Network


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

Hotspot


Dive into the research topics where Raisa Yu. Kiseleva is active.

Publication


Featured researches published by Raisa Yu. Kiseleva.


Journal of Controlled Release | 2018

Targeting superoxide dismutase to endothelial caveolae profoundly alleviates inflammation caused by endotoxin

Vladimir V. Shuvaev; Raisa Yu. Kiseleva; Evguenia Arguiri; Carlos H. Villa; Silvia Muro; Melpo Christofidou-Solomidou; Radu V. Stan; Vladimir R. Muzykantov

ABSTRACT Inflammatory mediators binding to Toll‐Like receptors (TLR) induce an influx of superoxide anion in the ensuing endosomes. In endothelial cells, endosomal surplus of superoxide causes pro‐inflammatory activation and TLR4 agonists act preferentially via caveolae‐derived endosomes. To test the hypothesis that SOD delivery to caveolae may specifically inhibit this pathological pathway, we conjugated SOD with antibodies (Ab/SOD, size ∽ 10 nm) to plasmalemmal vesicle‐associated protein (Plvap) that is specifically localized to endothelial caveolae in vivo and compared its effects to non‐caveolar target CD31/PECAM‐1. Plvap Ab/SOD bound to endothelial cells in culture with much lower efficacy than CD31 Ab/SOD, yet blocked the effects of LPS signaling with higher efficiency than CD31 Ab/SOD. Disruption of cholesterol‐rich membrane domains by filipin inhibits Plvap Ab/SOD endocytosis and LPS signaling, implicating the caveolae‐dependent pathway(s) in both processes. Both Ab/SOD conjugates targeted to Plvap and CD31 accumulated in the lungs after IV injection in mice, but the former more profoundly inhibited LPS‐induced pulmonary inflammation and elevation of plasma level of interferon‐beta and ‐gamma and interleukin‐27. Taken together, these results indicate that targeted delivery of SOD to specific cellular compartments may offer effective, mechanistically precise interception of pro‐inflammatory signaling mediated by reactive oxygen species. Graphical abstract Figure. No Caption available.


Advanced Materials | 2018

Flexible Nanoparticles Reach Sterically Obscured Endothelial Targets Inaccessible to Rigid Nanoparticles

Jacob W. Myerson; Bruce Braender; Olivia Mcpherson; Patrick M. Glassman; Raisa Yu. Kiseleva; Vladimir V. Shuvaev; Oscar A. Marcos-Contreras; Martha E. Grady; Hyun-Su Lee; Colin F. Greineder; Radu V. Stan; Russell J. Composto; David M. Eckmann; Vladimir R. Muzykantov

Molecular targeting of nanoparticle drug carriers promises maximized therapeutic impact to sites of disease or injury with minimized systemic effects. Precise targeting demands addressing to subcellular features. Caveolae, invaginations in cell membranes implicated in transcytosis and inflammatory signaling, are appealing subcellular targets. Caveolar geometry has been reported to impose a ≈50 nm size cutoff on nanocarrier access to plasmalemma vesicle associated protein (PLVAP), a marker found in caveolae in the lungs. The use of deformable nanocarriers to overcome that size cutoff is explored in this study. Lysozyme-dextran nanogels (NGs) are synthesized with ≈150 or ≈300 nm mean diameter. Atomic force microscopy indicates the NGs deform on complementary surfaces. Quartz crystal microbalance data indicate that NGs form softer monolayers (≈60 kPa) than polystyrene particles (≈8 MPa). NGs deform during flow through microfluidic channels, and modeling of NG extrusion through porous filters yields sieving diameters less than 25 nm for NGs with 150 and 300 nm hydrodynamic diameters. NGs of 150 and 300 nm diameter target PLVAP in mouse lungs while counterpart rigid polystyrene particles do not. The data in this study indicate a role for mechanical deformability in targeting large high-payload drug-delivery vehicles to sterically obscured targets like PLVAP.


Drug Delivery and Translational Research | 2018

Targeting therapeutics to endothelium: are we there yet?

