nontargeted enzymes, including polyethylene glycol (PEG)‐SOD, inhibited 2‐ to 3‐fold VCAM expression caused by tumor necrosis factor (TNF), interleukin‐1β, and lipopolysaccharide. Anti‐PECAM/SOD, but not nontargeted counterparts, accumulated in vascular endothelium after intravenous injection, localized in endothelial endosomes, and inhibited by 70% lipopolysaccharide‐caused VCAM‐1 expression in mice. Anti‐PECAM/SOD colocalized with EEA‐1‐positive endothelial vesicles and quenched ROS produced in response to TNF. Inhibitors of NADPH oxidase and anion channel ClC3 blocked TNF‐induced VCAM expression, affirming that superoxide produced and transported by these proteins, respectively, mediates inflammatory signaling. Anti‐PECAM/SOD abolished VCAM expression caused by poly(I:C)‐induced activation of toll‐like receptor 3 localized in intracellular vesicles. These results directly implicate endosomal influx of superoxide in endothelial inflammatory response and suggest that site‐specific interception of this signal attained by targeted delivery of anti‐PECAM/SOD into endothelial endo‐somes may have anti‐inflammatory effects.—Shuvaev, V. V., Han, J., Yu, K. J., Huang, S., Hawkins, B. J., Madesh, M., Nakada, M., and Muzykantov, V. R. PECAM‐targeted delivery of SOD inhibits endothelial inflammatory response. FASEB J. 25, 348–357 (2011). www.fasebj.org


Journal of Pharmacology and Experimental Therapeutics | 2007

Platelet-Endothelial Cell Adhesion Molecule-1-Directed Endothelial Targeting of Superoxide Dismutase Alleviates Oxidative Stress Caused by Either Extracellular or Intracellular Superoxide

Vladimir V. Shuvaev; Samira Tliba; Marian T. Nakada; Steven M. Albelda; Vladimir R. Muzykantov

Targeting of the antioxidant enzyme catalase to endothelial cells protects against vascular oxidative stress induced by hydrogen peroxide (H2O2)(Am J Physiol 285:L283–L292, 2003; Nat Biotechnol 21:392–398, 2003; Am J Physiol 293:L162–L169, 2007). However, another reactive oxygen species, superoxide anion, is also involved in many forms of vascular oxidative stress, including ischemia/reperfusion, hypertension, and inflammation. To protect endothelium against superoxide attack, we designed and tested antibody-directed targeting of superoxide dismutase (SOD) to the endothelial surface determinant, platelet-endothelial cell adhesion molecule (PECAM)-1. We synthesized anti-PECAM/SOD conjugates that retained 70% of enzymatic activity (superoxide anion dismutation) and specifically bound to endothelial cells, but not PECAM-negative cells. The effect of anti-PECAM/SOD delivery to cells was tested in two distinct models of oxidative stress induced by either extracellular or intracellular generation of superoxide anion. In the first model, anti-PECAM/SOD, but not unconjugated SOD, protected endothelial cells against injury caused by superoxide produced in the medium by hypoxanthine-xanthine oxidase. At the optimal dose, anti-PECAM/SOD provided up to 40 to 50% protection against cell death in this model. In the second model, anti-PECAM/SOD at the optimal dose provided complete protection against necrosis caused by paraquat-induced intracellular superoxide generation. Endothelial targeting of SOD represents a new molecular antioxidant approach that could be used for the management of vascular oxidative stress.


Free Radical Biology and Medicine | 2003

PECAM-directed immunotargeting of catalase: specific, rapid and transient protection against hydrogen peroxide.

Thomas Sweitzer; Anu Thomas; Rainer Wiewrodt; Marian T. Nakada; Francisco Branco; Vladimir R. Muzykantov

Vascular immunotargeting to Platelet-Endothelial Cell Adhesion Molecule-1 (PECAM) facilitates drug delivery to endothelium. We used human PECAM-transfected REN cells (REN/PECAM) as a model to compare targeting of antioxidant enzyme catalase conjugated with PECAM antibody (anti-PECAM/catalase) with adenoviral catalase delivery. Anti-PECAM/(125)I-catalase bound to REN/PECAM, but not to REN cells (70 vs. 1 ng/well vs. < 2 ng/well of unmodified catalase). At a virus-to-cell ratio of 1, elevated levels of catalase protein were detected by immunoblotting after adenoviral transfection of REN/PECAM and REN cells alike; H(2)O(2)-degrading activity of cell lysates was elevated at ratios of 10 and higher. REN/PECAM cells internalize 66% of cell-bound anti-PECAM/(125)I-catalase. Confocal microscopy localized anti-PECAM/catalase to intracellular vesicles, while catalase expressed by adenovirus was distributed in vesicles and throughout the cytosol. Within 15 min of delivery, anti-PECAM/catalase augmented H(2)O(2)-degrading activity and survival of H(2)O(2)-exposed REN/PECAM cells. The effects of conjugate delivery reached a plateau within 1 h and declined to the basal level within 12 h. In contrast, adenoviral delivery required several hours for transduction and development of the effects, but permitted much longer duration of protection (at least 48 h). Simultaneous exposure of REN/PECAM cells to anti-PECAM/catalase and catalase-encoding adenovirus afforded protection against H(2)O(2) with a rapid onset and a prolonged duration. Therefore, PECAM-directed immunotargeting provides a specific, antigen-directed intracellular delivery of catalase that affords a rapid but transient protection against H(2)O(2) and may complement gene delivery strategies for antioxidant protection.


Arteriosclerosis, Thrombosis, and Vascular Biology | 1998

Effects of β3-Integrin Blockade (c7E3) on the Response to Angioplasty and Intra-Arterial Stenting in Atherosclerotic Nonhuman Primates

Jonathan S. Deitch; J. Koudy Williams; Michael R. Adams; Christopher A. Fly; David M. Herrington; Robert E. Jordan; Marian T. Nakada; Joseph A. Jakubowski; Randolph L. Geary

Because the beta3-antagonist abciximab (c7E3 Fab) has significantly improved late outcomes after coronary angioplasty, the beta3 integrins have been implicated in the arterial response to injury. However, the mechanisms underlying this benefit are unknown. The observation that c7E3 binds beta3 integrins on vascular cells (alphavbeta3) with affinity equal to that for the platelet glycoprotein IIb/IIIa integrin has led to the hypothesis that c7E3 may act directly on the artery wall to prevent restenosis after angioplasty. To test this hypothesis, we studied the effects of c7E3 on structural changes within the artery wall after angioplasty or stent angioplasty in 23 male cynomolgus monkeys with established atherosclerosis. Animals were randomly assigned to receive either a bolus of c7E3 (0.4 mg/kg IV, n=11) followed by a 48-hour infusion (0. 2 microg. kg-1. min-1) or an equal volume of vehicle (n=12). Animals received weight-adjusted aspirin and heparin and then underwent unilateral iliac artery experimental angioplasty and subclavian artery stent angioplasty (Palmaz). Iliac artery lumen diameter (LD) was determined by angiography at baseline (LDPre), after angioplasty (LDPost), and 35 days later (LDDay35). Arteries were then fixed by perfusion and removed for analysis. Lumen, intima, media, and external elastic lamina (EEL) areas were measured in iliac artery cross sections. Values from each injured iliac artery were normalized to the contralateral uninjured iliac artery to control for interanimal variability in baseline artery size and atherosclerosis extent. Intimal area was also measured in subclavian stent cross sections. c7E3 blocked platelet aggregation and prolonged the bleeding time from 2.8+/-1.1 to 19.8+/-2.5 minutes, P<0.001. Experimental angioplasty increased LDPost an average of 28%, and the initial gain was similar in both groups (P=NS). Despite an anti-platelet effect, c7E3 did not inhibit iliac lumen narrowing (LDDay35-LDPost: c7E3, -0.69+/-0.17 versus vehicle, -0.99+/-.17 mm, P=0.35); intimal hyperplasia (neointima area: c7E3, 1.12+/-.28 versus vehicle, 1.22+/-.20 mm2, P=0.77); or decrease in artery wall size (EEL area [percent of uninjured control]: c7E3, 101+/-7% versus vehicle, 121+/-7%). Stent intimal hyperplasia was also unaltered by c7E3 treatment (neointimal area: c7E3, 1.09+/-0.16 versus vehicle, 1. 28+/-0.11 mm2, P=0.36). These results suggest that the benefits of c7E3 treatment in coronary angioplasty were not from inhibition of intimal hyperplasia or improved artery wall remodeling. Alternative mechanisms should be explored to explain improved late outcomes after angioplasty in patients treated with c7E3.


Angiogenesis | 2006

c7E3 Fab inhibits human tumor angiogenesis in a SCID mouse human skin xenograft model.

Marian T. Nakada; Gaoyuan Cao; Patricia M. Sassoli; Horace M. DeLisser

The αvβ3 integrin plays an important role in tumor growth and angiogenesis. Inhibition of this receptor by intact bivalent antibodies has been shown to inhibit angiogenesis and tumor growth. In this study we tested the chimeric Fab of 7E3 (c7E3 Fab), an antibody reactive with human platelet GPIIb/IIIa and αvβ3 to determine if it would inhibit in vivo angiogenesis and tumor growth in a SCID mouse/human skin tumor growth and angiogenesis model. c7E3 Fab inhibited human tumor angiogenesis and tumor growth. These data suggest monovalent antibody fragments devoid of antibody effector function can have efficacy in preclinical models of angiogenesis.


Journal of Immunology | 1997

Neutrophil Platelet Endothelial Cell Adhesion Molecule-1 Participates in Neutrophil Recruitment at Inflammatory Sites and Is Down-Regulated after Leukocyte Extravasation

Melpo Christofidou-Solomidou; Marian T. Nakada; John L. Williams; William A. Muller; Horace M. DeLisser


Biomaterials | 2008

Endothelial targeting of semi-permeable polymer nanocarriers for enzyme therapies

Thomas D. Dziubla; Vladimir V. Shuvaev; Nan Kang Hong; Brian J. Hawkins; Muniswamy Madesh; Hajime Takano; Eric Simone; Marian T. Nakada; Aron B. Fisher; Steven M. Albelda; Vladimir R. Muzykantov

Collaboration


Dive into the Marian T. Nakada's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Horace M. DeLisser

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven M. Albelda

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

George A. Heavner

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jing Sun

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aron B. Fisher

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catherine Arnold

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge