Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Oncol ; 10: 1659, 2020.
Article in English | MEDLINE | ID: mdl-32984039

ABSTRACT

INTRODUCTION: Colorectal cancer (CC) is the third most common type of cancer, accounting for 10% of all cancer cases. Adjuvant chemotherapy is recommended in stages II-III CC. Wheatgrass juice (WGJ) from wheat seeds has high nutritional values, may induce synergistic benefits to chemotherapy and may attenuate chemotherapy-related side effects. Extracellular vesicles (EVs) are subcellular membrane blebs. EVs include exosomes (generated in the endosome, in size <150 nm) and microvesicles (shed from the plasma cell membrane) provide information on their parental cells and play a role in intercellular communication. We aimed to elucidate the effects of chemotherapy administration with supportive treatment of WGJ on CC patients' EVs characteristics. METHODS: EVs were isolated from the blood samples of 15 healthy controls (HCs) and 50 CC patients post-surgery, treated by chemotherapy, with or without additional daily WGJ. Blood samples were taken before, during, and at the end of chemotherapy. EVs were characterized by size, concentration, membrane antigens and cytokine content using nanoparticle-tracking analysis, western blot, flow cytometry, and protein array methods. RESULTS: EVs were found to be similar by size and concentration with reduced levels of exosome markers (CD81) on samples at the end of combined treatment (chemotherapy and WGJ). Higher levels of endothelial EVs, which may indicate impairment of the vascular endothelial cells during treatment, were found in CC patients treated by chemotherapy only compared to those with chemotherapy and daily WGJ. Also, EVs thrombogenicity was lower in patients added WGJ compared to patients who had only chemotherapy (levels of tissue factor p = 0.029 and endothelial protein C receptor p = 0.005). Following treatments, levels of vascular endothelial growth factor receptors (VEGFR-1) and the majority of growth-factors/pro-inflammatory cytokines were higher in EVs of patients treated by chemotherapy only than in EVs obtained from patients with the combined treatment. CONCLUSION: Daily consumption of WGJ during chemotherapy may reduce vascular damage and chemotherapy-related thrombogenicity, growth factors and cytokines, as reflected by the characteristics of patient's EVs.

2.
Adv Healthc Mater ; 8(10): e1801589, 2019 05.
Article in English | MEDLINE | ID: mdl-30963725

ABSTRACT

Nanoghosts (NGs) are nanovesicles reconstructed from the cytoplasmic membranes of mesenchymal stem cells (MSCs). By retaining MSC membranes, the NGs retain the ability of these cells to home in on multiple tumors, laying the foundations, thereby, for the development of a targeted drug delivery platform. The susceptibility of MSCs to functional changes, following their exposure to cytokines or cancer-derived conditioned-media (CM), presents the opportunity to modify the NGs by conditioning their source cells. This opportunity is investigated by comparing the membrane protein composition and the tumor uptake of NGs derived from naïve MSCs (N-NG) against conditioned NGs made from MSCs pre-treated with conditioned-media (CM-NG) or with a mix of the proinflammatory cytokines TNF-α and IL-1ß (Cyto-NG). CM-NGs are found to be more targeted towards immune cells than Cyto- or N-NGs, while Cyto-NGs are the most tumor-targeted ones, with similar immune-targeting capacity as N-NGs but with a higher affinity towards endothelial cells. Proteomic variations were wider in the CM-NGs, with exceptionally higher levels of ICAM-1 compared to N- and Cyto-NGs. From a translational point of view, the data show that the tumor-targeting ability of the NGs, and possibly that of other MSC-derived extracellular vesicles, can be enhanced by simple conditioning of their source cells.


Subject(s)
Cell Membrane/metabolism , Culture Media, Conditioned/pharmacology , Cytokines/pharmacology , Mesenchymal Stem Cells/drug effects , Animals , Cell Line , Cell Membrane/chemistry , Humans , Integrins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Nanostructures/chemistry , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Proteome/metabolism
3.
J Control Release ; 293: 215-223, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30527755

ABSTRACT

The rapid development of biomimetic cell membrane-based nanoparticles is still overshadowed by many practical challenges, one of which is the difficulty to precisely measure the biodistribution of such nanoparticles. Currently, this challenge is mostly addressed using fluorescent techniques with limited sensitivity, or radioactive labeling methods, which rarely account for the nanoparticles themselves, but their payloads instead. Here we report the development of a robust method for the innate radioactive labeling of cells and membrane-based nanoparticles and their consequent sensitive detection and biodistribution measurements. The preclinical potential of this method was demonstrated with Nano-Ghosts (NGs), manufactured from the cytoplasmic membranes of mesenchymal stem cells cultured with radioactively-labeled linoleic acid and achieving a cell labeling efficiency of 36%. Radiolabeling did not affect the physiochemical properties of the NGs, which stably retained their radiolabels. Using radioactivity measurements, we are now able to determine precisely the amount of NGs uptaken by tissues and cells, thereby providing further support to our presumed active NG targeting mechanisms. Biodistribution studies comparing radiolabeled NGs to fluorescently-labeled ones have validated our method and revealed new information, which could not be obtained otherwise, regarding the NGs' unique kinetics and rapid clearance, supporting their excellent safety profiles. The reported approach may be expanded to other membrane-based entities to facilitate and hasten their preclinical development and be used in parallel with other labeling methods to provide different and additional information.


Subject(s)
Cell Membrane , Mesenchymal Stem Cells , Nanostructures/administration & dosage , A549 Cells , Animals , Carbon Radioisotopes , Humans , Linoleic Acid/administration & dosage , Male , Mice, Inbred C57BL , Mice, Nude , Tissue Distribution
4.
Cancer Res ; 78(5): 1253-1265, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29301792

ABSTRACT

Stromal cells residing in the tumor microenvironment contribute to the development of therapy resistance. Here we show that chemotherapy-educated mesenchymal stem cells (MSC) promote therapy resistance via cross-talk with tumor-initiating cells (TIC), a resistant tumor cell subset that initiates tumorigenesis and metastasis. In response to gemcitabine chemotherapy, MSCs colonized pancreatic adenocarcinomas in large numbers and resided in close proximity to TICs. Furthermore, gemcitabine-educated MSCs promoted the enrichment of TICs in vitro and enhance tumor growth in vivo These effects were dependent on the secretion of CXCL10 by gemcitabine-educated MSCs and subsequent activation of the CXCL10-CXCR3 axis in TICs. In an orthotopic pancreatic tumor model, targeting TICs using nanovesicles (called nanoghosts) derived from MSC membranes and loaded with a CXCR3 antagonist enhanced therapy outcome and delayed tumor regrowth when administered in combination with gemcitabine. Overall, our results establish a mechanism through which MSCs promote chemoresistance, and propose a novel drug delivery system to target TICs and overcome this resistance.Significance: These results establish a mechanism by which mesenchyme stem cells in the tumor microenvironment promote chemoresistance, and they propose a novel drug delivery system to overcome this challenge. Cancer Res; 78(5); 1253-65. ©2018 AACR.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/pathology , Deoxycytidine/analogs & derivatives , Lung Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Communication , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Deoxycytidine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
5.
Biomater Sci ; 5(6): 1183-1194, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28513656

ABSTRACT

Various extracellular matrix (ECM) scaffolds, isolated through decellularization, were suggested as ideal biomimetic materials for 'Functional tissue engineering' (FTE). The decellularization process comprises a compromise between damaging and preserving the ultrastructure and composition of ECM-previously shown to affect cell survival, proliferation, migration, organization, differentiation and maturation. Inversely, the effects of cells on the ECM constructs' biophysical properties, under physiological-like conditions, remain still largely unknown. We hypothesized that by re-cellularizing porcine cardiac ECM (pcECM, as a model scaffold) some of the original biophysical properties of the myocardial tissue can be restored, which are related to the scaffold's surface and the bulk modifications consequent to cellularization. We performed a systematic biophysical assessment of pcECM scaffolds seeded with human mesenchymal stem cells (MSCs), a common multipotent cell source in cardiac regenerative medicine. We report a new type of FTE study in which cell interactions with a composite-scaffold were evaluated from the perspective of their contribution to the biophysical properties of the construct surface (FTIR, WETSEM™) and bulk (DSC, TGA, and mechanical testing). The results obtained were compared with acellular pcECM and native ventricular tissue serving as negative and positive controls, respectively. MSC recellularization resulted in an inter-fiber plasticization effect, increased protein density, masking of acylated glycosaminoglycans (GAGs) and active pcECM remodelling which further stabilized the reseeded construct and increased its denaturation resistance. The systematic approach presented herein, therefore, identifies cells as "biological plasticizers" and yields important methodologies, understanding, and data serving both as a reference as well as possible 'design criteria' for future studies in FTE.


Subject(s)
Extracellular Matrix/chemistry , Mesenchymal Stem Cells/cytology , Myocardium/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Glycosaminoglycans/chemistry , Humans , Myocardium/chemistry , Swine , Tensile Strength
6.
Tissue Eng Part A ; 23(1-2): 69-79, 2017 01.
Article in English | MEDLINE | ID: mdl-27784199

ABSTRACT

Effective cellularization is a key approach to prevent small-caliber (<4 mm) tissue-engineered vascular graft (TEVG) failure and maintain patency and contractility following implantation. To achieve this goal, however, improved biomimicking designs and/or relatively long production times (typically several months) are required. We previously reported on porcine carotid artery decellularization yielding biomechanically stable and cell supportive small-caliber (3-4 mm diameter, 5 cm long) arterial extracellular matrix (scaECM) vascular grafts. In this study, we aimed to study the scaECM graft patency in vivo and possibly improve that patency by graft pre-endothelialization with the recipient porcine autologous cells using our previously reported custom-designed dynamic perfusion bioreactor system. Decellularized scaECM vascular grafts were histologically characterized, their immunoreactivity studied in vitro, and their biocompatibility profile evaluated as a xenograft subcutaneous implantation in a mouse model. To study the scaECM cell support and remodeling ability, pig autologous endothelial and smooth muscle cells (SMCs) were seeded and dynamically cultivated within the scaECM lumen and externa/media, respectively. Finally, endothelialized-only scaECMs-hypothesized as a prerequisite for maintaining graft patency and controlling intimal hyperplasia-were transplanted as an interposition carotid artery graft in a porcine model. Graft patency was evaluated through angiography online and endpoint pathological assessment for up to 6 weeks. Our results demonstrate the scaECM-TEVG biocompatibility preserving a structurally and mechanically stable vascular wall not just following decellularization and recellularization but also after implantation. Using our dynamic perfusion bioreactor, we successfully demonstrated the ability of this TEVG to support in vitro recellularization and remodeling by primary autologous endothelial and SMCs, which were seeded on the lumen and the externa/media layers, respectively. Following transplantation, dynamically endothelialized scaECM-TEVGs remained patent for 6 weeks in a pig carotid interposition bypass model. When compared with nonrevitalized control grafts, reendothelialized grafts provided excellent antithrombogenic activity, inhibited intimal hyperplasia formation, and encouraged media wall infiltration and reorganization with recruited host SMCs. We thus demonstrate that readily available decellularized scaECM can be promptly revitalized with autologous cells in a 3-week period before implantation, indicating applicability as a future platform for vascular reconstructive procedures.


Subject(s)
Blood Vessel Prosthesis Implantation , Blood Vessel Prosthesis , Carotid Arteries/surgery , Extracellular Matrix , Animals , Autografts , Bioprosthesis , Carotid Arteries/physiopathology , Mice , Swine
7.
Nano Lett ; 16(3): 1574-82, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26901695

ABSTRACT

Nanoghosts derived from mesenchymal stem cells and retaining their unique surface-associated tumor-targeting capabilities were redesigned as a selective and safe universal nonviral gene-therapy platform. pDNA-loaded nanoghosts efficiently targeted and transfected diverse cancer cells, in vitro and in vivo, in subcutaneous and metastatic orthotopic tumor models, leading to no adverse effects. Nanoghosts loaded with pDNA encoding for a cancer-toxic gene inhibited the growth of metastatic orthotopic lung cancer and subcutaneous prostate cancer models and dramatically prolonged the animals' survival.


Subject(s)
DNA/administration & dosage , Gene Transfer Techniques , Genetic Therapy , Lung Neoplasms/therapy , Mesenchymal Stem Cells , Nanostructures , Prostatic Neoplasms/therapy , Animals , Cell Line, Tumor , DNA/genetics , DNA/therapeutic use , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Nanostructures/administration & dosage , Nanostructures/adverse effects , Nanostructures/ultrastructure , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/therapeutic use , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
8.
Biomed Microdevices ; 18(2): 23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26888439

ABSTRACT

Strategies for cancer protein vaccination largely aim to activate the cellular arm of the immune system against cancer cells. This approach, however, is limited since protein vaccines mostly activate the system's humoral arm instead. One way to overcome this problem is to enhance the cross-presentation of such proteins by antigen-presenting cells, which may consequently lead to intense cellular response. Here we examined the ability of listeriolysin O (LLO) incorporated into poly-lactic-co-glycolic acid (PLGA) microspheres to modify the cytosolic delivery of low molecular weight peptides and enhance their cross-presentation. PLGA microspheres were produced in a size suitable for uptake by phagocytic cells. The peptide encapsulation and release kinetics were improved by adding NaCl to the preparation. PLGA microspheres loaded with the antigenic peptide and incorporated with LLO were readily up-taken by phagocytic cells, which exhibited an increase in the expression of peptide-MHC-CI complexes on the cell surface. Furthermore, this system enhanced the activation of a specific T hybridoma cell line, thus simulating cytotoxic T cells. These results establish, for the first time, a proof of concept for the use of PLGA microspheres incorporated with a pore-forming agent and the antigen peptide of choice as a unique cancer protein vaccination delivery platform.


Subject(s)
Antigens, Neoplasm/chemistry , Bacterial Toxins/chemistry , Cytosol/metabolism , Drug Carriers/chemistry , Heat-Shock Proteins/chemistry , Hemolysin Proteins/chemistry , Lactic Acid/chemistry , Microspheres , Polyglycolic Acid/chemistry , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Biological Transport , Cell Survival , Cross-Priming , Drug Liberation , Mice , Ovalbumin/immunology , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Phagocytes/cytology , Phagocytes/immunology , Polylactic Acid-Polyglycolic Acid Copolymer , RAW 264.7 Cells
9.
Biomed Microdevices ; 17(4): 69, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26044202

ABSTRACT

Studies with AZD2171-a new anti-angiogenic inhibitor of tyrosine kinases associated with VEGF signaling-have shown great promise for treating glioblastoma. Unfortunately, AZD2171 success is limited by low permeability through the blood-brain barrier. Due to AZD2171's short half-life and high toxicity, its local administration will require multiple intracranial procedures, making this approach clinically unfeasible. In this study, we investigated the potential of the highly hydrophobic AZD2171, released from modified polylactic-co-glycolic acid microspheres (PLGA-MS), to treat glioblastoma. To further demonstrate the versatile loading capacity of this system, the same PLGA formulation, which was found optimal for the loading and release of AZD2171, was tested with sTRAIL/Apo2L-a biologic drug that is very different than AZD2171 in its molecular weight, solubility, and charge. AZD2171 released from PLGA-MS was at least effective as the free drug in inhibiting endothelial growth and proliferation (in vitro), and, surprisingly, had a profound cytotoxic effect also towards in vitro cultured glioblastoma cell-lines (U87 and A172). Complete tumor inhibition was achieved following a single treatment with AZD2171-loaded PLGA-MS (6 (mg)/kg) administered locally adjacent to human U87 glioma tumors inoculated subcutaneously in nude mice. This improved effect, compared to other therapeutic approaches involving AZD2171, was shown to affect both tumor vasculature and the glioma cells. sTRAIL-loaded microspheres, administered at very low doses (0.3 (mg)/kg), led to 35 % inhibition of tumor growth in 2 weeks. Collectively, our results provide pre-clinical evidence for the potential of PLGA formulations of AZD2171 and sTRAIL to serve as an effective treatment for glioblastoma.


Subject(s)
Glioblastoma/drug therapy , Microspheres , Quinazolines/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Cell Line, Tumor , Delayed-Action Preparations/pharmacology , Disease Models, Animal , Glycolates/chemistry , Humans , Lactic Acid/chemistry , Male , Mice , Mice, Nude , Polyesters , Polymers/chemistry , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
10.
Tissue Eng Part A ; 21(9-10): 1507-19, 2015 May.
Article in English | MEDLINE | ID: mdl-25602926

ABSTRACT

Functional vascularization is a prerequisite for cardiac tissue engineering of constructs with physiological thicknesses. We previously reported the successful preservation of main vascular conduits in isolated thick acellular porcine cardiac ventricular ECM (pcECM). We now unveil this scaffold's potential in supporting human cardiomyocytes and promoting new blood vessel development ex vivo, providing long-term cell support in the construct bulk. A custom-designed perfusion bioreactor was developed to remodel such vascularization ex vivo, demonstrating, for the first time, functional angiogenesis in vitro with various stages of vessel maturation supporting up to 1.7 mm thick constructs. A robust methodology was developed to assess the pcECM maximal cell capacity, which resembled the human heart cell density. Taken together these results demonstrate feasibility of producing physiological-like constructs such as the thick pcECM suggested here as a prospective treatment for end-stage heart failure. Methodologies reported herein may also benefit other tissues, offering a valuable in vitro setting for "thick-tissue" engineering strategies toward large animal in vivo studies.


Subject(s)
Extracellular Matrix/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bioreactors , Coculture Techniques , Feasibility Studies , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Myocardium/cytology , Sus scrofa
11.
Biomatter ; 4: e28238, 2014.
Article in English | MEDLINE | ID: mdl-24553126

ABSTRACT

Human umbilical vein endothelial cells (HUVECs) were successfully entrapped in polyethylene oxide (PEO) core /polycaprolactone (PCL) shell electrospun fibers thus creating a "bioactive fiber." The viability and release of biomolecules from the entrapped cells in the bioactive fibers were characterized. A key modification to the core solution was the inclusion of 50% fetal bovine serum (FBS), which improved cell viability substantially. The fluorescein diacetate (FDA) staining revealed that the entrapped cells were intact and viable immediately after the electrospinning process. A long-term cell viability assay using AlamarBlue® showed that cells were viable for over two weeks. Secreted Interleukin-8 (IL-8) was monitored as a candidate released protein, which can also act as an indicator of HUVEC stress. These results demonstrated that HUVECs could be entrapped within the electrospun scaffold with the potential of controllable cell deposition and the creation of a bioactive fibrous scaffold with extended functionality.


Subject(s)
Biocompatible Materials/chemistry , Human Umbilical Vein Endothelial Cells , Tissue Scaffolds/chemistry , Cell Survival/drug effects , Cells, Immobilized , Humans , Interleukin-8/metabolism , Polyesters/chemistry , Polyethylene Glycols/chemistry , Porosity , Solutions/chemistry , Tissue Engineering/methods
12.
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
13.
Tissue Eng Part C Methods ; 19(8): 620-30, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23265414

ABSTRACT

The clinical success of tissue-engineered constructs commonly requires mechanical properties that closely mimic those of the human tissue. Determining the viscoelastic properties of such biomaterials and the factors governing their failure profiles, however, has proven challenging, although collecting extensive data regarding their tensile behavior is straightforward. The easily calculated Young's modulus remains the most reported mechanical measure, regardless of its limitations, even though single-relaxation-time (SRT) models can provide much more information, which remain scarce due to a lack of manageable tools for implementing these models. We developed an easy-to-use algorithm for applying the Zener SRT model and determining the elastic moduli, viscosity, and failure profiles of materials under different mechanical tests in a user-independent manner. The algorithm was validated on the data resulting from tensile tests on native and decellularized porcine cardiac tissue, previously suggested as a promising scaffold material for cardiac tissue engineering. This analysis yields new and more accurate measurements such as the elastic moduli and viscosity, the model's relaxation time, and information on the factors governing the materials' failure profiles. These measurements indicate that the viscoelasticity and strength of the decellularized acellular extracellular matrix (ECM) are similar to those of native tissue, although its elasticity and apparent viscosity are higher. Nonetheless, reseeding and culturing the ECM with mesenchymal stem cells was shown to partially restore the mechanical properties lost after decellularization. We propose this algorithm as a platform for soft-tissue analysis that can provide comparable and unbiased measures for characterizing viscoelastic biomaterials commonly used in tissue engineering.


Subject(s)
Elastic Modulus , Extracellular Matrix/chemistry , Models, Biological , Myocardium/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Humans , Swine , Viscosity
14.
Tissue Eng Part A ; 19(9-10): 1155-64, 2013 May.
Article in English | MEDLINE | ID: mdl-23216214

ABSTRACT

In most tissue engineering applications, understanding the factors affecting the growth dynamics of coculture systems is crucial for directing the population toward a desirable regenerative process. Yet, no comprehensive analysis method exists to quantify coculture population dynamics, let alone, a unifying model addressing the "environmental" factors influencing cell growth, all together. Here we suggest a modification of the Lotka-Volterra model to analyze the population dynamics of cocultured cells and predict their growth profiles for tissue engineering applications. This model, commonly used to describe the population dynamics of a prey and predator sharing a closed ecological niche, was found to fit our empirical data on cocultures of endothelial cells (ECs) and mesenchymal stem cells (MSCs) that have been widely investigated for their regenerative potential. Applying this model to cocultures of this sort allows us to quantify the effect that culturing conditions have on the way cell growth is affected by the same cells or by the other cells in the coculture. We found that in most cases, EC growth was inhibited by the same cells but promoted by MSCs. The principles resulting from this analysis can be used in various applications to guide the population toward a desired direction while shedding new light on the fundamental interactions between ECs and MSCs. Similar results were also demonstrated on complex substrates made from decellularized porcine cardiac extracellular matrix, where growth occurred only after coculturing ECs and MSCs together. Finally, this unique implementation of the Lotka-Volterra model may also be regarded as a roadmap for using such models with other potentially regenerative cocultures in various applications.


Subject(s)
Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Models, Theoretical , Cell Survival , Cells, Cultured , Humans , Tissue Engineering
15.
Tissue Eng Part A ; 18(19-20): 2125-37, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22663095

ABSTRACT

The decellularization of porcine heart tissue offers many opportunities for the production of physiologically relevant myocardial mimetic scaffolds. Earlier, we reported the successful isolation of a thin porcine cardiac extracellular matrix (pcECM) exhibiting relevant bio-mechanical properties for myocardial tissue engineering. Nevertheless, since native cardiac tissue is much thicker, such thin scaffolds may offer limited regeneration capacity. However, generation of thicker myocardial mimetic tissue constructs is hindered by diffusion limitations (~100 µm), and the lack of a proper vascular-like network within these constructs. In our present work, we focused on optimizing the decellularization procedure for thicker tissue slabs (10-15 mm), while retaining their inherent vasculature, and on characterizing the resulting pcECM. The trypsin/Triton-based perfusion procedure that resulted in a nonimmunogenic and cell-supportive pcECM was found to be more effective in cell removal and in the preservation of fiber morphology and structural characteristics than stirring, sonication, or sodium dodecyl sulfate/Triton-based procedures. Mass spectroscopy revealed that the pcECM is mainly composed of ECM proteins with no apparent cellular protein remains. Mechanical testing indicated that the obtained pcECM is viscoelastic in nature and possesses the typical stress-strain profile of biological materials. It is stiffer than native tissue yet exhibits matched mechanical properties in terms of energy dissipation, toughness, and ultimate stress behavior. Vascular network functionality was maintained to the first three-four branches from the main coronary vessels. Taken together, these results reaffirm the efficiency of the decellularization procedure reported herein for yielding thick nonimmunogenic cell-supportive pcECM scaffolds, preserving both native tissue ultra-structural properties and an inherent vascular network. When reseeded with the appropriate progenitor cells, these scaffolds can potentially serve as ex vivo screening platforms for new therapeutics, as models for human cardiac ECM, or as biomedical constructs for patch or transmural transplantation strategies.


Subject(s)
Extracellular Matrix/chemistry , Myocardium/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Adhesion/physiology , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Microscopy, Electron, Scanning , Rats , Swine
16.
J Control Release ; 151(2): 139-48, 2011 Apr 30.
Article in English | MEDLINE | ID: mdl-21362450

ABSTRACT

Anti-retroviral-therapies against HIV/AIDS focus on inhibiting viral growth and may slow AIDS progression, but not cure the disease. Here we describe an approach to treat HIV as a cellular pathology by targeting cell derived liposomes against HIV-infected cells. Cell-derived-liposomes were prepared from the cytoplasmatic membranes of cells expressing CCR5, the human receptor for gp120, that is found on the surface of virions and HIV-infected cells. The specific targeting and cytotoxicity of the cell-derived liposomes towards gp120-expressing cells were studied. Cell-derived liposomes exhibited unilamellar morphology and were found to be of 100-200 nm in diameter. Moreover, CCR5 that was expressed on the surface of the cell-derived liposomes was biologically active and correctly oriented. Cell-derived liposomes incubated with HIV-infected model cells exhibited significant and specific targeting to those gp120-expressing cells. To demonstrate the system efficacy, EDTA was selected as liposomal encapsulate and was shown to cause high cytotoxic effect when introduced into the cell cytoplasm. Finally, cell-derived liposomes containing EDTA led to a 60% reduction in the viability of gp120-expressing cells compared to no effect on control cells that do not express gp120. These results demonstrate the specific targeting and cytotoxic effect of CCR5-conjugated cell-derived liposomes towards gp120-expressing HIV model cells, suggesting for a potential new therapeutic approach.


Subject(s)
Drug Delivery Systems/methods , HIV Infections/drug therapy , HIV Infections/metabolism , HIV-1/metabolism , Receptors, CCR5/biosynthesis , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/metabolism , Cattle , Coculture Techniques , Cricetinae , HEK293 Cells , HIV Envelope Protein gp120/biosynthesis , HIV Infections/pathology , HIV-1/drug effects , Humans , Jurkat Cells , Liposomes , Receptors, CCR5/therapeutic use
17.
J Neurol ; 256(8): 1228-35, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19280101

ABSTRACT

Several recent studies proposed a role for innate immunity and inflammation in the pathogenesis of amyotrophic lateral sclerosis (ALS). However, possible links, if any, between disease and adaptive immunity are poorly understood. The present study probed for the role of B cells in ALS disease using the G93A-SOD-1 transgenic mouse model. In agreement with other studies, we show here that autoantibodies are detectable in SOD-1 mice. However, SOD-1 B cells did not express any altered phenotype and exhibited indistinguishable responsiveness to immunogenic stimuli relative to wild-type B cells. This was obtained for B cells isolated before, during and after the onset of ALS-like disease. Finally, to obtain an in vivo conclusion, we generated SOD-1 mice that are deficient of B cells, by crossing SOD-1 mice with Igmu-deficient mice (muMT), where B cell development is blocked at the proB stage. The meteoric assays performed on a rota-rod clearly showed the development of ALS-like disease in SOD-1 mice that are deficient of B cells not differently than in control SOD-1 mice. Our results propose that B lymphocytes do not have a major role in the pathogenesis of ALS-like disease in SOD-1 mice.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Antibody Formation/immunology , B-Lymphocytes/immunology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Autoantibodies/immunology , Autoimmunity/genetics , Autoimmunity/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Immunoglobulin Subunits/genetics , Immunoglobulin Subunits/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Depletion/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL
...