Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
ACS Biomater Sci Eng ; 8(8): 3526-3541, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35838679

ABSTRACT

Cyclic strain generated at the cell-material interface is critical for the engraftment of biomaterials. Mechanosensitive immune cells, macrophages regulate the host-material interaction immediately after implantation by priming the environment and remodeling ongoing regenerative processes. This study investigated the ability of mechanically active scaffolds to modulate macrophage function in vitro and in vivo. Remotely actuated magnetic scaffolds enhance the phenotype of murine classically activated (M1) macrophages, as shown by the increased expression of the M1 cell-surface marker CD86 and increased secretion of multiple M1 cytokines. When scaffolds were implanted subcutaneously into mice and treated with magnetic stimulation for 3 days beginning at either day 0 or day 5 post-implantation, the cellular infiltrate was enriched for host macrophages. Macrophage expression of the M1 marker CD86 was increased, with downstream effects on vascularization and the foreign body response. Such effects were not observed when the magnetic treatment was applied at later time points after implantation (days 12-15). These results advance our understanding of how remotely controlled mechanical cues, namely, cyclic strain, impact macrophage function and demonstrate the feasibility of using mechanically active nanomaterials to modulate the host response in vivo.


Subject(s)
Macrophages , Tissue Scaffolds , Animals , Biocompatible Materials , Macrophages/metabolism , Mice , Phenotype
2.
Cardiovasc Res ; 118(7): 1771-1784, 2022 06 22.
Article in English | MEDLINE | ID: mdl-34132787

ABSTRACT

AIMS: Hydrogen sulfide (H2S) is a potent signalling molecule that activates diverse cardioprotective pathways by post-translational modification (persulfidation) of cysteine residues in upstream protein targets. Heart failure patients with reduced ejection fraction (HFrEF) exhibit low levels of H2S. Sulfide:quinone oxidoreductase (SQOR) catalyses the first irreversible step in the metabolism of H2S and plays a key role in regulating H2S-mediated signalling. Here, the aim of this study was to discover a first-in-class inhibitor of human SQOR and evaluate its cardioprotective effect in an animal model of HFrEF. METHODS AND RESULTS: We identified a potent inhibitor of human SQOR (STI1, IC50 = 29 nM) by high-throughput screening of a small-molecule library, followed by focused medicinal chemistry optimization and structure-based design. STI1 is a competitive inhibitor that binds with high selectivity to the coenzyme Q-binding pocket in SQOR. STI1 exhibited very low cytotoxicity and attenuated the hypertrophic response of neonatal rat ventricular cardiomyocytes and H9c2 cells induced by neurohormonal stressors. A mouse HFrEF model was produced by transverse aortic constriction (TAC). Treatment of TAC mice with STI1 mitigated the development of cardiomegaly, pulmonary congestion, dilatation of the left ventricle, and cardiac fibrosis and decreased the pressure gradient across the aortic constriction. Moreover, STI1 dramatically improved survival, preserved cardiac function, and prevented the progression to HFrEF by impeding the transition from compensated to decompensated left ventricle hypertrophy. CONCLUSION: We demonstrate that the coenzyme Q-binding pocket in human SQOR is a druggable target and establish proof of concept for the potential of SQOR inhibitors to provide a novel therapeutic approach for the treatment of HFrEF.


Subject(s)
Heart Failure , Animals , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/prevention & control , Humans , Mice , Rats , Stroke Volume , Sulfides/pharmacology , Ubiquinone/therapeutic use , Ventricular Remodeling
3.
Anticancer Res ; 40(9): 4857-4867, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32878773

ABSTRACT

BACKGROUND/AIM: Anticancer peptide PNC-27 binds to HDM-2 protein on cancer cell membranes inducing the formation of cytotoxic transmembrane pores. Herein, we investigated HDM-2 membrane expression and the effect of PNC-27 treatment on human non-stem cell acute myelogenous leukemia cell lines: U937, acute monocytic leukemia; OCI-AML3, acute myelomonocytic leukemia and HL60, acute promyelocytic leukemia. MATERIALS AND METHODS: We measured cell surface membrane expression of HDM-2 using flow cytometry. Cell viability was assessed using MTT assay while direct cytotoxicity was measured by lactate dehydrogenase (LDH) release and induction of apoptotic markers annexin V and caspase-3. RESULTS: HDM-2 is expressed at high levels in membranes of U937, OCI-AML3 and HL-60 cells. PNC-27 can bind to membrane HDM-2 to induce cell necrosis and LDH release within 4 h. CONCLUSION: Targeting membrane HDM-2 can be a potential strategy to treat leukemia. PNC-27 targeting membrane HDM-2 demonstrated significant anti-leukemia activity in a variety of leukemic cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/pharmacology , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , L-Lactate Dehydrogenase/metabolism , Leukemia, Myeloid/metabolism , Necrosis , Tumor Suppressor Protein p53/metabolism
4.
Ann Surg Oncol ; 27(1): 76-84, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31187366

ABSTRACT

BACKGROUND: Treatment failure in pseudomyxoma peritonei (PMP) is partly attributed to the ineffective delivery of therapeutics through dense mucinous tumor barriers. We modified the surface of Poly (lactic-co-glycolic acid)-b-polyethylene glycol (PLGA-PEG-NPs) with a low-density, second PEG layer (PLGA-TPEG-NPs-20) to reduce their binding affinity to proteins and improve diffusion through mucin. METHODS: Nanoprecipitation was used to fabricate PLGA-PEG-NPs. To construct the second PEG layer of PLGA-TPEG-NPs-20, PEG-Thiol was conjugated to PLGA-PEG-NPs composed of 80% methoxy PLGA-PEG and 20% of PLGA-PEG-Maleimide. DiD-labeled nanoparticles (NPs) were added to the inner well of a trans-well system containing cultured LS174T or human PMP tissue. Diffusion of NPs was measured via fluorescence signal in the bottom well. In an ex vivo rat model, small intestine was treated with DiD-labeled NPs. In an in vivo murine LS174T subcutaneous tumor model, Nu/Nu nude mice received supratumoral injections (subcutaneous injection above the tumor) of DiD-labeled NPs. Thirty minutes after injection, mice were sacrificed, and tumors were collected. All tissue was cryosectioned, mounted with DAPI-containing media, and inspected via confocal microscopy. RESULTS: Diffusion profiles of NPs through PMP and cultured LS174T cells were generated. PLGA-TPEG-NPs-20 diffused faster with ~ 100% penetration versus PLGA-PEG-NPs with ~ 40% penetration after 8 h. Increased diffusion of PLGA-TPEG-NPs-20 was further observed in ex vivo rat small intestine as evidenced by elevated luminal NP fluorescence signal on the luminal surface. Subcutaneous LS174T tumors treated with PLGA-TPEG-NPs-20 demonstrated greater diffusion of NPs, showing homogenous fluorescence signal throughout the tumor. CONCLUSIONS: PLGA-TPEG-NPs-20 can be an effective mucin penetrating drug delivery system.


Subject(s)
Drug Delivery Systems , Intestine, Small/metabolism , Mucin-1/metabolism , Nanoparticles/administration & dosage , Peritoneal Neoplasms/metabolism , Polyesters/chemistry , Polyethylene Glycols/chemistry , Pseudomyxoma Peritonei/metabolism , Animals , Apoptosis , Cell Proliferation , Diffusion , Female , Humans , Intestine, Small/drug effects , Mice , Mice, Nude , Nanoparticles/chemistry , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/pathology , Pseudomyxoma Peritonei/drug therapy , Pseudomyxoma Peritonei/pathology , Rats , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Cereb Blood Flow Metab ; 40(10): 1953-1965, 2020 10.
Article in English | MEDLINE | ID: mdl-31601141

ABSTRACT

Most neurological diseases, including stroke, lead to some degree of blood-brain barrier (BBB) dysfunction. A significant portion of BBB injury is caused by inflammation, due to pro-inflammatory factors produced in the brain, and by leukocyte engagement of the brain endothelium. Recently, microRNAs (miRNAs) have appeared as major regulators of inflammation-induced changes to gene expression in the microvascular endothelial cells (BMVEC) that comprise the BBB. However, miRNAs' role during cerebral ischemia/reperfusion is still underexplored. Endothelial levels of miR-98 were significantly altered following ischemia/reperfusion insults, both in vivo and in vitro, transient middle cerebral artery occlusion (tMCAO), and oxygen-glucose deprivation (OGD), respectively. Overexpression of miR-98 reduced the mouse's infarct size after tMCAO. Further, miR-98 lessened infiltration of proinflammatory Ly6CHI leukocytes into the brain following stroke and diminished the prevalence of M1 (activated) microglia within the impacted area. miR-98 attenuated BBB permeability, as demonstrated by changes to fluorescently-labeled dextran penetration in vivo and improved transendothelial electrical resistance (TEER) in vitro. Treatment with miR-98 improved significantly the locomotor impairment. Our study provides identification and functional assessment of miRNAs in brain endothelium and lays the groundwork for improving therapeutic approaches for patients suffering from ischemic attacks.


Subject(s)
Blood-Brain Barrier , Endothelium, Vascular , MicroRNAs/therapeutic use , Reperfusion Injury/prevention & control , Stroke/prevention & control , Animals , Electric Impedance , Encephalitis/pathology , Glucose/deficiency , Infarction, Middle Cerebral Artery/pathology , Leukocytes/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Microglia/pathology , Movement Disorders/drug therapy , Movement Disorders/etiology , Reperfusion Injury/genetics , Stroke/complications , Stroke/genetics , Transfection
6.
Curr Drug Deliv ; 16(7): 637-644, 2019.
Article in English | MEDLINE | ID: mdl-31237208

ABSTRACT

BACKGROUND: Inflammation is a hallmark of epileptogenic brain tissue. Previously, we have shown that inflammation in epilepsy can be delineated using systemically-injected fluorescent and magnetite- laden nanoparticles. Suggested mechanisms included distribution of free nanoparticles across a compromised blood-brain barrier or their transfer by monocytes that infiltrate the epileptic brain. OBJECTIVE: In the current study, we evaluated monocytes as vehicles that deliver nanoparticles into the epileptic brain. We also assessed the effect of epilepsy on the systemic distribution of nanoparticleloaded monocytes. METHODS: The in vitro uptake of 300-nm nanoparticles labeled with magnetite and BODIPY (for optical imaging) was evaluated using rat monocytes and fluorescence detection. For in vivo studies we used the rat lithium-pilocarpine model of temporal lobe epilepsy. In vivo nanoparticle distribution was evaluated using immunohistochemistry. RESULTS: 89% of nanoparticle loading into rat monocytes was accomplished within 8 hours, enabling overnight nanoparticle loading ex vivo. The dose-normalized distribution of nanoparticle-loaded monocytes into the hippocampal CA1 and dentate gyrus of rats with spontaneous seizures was 176-fold and 380-fold higher compared to the free nanoparticles (p<0.05). Seizures were associated with greater nanoparticle accumulation within the liver and the spleen (p<0.05). CONCLUSION: Nanoparticle-loaded monocytes are attracted to epileptogenic brain tissue and may be used for labeling or targeting it, while significantly reducing the systemic dose of potentially toxic compounds. The effect of seizures on monocyte biodistribution should be further explored to better understand the systemic effects of epilepsy.


Subject(s)
Drug Delivery Systems , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Magnetite Nanoparticles/administration & dosage , Monocytes , Animals , Boron Compounds/administration & dosage , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Fluorescent Dyes/administration & dosage , Inflammation/metabolism , Kidney/metabolism , Lithium Chloride , Liver/metabolism , Male , Pilocarpine , Rats, Wistar , Spleen/metabolism
7.
Nano Lett ; 18(11): 7314-7322, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30380888

ABSTRACT

Nano- and microscale topographical cues have become recognized as major regulators of cell growth, migration, and phenotype. In tissue engineering, the complex and anisotropic architecture of culture platforms is aimed to imitate the high degree of spatial organization of the extracellular matrix and basement membrane components. Here, we developed a method of creating a novel, magnetically aligned, three-dimensional (3D) tissue culture matrix with three distinct classes of anisotropy-surface topography, microstructure, and physical properties. Alginate-stabilized magnetic nanoparticles (MNPs) were added to a cross-linked alginate solution, and an external magnetic field of about 2400 G was applied during freezing to form the aligned macroporous scaffold structure. The resultant scaffold exhibited anisotropic topographic features on the submicron scale, the directionality of the pore shape, and increased scaffold stiffness in the direction of magnetic alignment. These scaffold features were modulated by an alteration in the impregnated MNP size and concentration, as quantified by electron microscopy, advanced image processing analyses, and rheological methods. Mouse myoblasts (C2C12) cultured on the magnetically aligned scaffolds, demonstrated co-oriented morphology in the direction of the magnetic alignment. In summary, magnetic alignment introduces several degrees of anisotropy in the scaffold structure, providing diverse mechanical cues that can affect seeded cells and further tissue development. Multiscale anisotropy together with the capability of the MNP-containing alginate scaffolds to undergo reversible shape deformation in an oscillating magnetic field creates interesting opportunities for multifarious stimulation of cells and functional tissue development.

8.
ACS Nano ; 10(10): 9559-9569, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27622988

ABSTRACT

Although drug-eluting stents have dramatically reduced the recurrence of restenosis after vascular interventions, the nonselective antiproliferative drugs released from these devices significantly delay reendothelialization and vascular healing, increasing the risk of short- and long-term stent failure. Efficient repopulation of endothelial cells in the vessel wall following injury may limit complications, such as thrombosis, neoatherosclerosis, and restenosis, through reconstitution of a luminal barrier and cellular secretion of paracrine factors. We assessed the potential of magnetically mediated delivery of endothelial cells (ECs) to inhibit in-stent stenosis induced by mechanical injury in a rat carotid artery stent angioplasty model. ECs loaded with biodegradable superparamagnetic nanoparticles (MNPs) were administered at the distal end of the stented artery and localized to the stent using a brief exposure to a uniform magnetic field. After two months, magnetic localization of ECs demonstrated significant protection from stenosis at the distal part of the stent in the cell therapy group compared to both the proximal part of stent in the cell therapy group and the control (stented, nontreated) group: 1.7-fold (p < 0.001) less reduction in lumen diameter as measured by B-mode and color Doppler ultrasound, 2.3-fold (p < 0.001) less reduction in the ratios of peak systolic velocities as measured by pulsed wave Doppler ultrasound, and 2.1-fold (p < 0.001) attenuation of stenosis as determined through end point morphometric analysis. The study thus demonstrates that magnetically assisted delivery of ECs is a promising strategy for prevention of vessel lumen narrowing after stent angioplasty procedure.

9.
PLoS One ; 11(6): e0156818, 2016.
Article in English | MEDLINE | ID: mdl-27270230

ABSTRACT

Non-thermal atmospheric pressure plasma has attracted great interest due to its multiple potential biomedical applications with cancer treatment being among the most urgent. To realize the clinical potential of non-thermal plasma, the exact cellular and molecular mechanisms of plasma effects must be understood. This work aimed at studying the prostate cancer specific mechanisms of non-thermal plasma effects on energy metabolism as a central regulator of cell homeostasis and proliferation. It was found that cancer cells with higher metabolic rate initially are more resistant to plasma treated phosphate-buffered saline (PBS) since the respiratory and calcium sensitive signaling systems were not responsive to plasma exposure. However, dramatic decline of cancer oxidative phosphorylation developed over time resulted in significant progression of cell lethality. The normal prostate cells with low metabolic activity immediately responded to plasma treated PBS by suppression of respiratory functions and sustained elevation of cytosolic calcium. However, over time the normal cells start recovering their mitochondria functions, proliferate and restore the cell population. We found that the non-thermal plasma induced increase in intracellular ROS is of primarily non-mitochondrial origin. The discriminate non-thermal plasma effects hold a promise for clinical cancer intervention.


Subject(s)
Antineoplastic Agents/pharmacology , Mitochondria/physiology , Plasma Gases/pharmacology , Prostatic Neoplasms/pathology , Calcium Signaling/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Homeostasis/drug effects , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Prostatic Neoplasms/therapy , Reactive Oxygen Species/metabolism
10.
Nanomedicine ; 12(5): 1335-45, 2016 07.
Article in English | MEDLINE | ID: mdl-26964483

ABSTRACT

Correct localization of epileptic foci can improve surgical outcome in patients with drug-resistant seizures. Our aim was to demonstrate that systemically injected nanoparticles identify activated immune cells, which have been reported to accumulate in epileptogenic brain tissue. Fluorescent and magnetite-labeled nanoparticles were injected intravenously to rats with lithium-pilocarpine-induced chronic epilepsy. Cerebral uptake was studied ex vivo by confocal microscopy and MRI. Cellular uptake and biological effects were characterized in vitro in murine monocytes and microglia cell lines. Microscopy confirmed that the nanoparticles selectively accumulate within myeloid cells in the hippocampus, in association with inflammation. The nanoparticle signal was also detectable by MRI. The in vitro studies demonstrate rapid nanoparticle uptake and good cellular tolerability. We show that nanoparticles can target myeloid cells in epileptogenic brain tissue. This system can contribute to pre-surgical and intra-surgical localization of epileptic foci, and assist in detecting immune system involvement in epilepsy.


Subject(s)
Brain , Epilepsy/surgery , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Animals , Hippocampus , Humans , Inflammation , Mice , Microscopy, Confocal , Rats
11.
Nanomedicine (Lond) ; 11(4): 345-58, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26785794

ABSTRACT

AIM: To achieve high and sustained magnetic particle loading in a proliferative and endocytotically active neural transplant population (astrocytes) through tailored magnetite content in polymeric iron oxide particles. MATERIALS & METHODS: MPs of varying magnetite content were applied to primary-derived rat cortical astrocytes ± static/oscillating magnetic fields to assess labeling efficiency and safety. RESULTS: Higher magnetite content particles display high but safe accumulation in astrocytes, with longer-term label retention versus lower/no magnetite content particles. Magnetic fields enhanced loading extent. Dynamic live cell imaging of dividing labeled astrocytes demonstrated that particle distribution into daughter cells is predominantly 'asymmetric'. CONCLUSION: These findings could inform protocols to achieve efficient MP loading into neural transplant cells, with significant implications for post-transplantation tracking/localization.


Subject(s)
Astrocytes/cytology , Cell Division , Endocytosis , Magnetite Nanoparticles/administration & dosage , Animals , Cells, Cultured , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley
12.
Nanomedicine (Lond) ; 11(3): 189-92, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26783731

Subject(s)
Nanoparticles , Animals , Humans
13.
Nanoscale ; 8(6): 3386-99, 2016 Feb 14.
Article in English | MEDLINE | ID: mdl-26790538

ABSTRACT

Providing the right stimulatory conditions resulting in efficient tissue promoting microenvironment in vitro and in vivo is one of the ultimate goals in tissue development for regenerative medicine. It has been shown that in addition to molecular signals (e.g. growth factors) physical cues are also required for generation of functional cell constructs. These cues are particularly relevant to engineering of biological tissues, within which mechanical stress activates mechano-sensitive receptors, initiating biochemical pathways which lead to the production of functionally mature tissue. Uniform magnetic fields coupled with magnetizable nanoparticles embedded within three dimensional (3D) scaffold structures remotely create transient physical forces that can be transferrable to cells present in close proximity to the nanoparticles. This study investigated the hypothesis that magnetically responsive alginate scaffold can undergo reversible shape deformation due to alignment of scaffold's walls in a uniform magnetic field. Using custom made Helmholtz coil setup adapted to an Atomic Force Microscope we monitored changes in matrix dimensions in situ as a function of applied magnetic field, concentration of magnetic particles within the scaffold wall structure and rigidity of the matrix. Our results show that magnetically responsive scaffolds exposed to an externally applied time-varying uniform magnetic field undergo a reversible shape deformation. This indicates on possibility of generating bending/stretching forces that may exert a mechanical effect on cells due to alternating pattern of scaffold wall alignment and relaxation. We suggest that the matrix structure deformation is produced by immobilized magnetic nanoparticles within the matrix walls resulting in a collective alignment of scaffold walls upon magnetization. The estimated mechanical force that can be imparted on cells grown on the scaffold wall at experimental conditions is in the order of 1 pN, which correlates well with reported threshold to induce mechanotransduction effects on cellular level. This work is our next step in understanding of how to accurately create proper stimulatory microenvironment for promotion of cellular organization to form mature tissue engineered constructs.


Subject(s)
Magnetic Fields , Nanoparticles/chemistry , Stress, Mechanical , Tissue Scaffolds/chemistry , Microscopy, Atomic Force , Nanoparticles/ultrastructure
14.
Nanomedicine (Lond) ; 10(10): 1555-68, 2015 May.
Article in English | MEDLINE | ID: mdl-26008193

ABSTRACT

AIM: To successfully translate magnetically mediated cell targeting from bench to bedside, there is a need to systematically assess the potential adverse effects of magnetic nanoparticles (MNPs) interacting with 'therapeutic' cells. Here, we examined in detail the effects of internalized polymeric MNPs on primary rat endothelial cells' structural intactness, metabolic integrity and proliferation potential. MATERIALS & METHODS: The intactness of cytoskeleton and organelles was studied by fluorescent confocal microscopy, flow cytometry and high-resolution respirometry. RESULTS: MNP-loaded primary endothelial cells preserve intact cytoskeleton and organelles, maintain normal rate of proliferation, calcium signaling and mitochondria energy metabolism. CONCLUSION: This study provides supportive evidence that MNPs at doses necessary for targeting did not induce significant adverse effects on structural integrity and functionality of primary endothelial cells - potential cell therapy vectors.


Subject(s)
Cell- and Tissue-Based Therapy , Endothelium, Vascular/cytology , Magnetics , Nanoparticles , Animals , Cells, Cultured , Rats
15.
Nanomedicine (Lond) ; 10(9): 1391-406, 2015 May.
Article in English | MEDLINE | ID: mdl-25996117

ABSTRACT

AIM: To assess functional competence and gene expression of magnetic nanoparticle (MNP)-loaded primary endothelial cells (ECs) as potential cell-based therapy vectors. MATERIALS & METHODS: A quantitative tube formation, nitric oxide and adhesion assays were conducted to assess functional potency of the MNP-loaded ECs. A quantitative real-time PCR was used to profile genes in both MNP-loaded at static conditions and in vitro targeted ECs. RESULTS: Functional behavior of MNP-loaded and unloaded cells was comparable. MNPs induce expression of genes involved in EC growth and survival, while repress genes involved in coagulation. CONCLUSION: MNPs do not adversely affect cellular function. Gene expression indicates that targeting MNP-loaded ECs to vascular stents may potentially stimulate re-endothelialization of an implant and attenuate neointimal hyperplasia.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Profiling , Magnetics , Nanoparticles , Stents , Animals , Cell Adhesion , Cells, Cultured , Endothelium, Vascular/cytology , Rats
16.
Nanomedicine ; 11(1): 19-29, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25038496

ABSTRACT

Safe and efficient delivery of therapeutic cells to sites of injury/disease in the central nervous system is a key goal for the translation of clinical cell transplantation therapies. Recently, 'magnetic cell localization strategies' have emerged as a promising and safe approach for targeted delivery of magnetic particle (MP) labeled stem cells to pathology sites. For neuroregenerative applications, this approach is limited by the lack of available neurocompatible MPs, and low cell labeling achieved in neural stem/precursor populations. We demonstrate that high magnetite content, self-sedimenting polymeric MPs [unfunctionalized poly(lactic acid) coated, without a transfecting component] achieve efficient labeling (≥90%) of primary neural stem cells (NSCs)-a 'hard-to-label' transplant population of major clinical relevance. Our protocols showed high safety with respect to key stem cell regenerative parameters. Critically, labeled cells were effectively localized in an in vitro flow system by magnetic force highlighting the translational potential of the methods used.


Subject(s)
Biocompatible Materials/chemistry , Magnetite Nanoparticles/chemistry , Nanomedicine/methods , Neural Stem Cells/cytology , Polymers/chemistry , Stem Cell Transplantation , Animals , Cell Differentiation , Cell Proliferation , Humans , Lactic Acid/chemistry , Magnetics , Neurons/metabolism , Polyesters , Regeneration
17.
Methods Mol Biol ; 1181: 83-95, 2014.
Article in English | MEDLINE | ID: mdl-25070329

ABSTRACT

Among the greatest hurdles hindering the successful implementation of tissue-engineered cardiac patch as a therapeutic strategy for myocardial repair is the know-how to promote its rapid integration into the host. We previously demonstrated that prevascularization of the engineered cardiac patch improves cardiac repair after myocardial infarction (MI); the mature vessel networks were generated by including affinity-bound angiogenic factors in the patch and its transplantation on the blood vessel-enriched omentum. Here, we describe a novel in vitro strategy to promote the formation of capillary-like networks in cell constructs without supplementing with angiogenic factors. Endothelial cells (ECs) were seeded into macroporous alginate scaffolds impregnated with magnetically responsive nanoparticles (MNPs), and after pre-culture for 24 h under standard conditions the constructs were subjected to an alternating magnetic field of 40 Hz for 7 days. The magnetic stimulation per se promoted EC organization into capillary-like structures with no supplementation of angiogenic factors; in the non-stimulated constructs, the cells formed sheets or aggregates. This chapter describes in detail the preparation method of the MNP-impregnated alginate scaffold, the cultivation setup for the cell construct under magnetic field conditions, and the set of analyses performed to characterize the resultant cell constructs.


Subject(s)
Alginates/chemistry , Alginates/pharmacology , Cell Culture Techniques/methods , Endothelial Cells/cytology , Magnetic Fields , Neovascularization, Physiologic/drug effects , Tissue Scaffolds/chemistry , Animals , Cattle , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Glucuronic Acid/chemistry , Glucuronic Acid/pharmacology , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacology , Magnetite Nanoparticles/chemistry , Microscopy, Confocal , Tissue Engineering
18.
Nanotechnology ; 25(1): 014009, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24334551

ABSTRACT

Cardiac tissue engineering offers new possibilities for the functional and structural restoration of damaged or lost heart tissue by applying cardiac patches created in vitro. Engineering such functional cardiac patches is a complex mission, involving material design on the nano- and microscale as well as the application of biological cues and stimulation patterns to promote cell survival and organization into a functional cardiac tissue. Herein, we present a novel strategy for creating a functional cardiac patch by combining the use of a macroporous alginate scaffold impregnated with magnetically responsive nanoparticles (MNPs) and the application of external magnetic stimulation. Neonatal rat cardiac cells seeded within the magnetically responsive scaffolds and stimulated by an alternating magnetic field of 5 Hz developed into matured myocardial tissue characterized by anisotropically organized striated cardiac fibers, which preserved its features for longer times than non-stimulated constructs. A greater activation of AKT phosphorylation in cardiac cell constructs after applying a short-term (20 min) external magnetic field indicated the efficacy of magnetic stimulation to actuate at a distance and provided a possible mechanism for its action. Our results point to a synergistic effect of magnetic field stimulation together with nanoparticulate features of the scaffold surface as providing the regenerating environment for cardiac cells driving their organization into functionally mature tissue.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Animals, Newborn , Cell Adhesion , Cells, Cultured , Ferrosoferric Oxide/chemistry , Magnetic Fields , Rats , Rats, Sprague-Dawley , Signal Transduction , Tissue Scaffolds/chemistry
19.
Nanomedicine (Lond) ; 7(9): 1425-42, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22994959

ABSTRACT

Magnetic-based systems utilizing superparamagnetic nanoparticles and a magnetic field gradient to exert a force on these particles have been used in a wide range of biomedical applications. This review is focused on drug targeting applications that require penetration of a cellular barrier as well as strategies to improve the efficacy of targeting in these biomedical applications. Another focus of this review is regenerative applications utilizing tissue engineered scaffolds prepared with the aid of magnetic particles, the use of remote actuation for release of bioactive molecules and magneto-mechanical cell stimulation, cell seeding and cell patterning.


Subject(s)
Drug Delivery Systems/methods , Magnetite Nanoparticles/chemistry , Regenerative Medicine/methods , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Humans , Magnetics/methods , Magnetite Nanoparticles/analysis , Nanomedicine/methods
20.
Biomaterials ; 33(16): 4100-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22417620

ABSTRACT

One of the major challenges in engineering thick, complex tissues such as cardiac muscle, is the need to pre-vascularize the engineered tissue in vitro to enable its efficient integration with host tissue upon implantation. Herein, we explored new magnetic alginate composite scaffolds to provide means of physical stimulation to cells. Magnetite-impregnated alginate scaffolds seeded with aortic endothelial cells stimulated during the first 7 days out of a total 14 day experimental course showed significantly elevated metabolic activity during the stimulation period. Expression of proliferating cell nuclear antigen (PCNA) indicated that magnetically stimulated cells had a lower proliferation index as compared to the non-stimulated cells. This suggests that the elevated metabolic activity could instead be related to cell migration and re-organization. Immunostaining and confocal microscopy analyses supported this observation showing that on day 14 in magnetically stimulated scaffolds without supplementation of any growth factors, cellular vessel-like (loop) structures, known as indicators of vasculogenesis and angiogenesis were formed as compared to cell sheets or aggregates observed in the non-stimulated (control) scaffolds. This work is the first step in our understanding of how to accurately control cellular organization to form tissue engineered constructs, which together with additional molecular signals could lead to a creation of an efficient pre-vascularized tissue construct with potential applicability for transplantation.


Subject(s)
Alginates , Endothelium, Vascular/cytology , Magnetics , Animals , Blotting, Western , Cattle , Cells, Cultured , Ferrosoferric Oxide , Glucuronic Acid , Hexuronic Acids , Microscopy, Electron, Scanning , Proliferating Cell Nuclear Antigen/metabolism , Rheology , Tissue Engineering , Wettability
SELECTION OF CITATIONS
SEARCH DETAIL
...