Raisa Yu. Kiseleva; Patrick M. Glassman; Colin F. Greineder; Elizabeth D. Hood; Vladimir V. Shuvaev; Vladimir R. Muzykantov

Vascular endothelial cells represent an important therapeutic target in many pathologies, including inflammation, oxidative stress, and thrombosis; however, delivery of drugs to this site is often limited by the lack of specific affinity of therapeutics for these cells. Selective delivery of both small molecule drugs and therapeutic proteins to the endothelium has been achieved through the use of targeting ligands, such as monoclonal antibodies, directed against endothelial cell surface markers, particularly cell adhesion molecules (CAMs). Careful selection of target molecules and targeting agents allows for precise delivery to sites of inflammation, thereby maximizing therapeutic drug concentrations at the site of injury. A good understanding of the physiological and pathological determinants of drug and drug carrier pharmacokinetics and biodistribution may allow for a priori identification of optimal properties of drug carrier and targeting agent. Targeted delivery of therapeutics such as antioxidants and antithrombotic agents to the injured endothelium has shown efficacy in preclinical models, suggesting the potential for translation into clinical practice. As with all therapeutics, demonstration of both efficacy and safety are required for successful clinical implementation, which must be considered not only for the individual components (drug, targeting agent, etc.) but also for the sum of the parts (e.g., the drug delivery system), as unexpected toxicities may arise with complex delivery systems. While the use of endothelial targeting has not been translated into the clinic to date, the preclinical results summarized here suggest that there is hope for successful implementation of these agents in the years to come.


PLOS ONE | 2017

Mechanism of Collaborative Enhancement of Binding of Paired Antibodies to Distinct Epitopes of Platelet Endothelial Cell Adhesion Molecule-1

Raisa Yu. Kiseleva; Colin F. Greineder; Carlos H. Villa; Elizabeth D. Hood; Vladimir V. Shuvaev; Jing Sun; Ann-Marie Chacko; Valsamma Abraham; Horace M. DeLisser; Vladimir R. Muzykantov

Monoclonal antibodies (mAbs) directed to extracellular epitopes of human and mouse Platelet Endothelial Cell Adhesion Molecule-1 (CD31 or PECAM-1) stimulate binding of other mAbs to distinct adjacent PECAM-1 epitopes. This effect, dubbed Collaborative Enhancement of Paired Affinity Ligands, or CEPAL, has been shown to enhance delivery of mAb-targeted drugs and nanoparticles to the vascular endothelium. Here we report new insights into the mechanism underlying this effect, which demonstrates equivalent amplitude in the following models: i) cells expressing a full length PECAM-1 and mutant form of PECAM-1 unable to form homodimers; ii) isolated fractions of cellular membranes; and, iii) immobilized recombinant PECAM-1. These results indicate that CEPAL is mediated not by interference in cellular functions or homophilic PECAM-1 interactions, but rather by conformational changes within the cell adhesion molecule induced by ligand binding. This mechanism, mediated by exposure of partially occult epitopes, is likely to occur in molecules other than PECAM-1 and may represent a generalizable phenomenon with valuable practical applications.


Bioconjugate Chemistry | 2017

Site-Specific Modification of Single-Chain Antibody Fragments for Bioconjugation and Vascular Immunotargeting

Colin F. Greineder; Carlos H. Villa; Landis Walsh; Raisa Yu. Kiseleva; Elizabeth D. Hood; Makan Khoshnejad; Robert Warden-Rothman; Andrew Tsourkas; Vladimir R. Muzykantov

The conjugation of antibodies to drugs and drug carriers improves delivery to target tissues. Widespread implementation and effective translation of this pharmacologic strategy awaits the development of affinity ligands capable of a defined degree of modification and highly efficient bioconjugation without loss of affinity. To date, such ligands are lacking for the targeting of therapeutics to vascular endothelial cells. To enable site-specific, click-chemistry conjugation to therapeutic cargo, we used the bacterial transpeptidase, sortase A, to attach short azidolysine containing peptides to three endothelial-specific single chain antibody fragments (scFv). While direct fusion of a recognition motif (sortag) to the scFv C-terminus generally resulted in low levels of sortase-mediated modification, improved reaction efficiency was observed for one protein, in which two amino acids had been introduced during cloning. This prompted insertion of a short, semi-rigid linker between scFv and sortag. The linker significantly enhanced modification of all three proteins, to the extent that unmodified scFv could no longer be detected. As proof of principle, purified, azide-modified scFv was conjugated to the antioxidant enzyme, catalase, resulting in robust endothelial targeting of functional cargo in vitro and in vivo.


