Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 13(4): 599-611, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31495714

ABSTRACT

The derivation of human brain capillary endothelial cells is of utmost importance for drug discovery programs focusing on diseases of the central nervous system. Here, we describe a two-step differentiation protocol to derive brain capillary-like endothelial cells from human pluripotent stem cells. The cells were initially differentiated into endothelial progenitor cells followed by specification into a brain capillary-like endothelial cell phenotype using a protocol that combined the induction, in a time-dependent manner, of VEGF, Wnt3a, and retinoic acid signaling pathways and the use of fibronectin as the extracellular matrix. The brain capillary-like endothelial cells displayed a permeability to lucifer yellow of 1 × 10-3 cm/min, a transendothelial electrical resistance value of 60 Ω cm2 and were able to generate a continuous monolayer of cells expressing ZO-1 and CLAUDIN-5 but moderate expression of P-glycoprotein. Further maturation of these cells required coculture with pericytes. The study presented here opens a new approach for the study of soluble and non-soluble factors in the specification of endothelial progenitor cells into brain capillary-like endothelial cells.


Subject(s)
Cell Differentiation , Endothelial Progenitor Cells/cytology , Pluripotent Stem Cells/cytology , Biomarkers , Blood-Brain Barrier/cytology , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Endothelium, Vascular/cytology , Extracellular Matrix/metabolism , Humans , Immunophenotyping , Pluripotent Stem Cells/metabolism
2.
Nanoscale ; 11(28): 13243-13248, 2019 Jul 18.
Article in English | MEDLINE | ID: mdl-31290510

ABSTRACT

Here we report a two-step surface modification methodology to radiolabel small extracellular vesicles (SEVs) with 64CuCl2 for PET/MRI imaging. The modification did not change or damage the morphology, surface receptor proteins and internal RNA content. Radiolabeled SEVs could be detected in organs with low accumulation such as the brain (0.4-0.5% ID g-1) and their brain location determined by MRI.


Subject(s)
Copper/metabolism , Extracellular Vesicles/metabolism , Magnetic Resonance Imaging , Multimodal Imaging , Positron-Emission Tomography , Animals , Brain/diagnostic imaging , Brain/metabolism , Copper/chemistry , Extracellular Vesicles/chemistry , Extracellular Vesicles/ultrastructure , Humans , Mice , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism
3.
J Control Release ; 284: 57-72, 2018 08 28.
Article in English | MEDLINE | ID: mdl-29902485

ABSTRACT

Stimulation of adult neurogenesis by targeting the endogenous neural stem cells (NSCs), located in hippocampus and subventricular zone (SVZ), with nanoformulations has been proposed for brain repair in cases of neurodegenerative diseases. Unfortunately, it is relatively unknown the nanoformulation properties to facilitate their accumulation in the neurogenic niches after intravenous injection. Here, we have screened different gold-based formulations having variable morphology, surface chemistry and responsiveness to light for their capacity to cross the blood brain barrier (BBB) and accumulate preferentially in the neurogenic niches. Results obtained in a human in vitro BBB model showed that gold nanoparticles (Au NPs) and gold nanorods (Au NRs) conjugated with medium density of transferrin (Tf) peptides (i.e. between 169 and 230 peptides per NP) crossed more efficiently the BBB than the remaining formulations. This is due to a relatively lower avidity of these formulations to Tf receptor (TfR) and lower accumulation in the lysosomes, as compared to the other formulations. We further show that the near infrared light (NIR) irradiation of Au NRs, under a certain concentration and at specific cell culture time, lead to the opening of the BBB. Finally, we demonstrate that Au NRs conjugated with Tf administered intravenously in mice and activated by NIR had the highest accumulation in the neurogenic niches. Our results open the possibility of targeting more effectively the neurogenic niches by controlling the properties of the nanoformulations.


Subject(s)
Brain/metabolism , Drug Carriers/metabolism , Gold/metabolism , Nanoparticles/metabolism , Transferrin/metabolism , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Cell Line , Drug Carriers/analysis , Gold/analysis , Humans , Metal Nanoparticles/chemistry , Mice , Nanoparticles/analysis , Nanotubes/analysis , Neurogenesis , Transferrin/analysis
4.
J Control Release ; 194: 138-47, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25173841

ABSTRACT

Wound treatment remains one of the most prevalent and economically burdensome healthcare issues in the world. Poly (lactic-co-glycolic acid) (PLGA) supplies lactate that accelerates neovascularization and promotes wound healing. LL37 is an endogenous human host defense peptide that modulates wound healing and angiogenesis and fights infection. Hence, we hypothesized that the administration of LL37 encapsulated in PLGA nanoparticles (PLGA-LL37 NP) promotes wound closure due to the sustained release of both LL37 and lactate. In full thickness excisional wounds, the treatment with PLGA-LL37 NP significantly accelerated wound healing compared to PLGA or LL37 administration alone. PLGA-LL37 NP-treated wounds displayed advanced granulation tissue formation by significant higher collagen deposition, re-epithelialized and neovascularized composition. PLGA-LL37 NP improved angiogenesis, significantly up-regulated IL-6 and VEGFa expression, and modulated the inflammatory wound response. In vitro, PLGA-LL37 NP induced enhanced cell migration but had no effect on the metabolism and proliferation of keratinocytes. It displayed antimicrobial activity on Escherichia coli. In conclusion, we developed a biodegradable drug delivery system that accelerated healing processes due to the combined effects of lactate and LL37 released from the nanoparticles.


Subject(s)
Cathelicidins/administration & dosage , Cathelicidins/pharmacology , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides , Cell Movement/drug effects , Collagen/metabolism , Dose-Response Relationship, Drug , Drug Delivery Systems , Epithelial Cells/drug effects , Escherichia coli/drug effects , Female , Granulation Tissue/drug effects , Inflammation/pathology , Keratinocytes/drug effects , Mice , Neovascularization, Physiologic/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer , Wounds and Injuries/pathology
5.
Sci Transl Med ; 6(218): 218ra6, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24401941

ABSTRACT

Currently, there are no clinically approved surgical glues that are nontoxic, bind strongly to tissue, and work well within wet and highly dynamic environments within the body. This is especially relevant to minimally invasive surgery that is increasingly performed to reduce postoperative complications, recovery times, and patient discomfort. We describe the engineering of a bioinspired elastic and biocompatible hydrophobic light-activated adhesive (HLAA) that achieves a strong level of adhesion to wet tissue and is not compromised by preexposure to blood. The HLAA provided an on-demand hemostatic seal, within seconds of light application, when applied to high-pressure large blood vessels and cardiac wall defects in pigs. HLAA-coated patches attached to the interventricular septum in a beating porcine heart and resisted supraphysiologic pressures by remaining attached for 24 hours, which is relevant to intracardiac interventions in humans. The HLAA could be used for many cardiovascular and surgical applications, with immediate application in repair of vascular defects and surgical hemostasis.


Subject(s)
Biocompatible Materials/therapeutic use , Carotid Arteries/pathology , Carotid Arteries/surgery , Heart Defects, Congenital/drug therapy , Heart Defects, Congenital/surgery , Minimally Invasive Surgical Procedures/instrumentation , Tissue Adhesives/therapeutic use , Animals , Biocompatible Materials/pharmacology , Blood , Carotid Arteries/drug effects , Collagen/pharmacology , Elasticity , Heart Defects, Congenital/diagnostic imaging , Humans , Male , Materials Testing , Rats , Rats, Wistar , Sus scrofa , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Tissue Engineering , Ultrasonography , Ultraviolet Rays
6.
Adv Healthc Mater ; 3(4): 565-71, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24106240

ABSTRACT

The use of tissue adhesives for internal clinical applications is limited due to a lack of materials that balance strong adhesion with biocompatibility. The use of substrate topography is explored to reduce the volume of a highly reactive and toxic glue without compromising adhesive strength. Micro-textured patches coated with a thin layer of cyanoacrylate glue achieve similar adhesion levels to patches employing large amounts of adhesive, and is superior to the level of adhesion achieved when a thin coating is applied to a non-textured patch. In vivo studies demonstrate reduced tissue inflammation and necrosis for patterned patches with a thinly coated layer of reactive glue, thus overcoming a significant challenge with existing tissue adhesives such as cyanoacrylate. Closure of surgical stomach and colon defects in a rat model is achieved without abdominal adhesions. Harnessing the synergy between surface topography and reactive chemistry enables controlled tissue adhesion with an improved biocompatibility profile without requiring changes in the chemical composition of reactive tissue glues.


Subject(s)
Abdominal Wound Closure Techniques/instrumentation , Biocompatible Materials/chemistry , Cyanoacrylates/chemistry , Inflammation/chemically induced , Tissue Adhesives/chemistry , Animals , Biocompatible Materials/adverse effects , Colon/drug effects , Colon/pathology , Colon/surgery , Cyanoacrylates/adverse effects , Female , Inflammation/pathology , Necrosis , Rats , Stomach/drug effects , Stomach/pathology , Stomach/surgery , Surface Properties , Tissue Adhesives/adverse effects
7.
Dent Mater ; 29(10): e252-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23981323

ABSTRACT

OBJECTIVES: To evaluate the antifungal activity, biocompatibility and mechanical properties of dental resins containing silica nanoparticles functionalized with amphotericin B (SNP-DexOxAmB) against five species of Candida. METHODS: Dental resin composites (Spectrum, Dentsply DeTrey, GmbH, Germany) having 2% (w/w) of SNP-DexOxAmB (SNPs of 5 and 80nm, denoted as SNP5 and SNP80) were aged for 10, 20 and 30 days at 37°C, in phosphate buffer saline buffer pH 7.4 (PBS). At different time, the antifungal activity was evaluated by a direct contact assay against 1×10(4)cells of Candida. The biocompatibility of the resins was tested against human fibroblasts, endothelial cells and red blood cells. RESULTS: Dental resins containing SNP5-DexOxAmB have high (1×10(4)cells killed in 5h by ∼70mg of dental resin composite containing 2% (w/w) of SNP-DexOxAmB) and durable (for at least 1 month) antifungal activity against five strains of Candida. The incorporation of the nanoparticles (NPs) had no significant change in the mechanical properties of the resin, specifically the flexural strength and modulus. Our results further show that the antifungal activity is mainly mediated by direct contact and not by leaching of NPs from the resin. Resins incorporating SNP5-DexOxAmB have longer-term antifungal activity than SNP80-DexOxAmB. The antimicrobial activity of resins with SNP5-DexOxAmB persists after 4 cycles of re-use and it is superior to the activity obtained for dental resins containing silver NPs. In addition, dental resins incorporating SNP5-DexOxAmB are non-cytotoxic against human skin fibroblasts and human umbilical vein endothelial cells, and non-hemolytic against human red blood cells. SIGNIFICANCE: The incorporation of SNP5-DexOxAmB in dental resins resulted in a non-cytotoxic composite with high and durable antifungal activity.


Subject(s)
Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Nanoparticles , Resins, Synthetic , Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Biocompatible Materials , Candida albicans/drug effects , Cells, Cultured , Humans
8.
Biomaterials ; 34(21): 5281-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23578560

ABSTRACT

Although a variety of nanoparticles (NPs) functionalized with amphotericin B, an antifungal agent widely used in the clinic, have been studied in the last years their cytotoxicity profile remains elusive. Here we show that human endothelial cells take up high amounts of silica nanoparticles (SNPs) conjugated with amphotericin B (AmB) (SNP-AmB) (65.4 ± 12.4 pg of Si per cell) through macropinocytosis while human fibroblasts internalize relatively low amounts (2.3 ± 0.4 pg of Si per cell) because of their low capacity for macropinocytosis. We further show that concentrations of SNP-AmB and SNP up to 400 µg/mL do not substantially affect fibroblasts. In contrast, endothelial cells are sensitive to low concentrations of NPs (above 10 µg/mL), in particular to SNP-AmB. This is because of their capacity to internalize high concentration of NPs and high sensitivity of their membrane to the effects of AmB. Low-moderate concentrations of SNP-AmB (up to 100 µg/mL) induce the production of reactive oxygen species (ROS), LDH release, high expression of pro-inflammatory cytokines and chemokines (IL-8, IL-6, G-CSF, CCL4, IL-1ß and CSF2) and high expression of heat shock proteins (HSPs) at gene and protein levels. High concentrations of SNP-AmB (above 100 µg/mL) disturb membrane integrity and kill rapidly human cells (60% after 5 h). This effect is higher in SNP-AmB than in SNP.


Subject(s)
Amphotericin B/pharmacology , Endocytosis/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , HSP70 Heat-Shock Proteins/metabolism , Nanoparticles/chemistry , Amphotericin B/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/metabolism , Fibroblasts/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Inflammation Mediators/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Nanoparticles/ultrastructure , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Silicon Dioxide/pharmacology , Skin/cytology , Transcriptome/genetics
10.
Nanotechnology ; 22(49): 494002, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22101232

ABSTRACT

Nanoparticles (NPs) are very promising for the intracellular delivery of anticancer and immunomodulatory drugs, stem cell differentiation biomolecules and cell activity modulators. Although initial studies in the area of intracellular drug delivery have been performed in the delivery of DNA, there is an increasing interest in the use of other molecules to modulate cell activity. Herein, we review the latest advances in the intracellular-targeted delivery of short interference RNA, proteins and small molecules using NPs. In most cases, the drugs act at different cellular organelles and therefore the drug-containing NPs should be directed to precise locations within the cell. This will lead to the desired magnitude and duration of the drug effects. The spatial control in the intracellular delivery might open new avenues to modulate cell activity while avoiding side-effects.


Subject(s)
Drug Delivery Systems/methods , Intracellular Space/metabolism , Nanoparticles/chemistry , Pharmaceutical Preparations/administration & dosage , Proteins/administration & dosage , RNA, Small Interfering/administration & dosage , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Nanoparticles/analysis
11.
Nat Methods ; 8(9): 731-6, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21878920

ABSTRACT

The self-renewal and differentiation of human pluripotent stem cells (hPSCs) have typically been studied in flat, two-dimensional (2D) environments. In this Perspective, we argue that 3D model systems may be needed in addition, as they mimic the natural 3D tissue organization more closely. We survey methods that have used 3D biomaterials for expansion of undifferentiated hPSCs, directed differentiation of hPSCs and transplantation of differentiated hPSCs in vivo.


Subject(s)
Pluripotent Stem Cells/cytology , Animals , Biocompatible Materials/chemistry , Cell Culture Techniques/methods , Cell Differentiation , Extracellular Matrix/physiology , Humans , Pluripotent Stem Cells/physiology , Receptors, Cell Surface/physiology
12.
J Cardiovasc Transl Res ; 4(5): 616-30, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21735303

ABSTRACT

Recent pre-clinical and clinical studies indicate that certain exogenous stem cells and biomaterials can preserve cardiac tissue after myocardial infarction. Regarding stem cells, a growing body of data suggests that the short-term positive outcomes are mainly attributed to paracrine signaling mechanisms. The release of such factors is due to the cell's ability to sense cardiac environmentally derived cues, though the exact feedback loops are still poorly understood. However, given the limited engraftment and survival of transplanted cells in the ischemic environment, the long-term clinical benefits of these therapies have not yet been realized. To overcome this, the long-term controlled delivery of bioactive factors using biomaterials is a promising approach. A major challenge has been the ability to develop timely and spatially controlled gradients of different cues, pivotal for the development and regeneration of tissues. In addition, given the complexity of the remodeling process after myocardial infarction, multiple factors may be required at distinct disease stages to maximize therapeutic outcomes. Therefore, novel smart materials that can sense the surrounding environment and generate cues through on demand mechanisms will be of major importance in the translation of these promising advanced therapies. This article reviews how the cardiac environment can mediate the release profiles of bioactive cues from cells and biomaterials and how the controlled delivery impacts heart regeneration.


Subject(s)
Biocompatible Materials , Cellular Microenvironment , Myocardial Infarction/therapy , Myocardium/metabolism , Regeneration , Regenerative Medicine/methods , Stem Cell Transplantation , Tissue Engineering , Animals , Cell Communication , Delayed-Action Preparations , Drug Carriers , Humans , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Signal Transduction , Time Factors
13.
Biomaterials ; 32(4): 1102-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21035182

ABSTRACT

We present use of a synthetic, injectable matrix metalloproteinase (MMP)-responsive, bioactive hydrogel as an in situ forming scaffold to deliver thymosin ß4 (Tß4), a pro-angiogenic and pro-survival factor, along with vascular cells derived from human embryonic stem cells (hESC) in ischemic injuries to the heart in a rat model. The gel was found to substitute the degrading extracellular matrix in the infarcted myocardium of rats and to promote structural organization of native endothelial cells, while some of the delivered hESC-derived vascular cells formed de novo capillaries in the infarct zone. Magnetic resonance imaging (MRI) revealed that the microvascular grafts effectively preserved contractile performance 3 d and 6 wk after myocardial infarction, attenuated left ventricular dilation, and decreased infarct size as compared to infarcted rats treated with PBS injection as a control (3 d ejection fraction, + ∼7%, P < 0.001; 6 wk ejection faction, + ∼12%, P < 0.001). Elevation in vessel density was observed in response to treatment, which may be due in part to elevations in human (donor)-derived cytokines EGF, VEGF and HGF (1 d). Thus, a clinically relevant matrix for dual delivery of vascular cells and drugs may be useful in engineering sustained tissue preservation and potentially regenerating ischemic cardiac tissue.


Subject(s)
Embryonic Stem Cells/physiology , Myocardial Infarction/pathology , Myocardium/pathology , Tissue Engineering/methods , Transplants , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Cell Survival , Embryonic Stem Cells/cytology , Humans , Hydrogels/chemistry , Hydrogels/metabolism , Materials Testing , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Myocytes, Cardiac/pathology , Rats , Regeneration/physiology , Thymosin/administration & dosage , Thymosin/metabolism
14.
Biomacromolecules ; 11(10): 2810-7, 2010 Oct 11.
Article in English | MEDLINE | ID: mdl-20845938

ABSTRACT

Nosocomial fungal infections, an increasing healthcare concern worldwide, are often associated with medical devices. We have developed antifungal nanoparticle conjugates that can act in suspension or attach to a surface, efficiently killing fungi. For that purpose, we immobilized covalently amphotericin B (AmB), a potent antifungal agent approved by the FDA, widely used in clinical practice and effective against a large spectrum of fungi, into silica nanoparticles. These antifungal nanoparticle conjugates are fungicidal against several strains of Candida sp., mainly by contact. In addition, they can be reused up to 5 cycles without losing their activity. Our results show that the antifungal nanoparticle conjugates are more fungistatic and fungicidal than 10 nm colloidal silver. The antifungal activity of the antifungal nanoparticle conjugates is maintained when they are immobilized on a surface using a chemical adhesive formed by polydopamine. The antifungal nanocoatings have no hemolytic or cytotoxic effect against red blood cells and blood mononuclear cells, respectively. Surfaces coated with these antifungal nanoparticle conjugates can be very useful to render medical devices with antifungal properties.


Subject(s)
Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Candida/drug effects , Drug Carriers/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Amphotericin B/chemistry , Antifungal Agents/chemistry , Candida/growth & development , Equipment and Supplies/microbiology , Microbial Sensitivity Tests , Microscopy, Electron, Transmission , Particle Size , Surface Properties
15.
Nat Protoc ; 5(6): 1115-26, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20539287

ABSTRACT

Herein, we describe a protocol for the isolation of human embryonic stem cells (hESCs)-derived vascular cells at various stages of development. The cells are isolated from 10 to 15-d-old human embryoid bodies (EBs) cultured in suspension. After dissociation, cells are labeled with anti-CD34 or anti-CD31 (PECAM1) antibody and separated from the cell mixture by magnetic-activated cell separation (MACS) or fluorescent-activated cell sorting (FACS). Isolated vascular cells are then cultured in media conditions that support specific differentiation and expansion pathways. The resulting vascular cell populations contain >80% endothelial-like or smooth muscle-like cells. Assuming typical initial cell adhesion and proliferation rates, the entire procedure can be completed within 1.5 months. Vascular cells isolated and differentiated under the described conditions may constitute a potential cell source for therapeutic application toward repair of ischemic tissues, preparation of tissue-engineered vascular grafts and design of cellular kits for drug screening applications.


Subject(s)
Cell Separation/methods , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Myocytes, Smooth Muscle/cytology , Animals , Base Sequence , Cell Differentiation , Cells, Cultured , DNA Primers/genetics , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Flow Cytometry , Humans , Mice , Myocytes, Smooth Muscle/metabolism , Polymerase Chain Reaction , Tissue Engineering
16.
Methods Mol Biol ; 584: 333-54, 2010.
Article in English | MEDLINE | ID: mdl-19907986

ABSTRACT

The vascularization of tissue constructs remains a major challenge in regenerative medicine, as the diffusional supply of oxygen can support only 100-200 mum thick layers of viable tissue. The formation of a mature and functional vascular network requires communication between endothelial cells (ECs) and smooth muscle cells (SMCs). Potential sources of these cells that involve noninvasive methodologies are required for numerous applications including tissue-engineered vascular grafts, myocardial ischemia, wound healing, plastic surgery, and general tissue-engineering applications. Human embryonic stem cells (hESCs) can be an unlimited source of these cells. They can be expanded in vitro in an undifferentiated state without apparent limit, and hES-derived cells can be created in virtually unlimited amounts for potential clinical uses. Recently, vascular progenitor cells as well as endothelial and smooth muscle cells have been isolated from hESCs.


Subject(s)
Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Myocytes, Smooth Muscle/cytology , Tissue Scaffolds , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cell Separation , Cell Survival , Coculture Techniques , Dextrans , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Flow Cytometry , Humans , Hyaluronic Acid , Hydrogels , Mice , Microscopy, Electron, Scanning , Myocytes, Smooth Muscle/metabolism , Tissue Engineering
17.
Biomaterials ; 30(26): 4318-24, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19500842

ABSTRACT

The in vitro potential of a synthetic matrix metalloproteinase (MMP)-responsive poly(ethylene glycol) (PEG)-based hydrogel as a bioactive co-encapsulation system for vascular cells and a small bioactive peptide, thymosin beta4 (Tbeta4), was examined. We show that the physical incorporation of Tbeta4 in this bioactive matrix creates a three-dimensional (3D) environment conducive for human umbilical vein endothelial cell (HUVEC) adhesion, survival, migration and organization. Gels with entrapped Tbeta4 increased the survival of HUVEC compared to gels without Tbeta4, and significantly up-regulated the endothelial genes vascular endothelial-cadherin and angiopoietin-2, whereas von Willebrand factor was significantly down-regulated. Incorporation of Tbeta4 significantly increased MMP-2 and MMP-9 secretion of encapsulated HUVEC. The gel acts as a controlled Tbeta4-release system, as MMP-2 and MMP-9 enzymes trigger the release. In addition, Tbeta4 facilitated HUVEC attachment and induced vascular-like network formation upon the PEG-hydrogels. These MMP-responsive PEG-hydrogels may thus serve as controlled co-encapsulation system of vascular cells and bioactive factors for in situ regeneration of ischemic tissues.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Gene Expression Regulation/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Polyethylene Glycols/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Thymosin/pharmacology , Umbilical Veins/cytology
18.
Biomaterials ; 29(10): 1526-32, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18096220

ABSTRACT

Highly fluorescent core-shell silica nanoparticles made by the modified Stöber process (C dots) are promising as tools for sensing and imaging subcellular agents and structures but will only be useful if they can be easily delivered to the cytoplasm of the subject cells. This work shows that C dots can be electrostatically coated with cationic polymers, changing their surface charge and enabling them to escape from endosomes and enter the cytoplasm and nucleus. As an example of cellular delivery, we demonstrate that these particles can also be complexed with DNA and mediate and trace DNA delivery and gene expression.


Subject(s)
Nanoparticles/chemistry , Silicon Dioxide/chemistry , Animals , COS Cells , Cell Nucleus/chemistry , Cell Proliferation , Cell Survival , Chlorocebus aethiops , Cytoplasm/chemistry , DNA/chemistry , Drug Delivery Systems/methods , Flow Cytometry , Fluorescence , HeLa Cells , Humans , Microscopy, Confocal , Microscopy, Electron, Scanning , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure
19.
Circ Res ; 101(3): 286-94, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17569886

ABSTRACT

We report that human embryonic stem cells contain a population of vascular progenitor cells that have the ability to differentiate into endothelial-like and smooth muscle (SM)-like cells. Vascular progenitor cells were isolated from EBs grown in suspension for 10 days and were characterized by expression of the endothelial/hematopoietic marker CD34 (CD34+ cells). When these cells are subsequently cultured in EGM-2 (endothelial growth medium) supplemented with vascular endothelial growth factor-165 (50 ng/mL), they give rise to endothelial-like cells characterized by a cobblestone cell morphology, expression of endothelial markers (platelet endothelial cell-adhesion molecule-1, CD34, KDR/Flk-1, vascular endothelial cadherin, von Willebrand factor), incorporation of acetylated low-density lipoprotein, and formation of capillary-like structures when placed in Matrigel. In contrast, when CD34+ cells are cultured in EGM-2 supplemented with platelet-derived growth factor-BB (50 ng/mL), they give rise to SM-like cells characterized by spindle-shape morphology, expression of SM cell markers (alpha-SM actin, SM myosin heavy chain, calponin, caldesmon, SM alpha-22), and the ability to contract and relax in response to common pharmacological agents such as carbachol and atropine but rarely form capillary-like structures when placed in Matrigel. Implantation studies in nude mice show that both cell types contribute to the formation of human microvasculature. Some microvessels contained mouse blood cells, which indicates functional integration with host vasculature. Therefore, the vascular progenitors isolated from human embryonic stem cells using methods established in the present study could provide a means to examine the mechanisms of endothelial and SM cell development, and they could also provide a potential source of cells for vascular tissue engineering.


Subject(s)
Blood Vessels/cytology , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Myocytes, Smooth Muscle/cytology , Animals , Antigens, CD34/biosynthesis , Becaplermin , Biomarkers , Blood Vessels/ultrastructure , Cattle , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/transplantation , Collagen , Culture Media/pharmacology , Drug Combinations , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Fetal Blood , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Injections, Subcutaneous , Laminin , Mice , Mice, Nude , Morphogenesis , Muscle Proteins/biosynthesis , Neovascularization, Physiologic , Organ Specificity , Platelet-Derived Growth Factor/pharmacology , Proteoglycans , Proto-Oncogene Proteins c-sis , Tissue Engineering , Vascular Endothelial Growth Factor A/pharmacology
20.
Proc Natl Acad Sci U S A ; 104(27): 11298-303, 2007 Jul 03.
Article in English | MEDLINE | ID: mdl-17581871

ABSTRACT

Control of self-renewal and differentiation of human ES cells (hESCs) remains a challenge. This is largely due to the use of culture systems that involve poorly defined animal products and do not mimic the normal developmental milieu. Routine protocols involve the propagation of hESCs on mouse fibroblast or human feeder layers, enzymatic cell removal, and spontaneous differentiation in cultures of embryoid bodies, and each of these steps involves significant variability of culture conditions. We report that a completely synthetic hydrogel matrix can support (i) long-term self-renewal of hESCs in the presence of conditioned medium from mouse embryonic fibroblast feeder layers, and (ii) direct cell differentiation. Hyaluronic acid (HA) hydrogels were selected because of the role of HA in early development and feeder layer cultures of hESCs and the controllability of hydrogel architecture, mechanics, and degradation. When encapsulated in 3D HA hydrogels (but not within other hydrogels or in monolayer cultures on HA), hESCs maintained their undifferentiated state, preserved their normal karyotype, and maintained their full differentiation capacity as indicated by embryoid body formation. Differentiation could be induced within the same hydrogel by simply altering soluble factors. We therefore propose that HA hydrogels, with their developmentally relevant composition and tunable physical properties, provide a unique microenvironment for the self-renewal and differentiation of hESCs.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Hyaluronic Acid/physiology , Hydrogels/pharmacology , Animals , Cell Differentiation/physiology , Cell Line , Embryonic Stem Cells/physiology , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...