Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
PLoS One ; 16(12): e0261599, 2021.
Article in English | MEDLINE | ID: mdl-34905584

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0233263.].

2.
PLoS One ; 16(5): e0251601, 2021.
Article in English | MEDLINE | ID: mdl-34038436

ABSTRACT

Traumatic brain injury (TBI) causes both physical disruption of the blood brain barrier (BBB) and altered immune responses that can lead to significant secondary brain injury and chronic inflammation within the central nervous system (CNS). Cell therapies, including mesenchymal stromal cells (MSC), have been shown to restore BBB integrity and augment endogenous splenic regulatory T cells (Treg), a subset of CD4+ T cells that function to regulate immune responses and prevent autoimmunity. We have recently shown that infusion of human cord blood-derived Treg decreased neuroinflammation after TBI in vivo and in vitro. However, while both cells have demonstrated anti-inflammatory and regenerative potential, they likely utilize differing, although potentially overlapping, mechanisms. Furthermore, studies investigating these two cell types together, as a combination therapy, are lacking. In this study, we compared the ability of Treg+MSC combination therapy, as well as MSC and Treg monotherapies, to improve BBB permeability in vivo and suppress inflammation in vitro. While Treg+MSC combination did not significantly augment potency in vivo, our in vitro data demonstrates that combination therapy may augment therapeutic potency and immunosuppressive potential compared to Treg or MSC monotherapy.


Subject(s)
Blood-Brain Barrier/immunology , Brain Injuries, Traumatic , Immune Tolerance , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , T-Lymphocytes, Regulatory , Animals , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/therapy , Disease Models, Animal , Humans , Male , Rats , Rats, Sprague-Dawley , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation
3.
Stem Cells ; 39(3): 358-370, 2021 03.
Article in English | MEDLINE | ID: mdl-33368792

ABSTRACT

The inflammatory response after traumatic brain injury (TBI) can lead to significant secondary brain injury and chronic inflammation within the central nervous system. Cell therapies, including mesenchymal stromal cells (MSC), have led to improvements in animal models of TBI and are under investigation in human trials. One potential mechanism for the therapeutic potential of MSC is their ability to augment the endogenous response of immune suppressive regulatory T cells (Treg). We have recently shown that infusion of human cord blood Treg decreased chronic microgliosis after TBI and altered the systemic immune response in a rodent model. These cells likely use both overlapping and distinct mechanisms to modulate the immune system; therefore, combining Treg and MSC as a combination therapy may confer therapeutic benefit over either monotherapy. However, investigation of Treg + MSC combination therapy in TBI is lacking. In this study, we compared the ability MSC + Treg combination therapy, as well as MSC and Treg monotherapies, to inhibit the neuroinflammatory response to TBI in vivo and in vitro. Treg + MSC combination therapy demonstrated increased potency to reduce the neuro- and peripheral inflammatory response compared to monotherapy; furthermore, the timing of infusion proved to be a significant variable in the efficacy of both MSC monotherapy and Treg + MSC combination therapy in vivo and in vitro.


Subject(s)
Brain Injuries, Traumatic/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Brain Injuries, Traumatic/immunology , Combined Modality Therapy/methods , Disease Models, Animal , Immunity , Inflammation/therapy , Mesenchymal Stem Cell Transplantation/methods , Rats, Sprague-Dawley
4.
Sci Rep ; 10(1): 10729, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612177

ABSTRACT

The immune system plays critical roles in promoting tissue repair during recovery from neurotrauma but is also responsible for unchecked inflammation that causes neuronal cell death, systemic stress, and lethal immunodepression. Understanding the immune response to neurotrauma is an urgent priority, yet current models of traumatic brain injury (TBI) inadequately recapitulate the human immune response. Here, we report the first description of a humanized model of TBI and show that TBI places significant stress on the bone marrow. Hematopoietic cells of the marrow are regionally decimated, with evidence pointing to exacerbation of underlying graft-versus-host disease (GVHD) linked to presence of human T cells in the marrow. Despite complexities of the humanized mouse, marrow aplasia caused by TBI could be alleviated by cell therapy with human bone marrow mesenchymal stromal cells (MSCs). We conclude that MSCs could be used to ameliorate syndromes triggered by hypercytokinemia in settings of secondary inflammatory stimulus that upset marrow homeostasis such as TBI. More broadly, this study highlights the importance of understanding how underlying immune disorders including immunodepression, autoimmunity, and GVHD might be intensified by injury.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Graft vs Host Disease/etiology , Immune Tolerance/immunology , Mesenchymal Stem Cells/cytology , T-Lymphocytes/immunology , Animals , Female , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Male , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred NOD , Mice, SCID
5.
Stem Cells Transl Med ; 9(8): 903-916, 2020 08.
Article in English | MEDLINE | ID: mdl-32383348

ABSTRACT

Traumatic brain injury (TBI) causes a profound inflammatory response within the central nervous system and peripheral immune system, which contributes to secondary brain injury and further morbidity and mortality. Preclinical investigations have demonstrated that treatments that downregulate microglia activation and polarize them toward a reparative/anti-inflammatory phenotype have improved outcomes in preclinical models. However, no therapy to date has translated into proven benefits in human patients. Regulatory T cells (Treg) have been shown to downregulate pathologic immune responses of the innate and adaptive immune system across a variety of pathologies. Furthermore, cellular therapy has been shown to augment host Treg responses in preclinical models; yet, studies investigating the use of Treg as a therapeutic for TBI are lacking. In a rodent TBI model, we demonstrate that human umbilical cord blood Treg modulate the central and peripheral immune response after injury in vitro and in vivo.


Subject(s)
Brain Injuries, Traumatic/immunology , Cell- and Tissue-Based Therapy/methods , Immunity/immunology , Immunophenotyping/methods , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Humans , Rats , Rats, Sprague-Dawley
6.
PLoS One ; 15(5): e0233263, 2020.
Article in English | MEDLINE | ID: mdl-32453741

ABSTRACT

In the U.S., approximately 1.7 million people suffer traumatic brain injury each year, with many enduring long-term consequences and significant medical and rehabilitation costs. The primary injury causes physical damage to neurons, glia, fiber tracts and microvasculature, which is then followed by secondary injury, consisting of pathophysiological mechanisms including an immune response, inflammation, edema, excitotoxicity, oxidative damage, and cell death. Most attempts at intervention focus on protection, repair or regeneration, with regenerative medicine becoming an intensively studied area over the past decade. The use of stem cells has been studied in many disease and injury models, using stem cells from a variety of sources and applications. In this study, human adipose-derived mesenchymal stromal cells (MSCs) were administered at early (3 days) and delayed (14 days) time points after controlled cortical impact (CCI) injury in rats. Animals were routinely assessed for neurological and vestibulomotor deficits, and at 32 days post-injury, brain tissue was processed by flow cytometry and immunohistochemistry to analyze neuroinflammation. Treatment with HB-adMSC at either 3d or 14d after injury resulted in significant improvements in neurocognitive outcome and a change in neuroinflammation one month after injury.


Subject(s)
Brain Injuries, Traumatic/therapy , Mesenchymal Stem Cell Transplantation , Adipose Tissue/cytology , Animals , Brain/pathology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/psychology , Disease Models, Animal , Humans , Inflammation/pathology , Male , Maze Learning , Mesenchymal Stem Cells/cytology , Motor Skills , Neurogenesis , Rats, Sprague-Dawley , Time Factors
7.
Stem Cells Transl Med ; 9(4): 491-498, 2020 04.
Article in English | MEDLINE | ID: mdl-31903737

ABSTRACT

Clinical trials in trauma populations are exploring the use of clinical cellular therapeutics (CCTs) like human mesenchymal stromal cells (MSC) and mononuclear cells (MNC). Recent studies demonstrate a procoagulant effect of these CCTs related to their expression of tissue factor (TF). We sought to examine this relationship in blood from severely injured trauma patients and identify methods to reverse this procoagulant effect. Human MSCs from bone marrow, adipose, and amniotic tissues and freshly isolated bone marrow MNC samples were tested. TF expression and phenotype were quantified using flow cytometry. CCTs were mixed individually with trauma patients' whole blood, assayed with thromboelastography (TEG), and compared with healthy subjects mixed with the same cell sources. Heparin was added to samples at increasing concentrations until TEG parameters normalized. Clotting time or R time in TEG decreased relative to the TF expression of the CCT treatment in a logarithmic fashion for trauma patients and healthy subjects. Nonlinear regression curves were significantly different with healthy subjects demonstrating greater relative decreases in TEG clotting time. In vitro coadministration of heparin normalized the procoagulant effect and required dose escalation based on TF expression. TF expression in human MSC and MNC has a procoagulant effect in blood from trauma patients and healthy subjects. The procoagulant effect is lower in trauma patients possibly because their clotting time is already accelerated. The procoagulant effect due to MSC/MNC TF expression could be useful in the bleeding trauma patient; however, it may emerge as a safety release criterion due to thrombotic risk. The TF procoagulant effect is reversible with heparin.


Subject(s)
Blood Coagulation , Wounds and Injuries/blood , Adult , Biomarkers/metabolism , Blood Coagulation/drug effects , Case-Control Studies , Female , Heparin/pharmacology , Humans , Male , Thrombelastography
8.
Front Immunol ; 10: 1645, 2019.
Article in English | MEDLINE | ID: mdl-31417542

ABSTRACT

For several decades, multipotent mesenchymal stromal cells (MSCs) have been extensively studied for their therapeutic potential across a wide range of diseases. In the preclinical setting, MSCs demonstrate consistent ability to promote tissue healing, down-regulate excessive inflammation and improve outcomes in animal models. Several proposed mechanisms of action have been posited and demonstrated across an array of in vitro models. However, translation into clinical practice has proven considerably more difficult. A number of prominent well-funded late-phase clinical trials have failed, thus calling out for new efforts to optimize product delivery in the clinical setting. In this review, we discuss novel topics critical to the successful translation of MSCs from pre-clinical to clinical applications. In particular, we focus on the major routes of cell delivery, aspects related to hemocompatibility, and potential safety concerns associated with MSC therapy in the different settings.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Animals , Disease Models, Animal , Humans
9.
Stem Cells Transl Med ; 7(10): 731-739, 2018 10.
Article in English | MEDLINE | ID: mdl-30070065

ABSTRACT

Clinical cellular therapeutics (CCTs) have shown preliminary efficacy in reducing inflammation after trauma, preserving cardiac function after myocardial infarction, and improving functional recovery after stroke. However, most clinically available cell lines express tissue factor (TF) which stimulates coagulation. We sought to define the degree of procoagulant activity of CCTs as related to TF expression. CCT samples from bone marrow, adipose, amniotic fluid, umbilical cord, multi-potent adult progenitor cell donors, and bone marrow mononuclear cells were tested. TF expression and phenotype were quantified using flow cytometry. Procoagulant activity of the CCTs was measured in vitro with thromboelastography and calibrated thrombogram. Fluorescence-activated cell sorting (FACS) separated samples into high- and low-TF expressing populations to isolate the contribution of TF to coagulation. A TF neutralizing antibody was incubated with samples to demonstrate loss of procoagulant function. All CCTs tested expressed procoagulant activity that correlated with expression of tissue factor. Time to clot and thrombin formation decreased with increasing TF expression. High-TF expressing cells decreased clotting time more than low-TF expressing cells when isolated from a single donor using FACS. A TF neutralizing antibody restored clotting time to control values in some, but not all, CCT samples. CCTs demonstrate wide variability in procoagulant activity related to TF expression. Time to clot and thrombin formation decreases as TF load increases and this procoagulant effect is neutralized by a TF blocking antibody. Clinical trials using CCTs are in progress and TF expression may emerge as a safety release criterion. Stem Cells Translational Medicine 2018;7:731-739.


Subject(s)
Blood Coagulation , Cell- and Tissue-Based Therapy/methods , Thromboplastin/metabolism , Adipose Tissue/cytology , Amniotic Fluid/cytology , Bone Marrow Cells/cytology , Fetal Blood/cytology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Thrombelastography , Thrombin/metabolism , Thromboplastin/genetics
10.
Sci Rep ; 8(1): 480, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323194

ABSTRACT

No current clinical intervention can alter the course of acute spinal cord injury (SCI), or appreciably improve neurological outcome. Mesenchymal stromal cells (MSCs) have been shown to modulate the injury sequelae of SCI largely via paracrine effects, although the mechanisms remain incompletely understood. One potential modality is through secretion of extracellular vesicles (EVs). In this study, we investigate whether systemic administration of EVs isolated from human MSCs (MSCEv) has the potential to be efficacious as an alternative to cell-based therapy for SCI. Additionally, we investigate whether EVs isolated from human MSCs stimulated with pro-inflammatory cytokines have enhanced anti-inflammatory effects when administered after SCI. Immunohistochemistry supported the quantitative analysis, demonstrating a diminished inflammatory response with apparent astrocyte and microglia disorganization in cord tissue up to 10 mm caudal to the injury site. Locomotor recovery scores showed significant improvement among animals treated with MSCEv. Significant increases in mechanical sensitivity threshold were observed in animals treated with EVs from either naïve MSC (MSCEvwt) or stimulated MSC (MSCEv+), with a statistically significant increase in threshold for MSCEv+-treated animals when compared to those that received MSCEvwt. In conclusion, these data show that treatment of acute SCI with extracellular vesicles derived from human MSCs attenuates neuroinflammation and improves functional recovery.


Subject(s)
Extracellular Vesicles/transplantation , Microglia/metabolism , Spinal Cord Injuries/therapy , Animals , Astrocytes/cytology , Astrocytes/metabolism , Disease Models, Animal , Extracellular Vesicles/metabolism , Humans , Inflammation , Locomotion/physiology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microglia/cytology , Neutrophils/cytology , Neutrophils/metabolism , Rats , Rats, Sprague-Dawley , Receptors, IgG/metabolism , Recovery of Function , Spleen/cytology , Spleen/immunology , Treatment Outcome
11.
Stem Cells ; 36(1): 79-90, 2018 01.
Article in English | MEDLINE | ID: mdl-29076623

ABSTRACT

Extracellular vesicles (EVs) secreted by mesenchymal stromal cells (MSCs) have been proposed to be a key mechanistic link in the therapeutic efficacy of cells in response to cellular injuries through paracrine effects. We hypothesize that inflammatory stimulation of MSCs results in the release of EVs that have greater anti-inflammatory effects. The present study evaluates the immunomodulatory abilities of EVs derived from inflammation-stimulated and naive MSCs (MSCEv+ and MSCEv, respectively) isolated using a current Good Manufacturing Practice-compliant tangential flow filtration system. Detailed characterization of both EVs revealed differences in protein composition, cytokine profiles, and RNA content, despite similarities in size and expression of common surface markers. MSCEv+ further attenuated release of pro-inflammatory cytokines in vitro when compared to MSCEv, with a distinctly different pattern of EV-uptake by activated primary leukocyte subpopulations. The efficacy of EVs was partially attributed to COX2/PGE2 expression. The present study demonstrates that inflammatory stimulation of MSCs renders release of EVs that have enhanced anti-inflammatory properties partially due to COX2/PGE2 pathway alteration. Stem Cells 2018;36:79-90.


Subject(s)
Extracellular Vesicles/metabolism , Inflammation/metabolism , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Transmission/methods , Humans
13.
Nanoscale ; 9(38): 14581-14591, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28932838

ABSTRACT

Inflammatory bowel disease (IBD), which includes Crohn's disease and ulcerative colitis, is a chronic inflammatory condition of the gastrointestinal (GI) tract. Currently, it is treated with immunosuppressant or biologics that often induce severe adverse effects. Thus, there is an urgent clinical need for more specific treatments. To provide a valid therapeutic tool for IBD therapy, in this work we developed biomimetic nanovesicles by manipulating leukocyte membranes to exploit mechanisms of T-cell recruitment during inflammation. A subset of T-lymphocytes participates in homing to inflamed tissue in the gastrointestinal tract by overexpressing the α4ß7 integrin, which is responsible for binding to its receptor on the endothelial membrane, the mucosal addressin cell adhesion molecule 1. Based on this principle, we engineered biomimetic vesicles, referred to as specialized leukosomes (SLKs), which are leukocyte-like carriers 'doped' with the α4ß7 integrin over-induced in purified immune cells. We tested SLKs in an in vivo murine model of IBD induced by treatment with dextran sulfate sodium. Notably, treatment of IBD mice with SLKs allowed us to observe a reduction of inflammation (favorable modulation of both pro- and anti-inflammatory genes, as well as reduction of immune cells infiltration into the colon tissue), and a consequent enhanced intestinal repair (low epithelial damage). In this study, we demonstrate that biological-derived nanoparticles can be used not only as naturally targeted drug delivery systems, but also as nano-therapeutics endowed with intrinsic anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biomimetic Materials , Inflammatory Bowel Diseases/drug therapy , Nanoparticles , Animals , Dextran Sulfate , Inflammatory Bowel Diseases/chemically induced , Integrins , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
14.
ACS Nano ; 11(3): 3262-3273, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28264157

ABSTRACT

Understanding interactions occurring at the interface between nanoparticles and biological components is an urgent challenge in nanomedicine due to their effect on the biological fate of nanoparticles. After the systemic injection of nanoparticles, a protein corona constructed by blood components surrounds the carrier's surface and modulates its pharmacokinetics and biodistribution. Biomimicry-based approaches in nanotechnology attempt to imitate what happens in nature in order to transfer specific natural functionalities to synthetic nanoparticles. Several biomimetic formulations have been developed, showing superior in vivo features as a result of their cell-like identity. We have recently designed biomimetic liposomes, called leukosomes, which recapitulate the ability of leukocytes to target inflamed endothelium and escape clearance by the immune system. To gain insight into the properties of leukosomes, we decided to investigate their protein corona in vivo. So far, most information about the protein corona has been obtained using in vitro experiments, which have been shown to minimally reproduce in vivo phenomena. Here we directly show a time-dependent quantitative and qualitative analysis of the protein corona adsorbed in vivo on leukosomes and control liposomes. We observed that leukosomes absorb fewer proteins than liposomes, and we identified a group of proteins specifically adsorbed on leukosomes. Moreover, we hypothesize that the presence of macrophage receptors on leukosomes' surface neutralizes their protein corona-meditated uptake by immune cells. This work unveils the protein corona of a biomimetic carrier and is one of the few studies on the corona performed in vivo.


Subject(s)
Nanoparticles/chemistry , Protein Corona/chemistry , Adsorption , Animals , Cell Line , Cryoelectron Microscopy , Electrophoresis, Capillary , Liposomes/blood , Liposomes/chemistry , Mass Spectrometry , Mice , Microscopy, Confocal
15.
Methodist Debakey Cardiovasc J ; 12(3): 141-145, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27826367

ABSTRACT

Atherosclerosis is a complex process responsible for a major burden of cardiovascular morbidity and mortality. Macrophages and smooth muscle cells (SMCs) are abundant within atherosclerotic plaques. This review discusses the role of macrophages and SMCs in plaque progression and provides an overview of nanoparticle-based approaches and other current methods for local targeting of atherosclerotic plaques.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Macrophages/drug effects , Muscle, Smooth, Vascular/drug effects , Plaque, Atherosclerotic/prevention & control , Animals , Cell Proliferation , Disease Progression , Humans , Macrophages/pathology , Muscle, Smooth, Vascular/pathology , Plaque, Atherosclerotic/pathology
16.
Methodist Debakey Cardiovasc J ; 12(3): 169-174, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27826372

ABSTRACT

Despite an improved understanding of its pathophysiology and a wide range of new treatments, cardiovascular disease (CVD) remains a serious public health issue and the number one cause of mortality in the United States. Conditions that promote chronic systemic inflammation, such as obesity, cancer, and autoimmune and infectious diseases, are now known to play an important role in promoting CVD by inducing the expression of endothelial adhesion molecules and chemokines; these in turn promote leukocyte adherence and infiltration, which initiates and spurs the progression of CVD. In response to this new understanding, researchers are evaluating the potential cardiovascular benefits of new-generation therapies based on endogenous molecules with anti-inflammatory properties. Similarly, targeted approaches that leverage the phenotypic differences between non-inflamed and inflamed endothelia have the potential to selectively deliver therapeutics and decrease the morbidity and mortality of CVD patients. In this review, we discuss the role of inflammation in CVD and explore the therapeutic potential of targeting inflamed vasculature through conventional and biomimetic approaches.


Subject(s)
Biological Factors/administration & dosage , Drug Delivery Systems , Nanomedicine/methods , Nanoparticles , Vasculitis/drug therapy , Humans
17.
Int J Nanomedicine ; 11: 3049-63, 2016.
Article in English | MEDLINE | ID: mdl-27445473

ABSTRACT

A thorough understanding of interactions occurring at the interface between nanocarriers and biological systems is crucial to predict and interpret their biodistribution, targeting, and efficacy, and thus design more effective drug delivery systems. Upon intravenous injection, nanoparticles are coated by a protein corona (PC). This confers a new biological identity on the particles that largely determines their biological fate. Liposomes have great pharmaceutical versatility, so, as proof of concept, their PC has recently been implicated in the mechanism and efficiency of their internalization into the cell. In an attempt to better understand the interactions between nanocarriers and biological systems, we analyzed the plasma proteins adsorbed on the surface of multicomponent liposomes. Specifically, we analyzed the physical properties and ultrastructure of liposome/PC complexes and the aggregation process that occurs when liposomes are dispersed in plasma. The results of combined confocal microscopy and flow cytometry experiments demonstrated that the PC favors liposome internalization by both macrophages and tumor cells. This work provides insights into the effects of the PC on liposomes' physical properties and, consequently, liposome-liposome and liposome-cell interactions.


Subject(s)
Cell Communication , Liposomes/chemistry , Protein Corona/chemistry , Adsorption , Animals , Blood Proteins/metabolism , Cell Line, Tumor , Cryoelectron Microscopy , Dynamic Light Scattering , Endocytosis , Flow Cytometry , Humans , Mice, Inbred BALB C , Microscopy, Atomic Force , Nanoparticles/chemistry , Proteomics
18.
Nanomedicine (Lond) ; 11(1): 81-100, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26653875

ABSTRACT

In a perfect sequence of events, nanoparticles (NPs) are injected into the bloodstream where they circulate until they reach the target tissue. The ligand on the NP surface recognizes its specific receptor expressed on the target tissue and the drug is released in a controlled manner. However, once injected in a physiological environment, NPs interact with biological components and are surrounded by a protein corona (PC). This can trigger an immune response and affect NP toxicity and targeting capabilities. In this review, we provide a survey of recent findings on the NP-PC interactions and discuss how the PC can be used to modulate both cytotoxicity and the immune response as well as to improve the efficacy of targeted delivery of nanocarriers.


Subject(s)
Nanoparticles/toxicity , Animals , Complement Activation , Drug Delivery Systems , Humans , Nanoparticles/administration & dosage
19.
Nano Lett ; 13(7): 3248-55, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23786263

ABSTRACT

The ultimate goal in cancer therapy is achieving selective targeting of cancer cells. We report a novel delivery platform, based on nanoghosts (NGs) produced from the membranes of mesenchymal stem cells (MSCs). Encompassing MSC surface molecules, the MSC-NGs retained MSC-specific in vitro and in vivo tumor targeting capabilities and were cleared from blood-filtering organs. MSC-NGs were found to be biocompatible. Systemic administration of drug loaded MSC-NGs demonstrated 80% inhibition of human prostate cancer.


Subject(s)
Cell Membrane/chemistry , Mesenchymal Stem Cells/chemistry , Nanocapsules/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Receptors, TNF-Related Apoptosis-Inducing Ligand/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Nanocapsules/administration & dosage , Nanocapsules/ultrastructure , Organ Specificity , Particle Size , Tissue Distribution , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...