Nature Communications | 2018

Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude

Jacob S. Brenner; Daniel C. Pan; Jacob W. Myerson; Oscar A. Marcos-Contreras; Carlos H. Villa; Priyal Patel; Hugh Hekierski; Shampa Chatterjee; Jian-Qin Tao; Hamideh Parhiz; Kartik Bhamidipati; Thomas Uhler; Elizabeth D. Hood; Raisa Yu. Kiseleva; Vladimir S. Shuvaev; Tea Shuvaeva; Makan Khoshnejad; Ian Johnston; Jason V. Gregory; Joerg Lahann; Tao Wang; Edward Cantu; William M. Armstead; Samir Mitragotri; Vladimir R. Muzykantov

Drug delivery by nanocarriers (NCs) has long been stymied by dominant liver uptake and limited target organ deposition, even when NCs are targeted using affinity moieties. Here we report a universal solution: red blood cell (RBC)-hitchhiking (RH), in which NCs adsorbed onto the RBCs transfer from RBCs to the first organ downstream of the intravascular injection. RH improves delivery for a wide range of NCs and even viral vectors. For example, RH injected intravenously increases liposome uptake in the first downstream organ, lungs, by ~40-fold compared with free NCs. Intra-carotid artery injection of RH NCs delivers >10% of the injected NC dose to the brain, ~10× higher than that achieved with affinity moieties. Further, RH works in mice, pigs, and ex vivo human lungs without causing RBC or end-organ toxicities. Thus, RH is a clinically translatable platform technology poised to augment drug delivery in acute lung disease, stroke, and several other diseases.Unwanted uptake in the liver and limited accumulation in target organs is a major obstacle to targeted drug delivery. Here, the authors report on the hitchhiking of nanocarriers on red blood cells and the targeted upstream delivery to different target organs in mice, pigs and ex vivo human lungs.


Scientific Reports | 2018

Vascular endothelial effects of collaborative binding to platelet/endothelial cell adhesion molecule-1 (PECAM-1)

Raisa Yu. Kiseleva; Colin F. Greineder; Carlos H. Villa; Oscar A. Marcos-Contreras; Elizabeth D. Hood; Vladimir V. Shuvaev; Horace M. DeLisser; Vladimir R. Muzykantov

Targeting drugs to endothelial cells has shown the ability to improve outcomes in animal models of inflammatory, ischemic and thrombotic diseases. Previous studies have revealed that certain pairs of ligands (antibodies and antibody fragments) specific for adjacent, but distinct, epitopes on PECAM-1 enhance each other’s binding, a phenomenon dubbed Collaborative Enhancement of Paired Affinity Ligands, or CEPAL. This discovery has been leveraged to enable simultaneous delivery of multiple therapeutics to the vascular endothelium. Given the known role of PECAM-1 in promoting endothelial quiescence and cell junction integrity, we sought here to determine if CEPAL might induce unintended vascular effects. Using a combination of in vitro and in vivo techniques and employing human and mouse endothelial cells under physiologic and pathologic conditions, we found only modest or non-significant effects in response to antibodies to PECAM-1, whether given solo or in pairs. In contrast, these methods detected significant elevation of endothelial permeability, pro-inflammatory vascular activation, and systemic cytokine release following antibody binding to the related endothelial junction protein, VE-Cadherin. These studies support the notion that PECAM-1-targeted CEPAL provides relatively well-tolerated endothelial drug delivery. Additionally, the analysis herein creates a template to evaluate potential toxicities of vascular-targeted nanoparticles and protein therapeutics.


Pulmonary circulation | 2018

The new frontiers of the targeted interventions in the pulmonary vasculature: precision and safety (2017 Grover Conference Series):

Jacob S. Brenner; Raisa Yu. Kiseleva; Patrick M. Glassman; Hamideh Parhiz; Colin F. Greineder; Elizabeth D. Hood; Vladimir V. Shuvaev; Vladimir R. Muzykantov

The pulmonary vasculature plays an important role in many lung pathologies, such as pulmonary arterial hypertension, primary graft dysfunction of lung transplant, and acute respiratory distress syndrome. Therapy for these diseases is quite limited, largely due to dose-limiting side effects of numerous drugs that have been trialed or approved. High doses of drugs targeting the pulmonary vasculature are needed due to the lack of specific affinity of therapeutic compounds to the vasculature. To overcome this problem, the field of targeted drug delivery aims to target drugs to the pulmonary endothelial cells, especially those in pathological regions. The field uses a variety of drug delivery systems (DDSs), ranging from nano-scale drug carriers, such as liposomes, to methods of conjugating drugs to affinity moieites, such as antibodies. These DDSs can deliver small molecule drugs, protein therapeutics, and imaging agents. Here we review targeted drug delivery to the pulmonary endothelium for the treatment of pulmonary diseases. Cautionary notes are made of the risk–benefit ratio and safety—parameters one should keep in mind when developing a translational therapeutic.


Journal of Controlled Release | 2018

PECAM-1 directed re-targeting of exogenous mRNA providing two orders of magnitude enhancement of vascular delivery and expression in lungs independent of apolipoprotein E-mediated uptake

Hamideh Parhiz; Vladimir V. Shuvaev; Norbert Pardi; Makan Khoshnejad; Raisa Yu. Kiseleva; Jacob S. Brenner; Thomas Uhler; Steven Tuyishime; Barbara L. Mui; Ying K. Tam; Thomas D. Madden; Michael J. Hope; Drew Weissman; Vladimir R. Muzykantov

ABSTRACT Systemic administration of lipid nanoparticle (LNP)‐encapsulated messenger RNA (mRNA) leads predominantly to hepatic uptake and expression. Here, we conjugated nucleoside‐modified mRNA‐LNPs with antibodies (Abs) specific to vascular cell adhesion molecule, PECAM‐1. Systemic (intravenous) administration of Ab/LNP‐mRNAs resulted in profound inhibition of hepatic uptake concomitantly with ˜200‐fold and 25‐fold elevation of mRNA delivery and protein expression in the lungs compared to non‐targeted counterparts. Unlike hepatic delivery of LNP‐mRNA, Ab/LNP‐mRNA is independent of apolipoprotein E. Vascular re‐targeting of mRNA represents a promising, powerful, and unique approach for novel experimental and clinical interventions in organs of interest other than liver. HIGHLIGHTSRobust pulmonary targeting of mRNA was achieved by endothelial targeted anti‐PECAM/LNP‐mRNA nanoparticles.Delivery of targeted Anti‐PECAM/LNP‐mRNA nanoparticles is independent of apolipoprotein E pathway.Around 200‐fold elevation in pulmonary mRNA expression was reached upon IV administration of targeted nanoparticles.Rapid, transient, and specific protein expression from reporter mRNA was observed, with limited off‐target biodistribution.


Biomaterials | 2018

Spatially controlled assembly of affinity ligand and enzyme cargo enables targeting ferritin nanocarriers to caveolae

Vladimir V. Shuvaev; Makan Khoshnejad; Katherine W. Pulsipher; Raisa Yu. Kiseleva; Evguenia Arguiri; Jasmina C. Cheung-Lau; Kathleen M. LeFort; Melpo Christofidou-Solomidou; Radu V. Stan; Ivan J. Dmochowski; Vladimir R. Muzykantov

One of the goals of nanomedicine is targeted delivery of therapeutic enzymes to the sub-cellular compartments where their action is needed. Endothelial caveolae-derived endosomes represent an important yet challenging destination for targeting, in part due to smaller size of the entry aperture of caveolae (ca. 30-50 nm). Here, we designed modular, multi-molecular, ferritin-based nanocarriers with uniform size (20 nm diameter) for easy drug-loading and targeted delivery of enzymatic cargo to these specific vesicles. These nanocarriers targeted to caveolar Plasmalemmal Vesicle-Associated Protein (Plvap) deliver superoxide dismutase (SOD) into endosomes in endothelial cells, the specific site of influx of superoxide mediating by such pro-inflammatory signaling as some cytokines and lipopolysaccharide (LPS). Cell studies showed efficient internalization of Plvap-targeted SOD-loaded nanocarriers followed by dissociation from caveolin-containing vesicles and intracellular transport to endosomes. The nanocarriers had a profound protective anti-inflammatory effect in an animal model of LPS-induced inflammation, in agreement with the characteristics of their endothelial uptake and intracellular transport, indicating that these novel, targeted nanocarriers provide an advantageous platform for caveolae-dependent delivery of biotherapeutics.

Collaboration


Dive into the Raisa Yu. Kiseleva's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elizabeth D. Hood

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Carlos H. Villa

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jacob S. Brenner

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Makan Khoshnejad

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hamideh Parhiz

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jacob W. Myerson

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge