Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Biofabrication ; 16(4)2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012007

ABSTRACT

Bone grafting is the most common treatment for repairing bone defects. However, current bone grafting methods have several drawbacks. Bone tissue engineering emerges as a promising solution to these problems. An ideal engineered bone graft should exhibit high mechanical strength, osteogenic properties, and pre-vascularization. Both top-down (using bulk scaffold) and bottom-up (using granular modules) approaches face challenges in fulfilling these requirements. In this paper, we propose a novel sectional modular bone approach to construct osteogenic, pre-vascularized bone grafts in anatomical shapes. We 3D-printed a series of rigid, thin, sectional, porous scaffolds from a biodegradable polymer, tailored to the dimensions of a femur bone shaft. These thin sectional modules promote efficient nutrition and waste removal due to a shorter diffusion distance. The modules were pre-vascularized viain-situangiogenesis, achieved through endothelial cell sprouting from the scaffold struts. Angiogenesis was further enhanced through co-culture with bioprinted fibroblast microtissues, which secreted pre-angiogenic growth factors. Sectional modules were assembled around a porous rod incorporated with Bone Morphogenetic Protein-2 (BMP-2), which released over 3 weeks, demonstrating sustained osteogenic activity. The assembled scaffold, in the anatomical shape of a human femur shaft, was pre-vascularized, osteogenic, and possessed high mechanical strength, supporting 12 times the average body weight. The feasibility of implanting the assembled bone graft was demonstrated using a 3D-printed femur bone defect model. Our method provides a novel modular engineering approach for regenerating tissues that require high mechanical strength and vascularization.


Subject(s)
Bioprinting , Bone Morphogenetic Protein 2 , Bone Transplantation , Neovascularization, Physiologic , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Bone Morphogenetic Protein 2/pharmacology , Neovascularization, Physiologic/drug effects , Humans , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Animals , Femur/blood supply , Delayed-Action Preparations/chemistry , Osteogenesis/drug effects , Bone and Bones/blood supply , Human Umbilical Vein Endothelial Cells , Angiogenesis
2.
Small ; : e2308694, 2024 May 19.
Article in English | MEDLINE | ID: mdl-38763898

ABSTRACT

Few studies have proved that bioprinting itself helps recapitulate native tissue functions mainly because the bioprinted macro shape can rarely, if ever, influence cell function. This can be more problematic in bioprinting cartilage, generally considered more challenging to engineer. Here a new method is shown to micro-pattern chondrocytes within bioprinted sub-millimeter micro tissues, denoted as patterned micro-articular-cartilages tissues (PA-MCTs). Under the sole influence of bioprinted cellular patterns. A pattern scoring system is developed after over 600 bioprinted cellular patterns are analyzed. The top-scored pattern mimics that of the isogenous group in native articular cartilage. Under the sole influence of this pattern during PA-MCTs bio-assembling into macro-cartilage and repairing cartilage defects, chondrogenic cell phenotype is preserved, and cartilagenesis is initiated and maintained. Neocartilage tissues from individual and assembled PA-MCTs are comparable to native articular cartilage and superior to cartilage bioprinted with homogeneously distributed cells in morphology, biochemical components, cartilage-specific protein and gene expression, mechanical properties, integration with host tissues, zonation forming and stem cell chondrogenesis. PA-MCTs can also be used as osteoarthritic and healthy cartilage models for therapeutic drug screening and cartilage development studies. This cellular patterning technique can pave a new way for bioprinting to recapitulate native tissue functions via tissue genesis.

3.
Cells ; 11(20)2022 10 16.
Article in English | MEDLINE | ID: mdl-36291114

ABSTRACT

Articular cartilage lesions are prevalent and affect one out of seven American adults and many young patients. Cartilage is not capable of regeneration on its own. Existing therapeutic approaches for articular cartilage lesions have limitations. Cartilage tissue engineering is a promising approach for regenerating articular neocartilage. Bioassembly is an emerging technology that uses microtissues or micro-precursor tissues as building blocks to construct a macro-tissue. We summarize and highlight the application of bioassembly technology in regenerating articular cartilage. We discuss the advantages of bioassembly and present two types of building blocks: multiple cellular scaffold-free spheroids and cell-laden polymer or hydrogel microspheres. We present techniques for generating building blocks and bioassembly methods, including bioprinting and non-bioprinting techniques. Using a data set of 5069 articles from the last 28 years of literature, we analyzed seven categories of related research, and the year trends are presented. The limitations and future directions of this technology are also discussed.


Subject(s)
Bioprinting , Cartilage, Articular , Humans , Bioprinting/methods , Tissue Engineering/methods , Hydrogels , Polymers
4.
Biomed Mater ; 15(5): 055028, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32485682

ABSTRACT

Collective cell migration refers to the movement of groups of cells and collective cell behavior and relies on cell-cell communication and cell-environment interactions. Collective cell migration plays a fundamental role in many aspects of cell biology and pathology. Current protocols for studying collective cell migration either use destructive methods or are not convenient for liquid handling. Here we present a novel 3D-printed insert-array and a 3D-coculture-array for collective cell migration study in high-throughput. The fabricated insert-array is comprised of 96 cylinder shaped inserts which can be placed in each well of a 96-well plate generating watertight contact with the bottom of each well. The insert-array has high manufacturing tolerance, and the coefficient of variations of the insert diameter and circularity are 0.67% and 0.03%, respectively. Each insert generates a circular cell-free area within the well without cell damage and provides convenient access for both manual and robotic liquid handling. Using the 3D-printed insert-array, we studied the migration of human umbilical vein endothelial cells (HUVECs) under the molecular influences of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) and under the cellular influences of human mesenchymal stem cells (hMSCs) using the 3D-coculture-array. Our results show that the migration of HUVECs was dose-dependent on the VEGF and bFGF with different correlation patterns. They also generated a synergic pro-migration effect. When cocultured with hMSCs, the migration rate increased significantly while dependent on the number of hMSCs. The effects were partially blocked by VEGF inhibitor which suggests that VEGF secreted from hMSCs plays an important role in cell-to-cell communication during cell migration. The 3D-coculture-array can be manufactured at very low cost and shows higher biomolecule transport efficiency than the commercially available transwell. The calculated Z-factor is 0.66, which classifies our system as a perfect high-throughput assay. In summary, our newly developed insert-array and 3D-coculture-array provide a versatile platform to study collective cell migration in high-throughput as well as the molecular and cellular influences upon it.


Subject(s)
Coculture Techniques/methods , Printing, Three-Dimensional , Cell Communication , Cell Movement , Cell-Free System , Cells, Cultured , Fibroblast Growth Factor 2/metabolism , High-Throughput Screening Assays/methods , Human Umbilical Vein Endothelial Cells , Humans , Materials Testing , Mesenchymal Stem Cells/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
5.
Int J Mol Sci ; 21(10)2020 May 14.
Article in English | MEDLINE | ID: mdl-32423161

ABSTRACT

Drop-on-demand (DOD) 3D bioprinting technologies currently hold the greatest promise for generating functional tissues for clinical use and for drug development. However, existing DOD 3D bioprinting technologies have three main limitations: (1) droplet volume inconsistency; (2) the ability to print only bioinks with low cell concentrations and low viscosity; and (3) problems with cell viability when dispensed under high pressure. We report our success developing a novel direct-volumetric DOD (DVDOD) 3D bioprinting technology that overcomes each of these limitations. DVDOD can produce droplets of bioink from < 10 nL in volume using a direct-volumetric mechanism with < ± 5% volumetric percent accuracy in an accurate spatially controlled manner. DVDOD has the capability of dispensing bioinks with high concentrations of cells and/or high viscosity biomaterials in either low- or high-throughput modes. The cells are subjected to a low pressure during the bioprinting process for a very short period of time that does not negatively impact cell viability. We demonstrated the functions of the bioprinter in two distinct manners: (1) by using a high-throughput drug-delivery model; and (2) by bioprinting micro-tissues using a variety of different cell types, including functional micro-tissues of bone, cancer, and induced pluripotent stem cells. Our DVDOD technology demonstrates a promising platform for generating many types of tissues and drug-delivery models.


Subject(s)
Biocompatible Materials/pharmacology , Bioprinting , Printing, Three-Dimensional , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Cell Survival/drug effects , Drug Delivery Systems , Extracellular Matrix/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Pharmaceutical Preparations , Tissue Engineering/trends
6.
Int J Mol Sci ; 21(1)2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31906530

ABSTRACT

Fused deposit modeling (FDM) 3D printing technology cannot generate scaffolds with high porosity while maintaining good integrity, anatomical-surface detail, or high surface area-to-volume ratio (S/V). Solvent casting and particulate leaching (SCPL) technique generates scaffolds with high porosity and high S/V. However, it is challenging to generate complex-shaped scaffolds; and solvent, particle and residual water removal are time consuming. Here we report techniques surmounting these problems, successfully generating a highly porous scaffold with the anatomical-shape characteristics of a human femur by polylactic acid polymer (PLA) and PLA-hydroxyapatite (HA) casting and salt leaching. The mold is water soluble and is easily removable. By perfusing with ethanol, water, and dry air sequentially, the solvent, salt, and residual water were removed 20 fold faster than utilizing conventional methods. The porosities are uniform throughout the femoral shaped scaffold generated with PLA or PLA-HA. Both scaffolds demonstrated good biocompatibility with the pre-osteoblasts (MC3T3-E1) fully attaching to the scaffold within 8 h. The cells demonstrated high viability and proliferation throughout the entire time course. The HA-incorporated scaffolds demonstrated significantly higher compressive strength, modulus and osteoinductivity as evidenced by higher levels of alkaline-phosphatase activity and calcium deposition. When 3D printing a 3D model at 95% porosity or above, our technology preserves integrity and surface detail when compared with FDM-generated scaffolds. Our technology can also generate scaffolds with a 31 fold larger S/V than FDM. We have developed a technology that is a versatile tool in creating personalized, patient-specific bone graft scaffolds efficiently with high porosity, good scaffold integrity, high anatomical-shaped surface detail and large S/V.


Subject(s)
Biocompatible Materials/chemistry , Osteoblasts/chemistry , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biocompatible Materials/chemical synthesis , Calcium/analysis , Cell Differentiation , Cell Proliferation , Cell Survival , Compressive Strength , Durapatite/chemistry , Femur , Humans , Materials Testing , Osteoblasts/enzymology , Osteoblasts/metabolism , Perfusion , Polyesters/chemistry , Porosity , Tissue Scaffolds/adverse effects
7.
Insects ; 10(4)2019 Apr 25.
Article in English | MEDLINE | ID: mdl-31027217

ABSTRACT

(E)-ß-Ocimene was the only volatile chemical found to be emitted by whole, live worker larvae of Apis mellifera L. when sampling in the vapor phase. In addition to (E)-ß-ocimene, there is evidence for the existence of other volatiles, but the changes in their composition and contents remain unknown during larval development, as are their differences from larvae to larval food. We investigated volatile components of worker larvae and larval food using solid phase dynamic extraction (SPDE) coupled with gas chromatography-mass spectrometry (GC-MS). Nine compounds were identified with certainty and six tentatively, including terpenoids, aldehydes, hydrocarbons, an ester and a ketone. The contents of volatiles in the second-instar worker larvae differ greatly from those in larvae of other stages. This is mainly attributable to terpenoids, which resulted in the second-instar worker larvae having significantly higher amounts of overall volatiles. Larval food contained significantly higher amounts of aldehydes and hydrocarbons than the corresponding larvae from the fourth to fifth-instar. We discovered volatiles in worker larvae and their food that were never reported before; we also determined the content changes of these volatiles during larval development.

8.
Tissue Eng Part C Methods ; 24(4): 205-213, 2018 04.
Article in English | MEDLINE | ID: mdl-29397786

ABSTRACT

Collective cell migration, in which cells migrate as a group, is fundamental in many biological and pathological processes. There is increasing interest in studying the collective cell migration in high throughput. Cell scratching, insertion blocker, and gel-dissolving techniques are some methodologies used previously. However, these methods have the drawbacks of cell damage, substrate surface alteration, limitation in medium exchange, and solvent interference. The superhydrophobic surface, on which the water contact angle is greater than 150 degrees, has been recently utilized to generate patterned arrays. Independent cell culture areas can be generated on a substrate that functions the same as a conventional multiple well plate. However, so far there has been no report on superhydrophobic patterning for the study of cell migration. In this study, we report on the successful development of a robotically patterned superhydrophobic array for studying collective cell migration in high throughput. The array was developed on a rectangular single-well cell culture plate consisting of hydrophilic flat microwells separated by the superhydrophobic surface. The manufacturing process is robotic and includes patterning discrete protective masks to the substrate using 3D printing, robotic spray coating of silica nanoparticles, robotic mask removal, robotic mini silicone blocker patterning, automatic cell seeding, and liquid handling. Compared with a standard 96-well plate, our system increases the throughput by 2.25-fold and generates a cell-free area in each well non-destructively. Our system also demonstrates higher efficiency than conventional way of liquid handling using microwell plates, and shorter processing time than manual operating in migration assays. The superhydrophobic surface had no negative impact on cell viability. Using our system, we studied the collective migration of human umbilical vein endothelial cells and cancer cells using assays of endpoint quantification, dynamic cell tracking, and migration quantification following varied drug treatments. This system provides a versatile platform to study collective cell migration in high throughput for a broad range of applications.


Subject(s)
Bone Neoplasms/pathology , Cell Movement , Cell Tracking/methods , Human Umbilical Vein Endothelial Cells/physiology , Nanotechnology/instrumentation , Osteosarcoma/pathology , Robotics/instrumentation , Cell Survival , Cells, Cultured , Human Umbilical Vein Endothelial Cells/cytology , Humans , Hydrophobic and Hydrophilic Interactions , Printing, Three-Dimensional , Robotics/methods , Silicon/chemistry
9.
Tissue Eng Part C Methods ; 21(10): 1025-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25962617

ABSTRACT

Vascularization is a key challenge in tissue engineering. Three-dimensional structure and microcirculation are two fundamental parameters for evaluating vascularization. Microscopic techniques with cellular level resolution, fast continuous observation, and robust 3D postimage processing are essential for evaluation, but have not been applied previously because of technical difficulties. In this study, we report novel video-rate confocal microscopy and 3D postimage processing techniques to accomplish this goal. In an immune-deficient mouse model, vascularized bone tissue was successfully engineered using human bone marrow mesenchymal stem cells (hMSCs) and human umbilical vein endothelial cells (HUVECs) in a poly (D,L-lactide-co-glycolide) (PLGA) scaffold. Video-rate (30 FPS) intravital confocal microscopy was applied in vitro and in vivo to visualize the vascular structure in the engineered bone and the microcirculation of the blood cells. Postimage processing was applied to perform 3D image reconstruction, by analyzing microvascular networks and calculating blood cell viscosity. The 3D volume reconstructed images show that the hMSCs served as pericytes stabilizing the microvascular network formed by HUVECs. Using orthogonal imaging reconstruction and transparency adjustment, both the vessel structure and blood cells within the vessel lumen were visualized. Network length, network intersections, and intersection densities were successfully computed using our custom-developed software. Viscosity analysis of the blood cells provided functional evaluation of the microcirculation. These results show that by 8 weeks, the blood vessels in peripheral areas function quite similarly to the host vessels. However, the viscosity drops about fourfold where it is only 0.8 mm away from the host. In summary, we developed novel techniques combining intravital microscopy and 3D image processing to analyze the vascularization in engineered bone. These techniques have broad applicability for evaluating vascularization in other engineered tissues as well.


Subject(s)
Bone and Bones/blood supply , Human Umbilical Vein Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Osteogenesis , Tissue Engineering , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, SCID
10.
Mol Biol Rep ; 41(6): 3569-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24515387

ABSTRACT

Immunological stress is the status of animal in active immune when they are challenged by bacterial, virus and endocrine. It is associated with immunological, neurological, and endocrinological response. An immunological stress model was established in this study using Chinese indigenous breed (Laiwu), crossbred (Lulai), and exotic breed (Yorkshire), to explore the capacity of immunological stress resistance among different breeds. The study was also to reveal the effect of chromium yeast to immunological stress. 48 post-weaning piglets were taken from three breeds, 16 piglets of each breed from Laiwu, Lulai and Yorkshire. The experiment was designed as 2 × 2 factors, immunological stress (Saline, LPS) and Chromium (with Cr, without Cr). There were four treatments: control, LPS, Cr, and Cr+LPS. Blood parameters related to immunological stress, such as IL-1ß, TNF-α, GH, and cortisol, were examined after blood sample were taken at 0, 2, 5, and 7 h of post-injection. The results showed that IL-1ß, TNF-α, and cortisol increased in group of LPS treatment while GH declined at 2 h of post-injection in comparison to the control (p < 0.01). However, IL-1ß, TNF-α, and cortisol in group of Cr+LPS were lower than that in group of LPS while GH were higher (p < 0.05). Total RNA was extractedfrom blood lymphocytes separation samples at 2 h of post-injection. Q-PCR was applied to determine the gene expression of IL-1ß, IL-6 and TNF-α. The results showed that LPS injection increased the gene expression of IL-1ß, IL-6 and TNF-α. Among three breeds, the expression of IL-1ß, IL-6 and TNF-α in Yorkshire were significantly higher than in Laiwu and Lulai (p < 0.05), but there was no difference between Laiwu and Lulai. Among four treatments, the expression of three genes in group of LPS was the highest, compared to the group of Cr+LPS (p < 0.05) and control (p < 0.01). This study concluded that Laiwu had stronger capacity of immunological stress resistance and next was Lulai among three breeds. Chromium yeast helped piglets relieve immunological stress.


Subject(s)
Gene Expression Regulation/drug effects , Immunization , Lipopolysaccharides/toxicity , Stress, Physiological/immunology , Animals , Breeding , Chromium/toxicity , Gene Expression Regulation/immunology , Hydrocortisone/blood , Hydrocortisone/genetics , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/blood , Interleukin-6/genetics , Neurosecretory Systems/drug effects , Neurosecretory Systems/immunology , Stress, Physiological/genetics , Swine , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
11.
Microvasc Res ; 90: 40-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23886898

ABSTRACT

These studies aimed to determine the effect of smooth muscle cells (SMCs) on angiogenic behavior of endothelial cells (ECs) within fibrin hydrogels, an extracellular matrix (ECM) commonly used in tissue engineering. We developed a 3-D, fibrin-based co-culture assay of angiogenesis consisting of aggregates of SMCs with ECs seeded onto the aggregates' surface. Using digital fluorescence micrography, EC matrix invasion was quantified by average length of sprouts (ALS) and density of sprout formation (DSF). We demonstrated that ECs and SMCs co-invade into the ECM in close proximity to one another. ECs that were co-cultured with SMCs demonstrated increased invasion compared to ECs that were cultured alone at all time points. At Day 19, the ALS of ECs in co-culture was 327+/-58µm versus 70+/-11µm of ECs cultured alone (p=.01). The DSF of co-cultured ECs was also significantly greater than that of ECs cultured alone (p=.007 on Day 19). This appeared to be a function of both increased EC invasion as well as improved persistence of EC sprout networks. At 7days, ECs in co-culture with proliferation-inhibited SMCs previously treated with Mitomycin-C (MMC) demonstrated significantly attenuated sprouting compared to ECs co-cultured with SMCs that were untreated with MMC (82+/-14µm versus 205+/-32µm; p<.05). In assays in which multiple co-culture aggregates were cultured within a single hydrogel, we observed directional invasion of sprouts preferentially towards the other aggregates within the hydrogel. In co-culture assays without early EC/SMC contact, the ALS of ECs cultured in the presence of SMCs was significantly greater than those cultured in the absence of SMCs by Day 3 (320+/-21µm versus 187+/-16µm; p<.005). We conclude that SMCs augment EC matrix invasion into 3-D fibrin hydrogels, at least in part resulting from SMC proliferative and invasive activities. Directed invasion between co-culture aggregates and augmented angiogenesis in the absence of early contact suggests a paracrine mechanism for the observed results.


Subject(s)
Cell Movement , Cell Proliferation , Endothelial Cells/metabolism , Fibrin/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Paracrine Communication , Animals , Cell Shape , Cells, Cultured , Coculture Techniques , Dogs , Hydrogels , Microscopy, Fluorescence , Time Factors
12.
Biomaterials ; 32(15): 3776-83, 2011 May.
Article in English | MEDLINE | ID: mdl-21388676

ABSTRACT

We developed a live imaging system enabling dynamic visualization of single cell alignment induced by external mechanical force in a 3-D collagen matrix. The alignment dynamics and migration of smooth muscle cells (SMCs) were studied by time lapse differential interference contrast and/or phase contrast microscopy. Fluorescent and reflection confocal microcopy were used to study the SMC morphology and the microscale collagen matrix remodeling induced by SMCs. A custom developed program was used to quantify the cell migration and matrix remodeling. Our system enables cell concentration-independent alignment eliminating cell-to-cell interference and enables dynamic cell tracking, high magnification observation and rapid cell alignment accomplished in a few hours compared to days in traditional models. We observed that cells sense and response to the mechanical signal before cell spreading. Under mechanical stretch the migration directionality index of SMCs is 46.3% more than those cells without external stretch; the dynamic direction of cell protrusion is aligned to that of the mechanical force; SMCs showed directional matrix remodeling and the alignment index calculated from the matrix in front of cell protrusions is about 3 fold of that adjacent to cell bodies. Our results indicate that the mechanism of cell alignment is directional cell protrusion. Mechano-sensing, directionality in cell protrusion dynamics, cell migration and matrix remodeling are highly integrated. Our system provides a platform for studying the role of mechanical force on the cell matrix interactions and thus finds strategies to optimize selected properties of engineered tissues.


Subject(s)
Cell Movement , Collagen/chemistry , Hydrogels/chemistry , Microscopy, Confocal/instrumentation , Myocytes, Smooth Muscle/cytology , Tissue Scaffolds/chemistry , Animals , Carotid Arteries/cytology , Cattle , Cell Survival , Cells, Cultured , Dogs , Equipment Design , Extracellular Matrix/chemistry , Mechanical Phenomena , Single-Cell Analysis/instrumentation
13.
J Tissue Eng Regen Med ; 5(5): 375-83, 2011 May.
Article in English | MEDLINE | ID: mdl-20718050

ABSTRACT

The development of a functional microvasculature is critical to the long-term survival of implanted tissue-engineered constructs. Dynamic culture conditions have been shown to significantly modulate phenotypic characteristics and stimulate proliferation of cells within hydrogel-based tissue engineered blood vessels. Although prior work has described the effects uniaxial or equibiaxial mechanical stimulation has on endothelial cells, no work has outlined effects of three-dimensional mechanical stimulation on endothelial cells within tubular vessel analogues. We demonstrate here that 7 days of 10% cyclic volumetric distension has a deleterious effect on the average length and density of angiogenic sprouts derived from pellets of bovine aortic endothelial cells. Although both groups demonstrated lumen formation, the sprouts grown under dynamic culture conditions typically had wider, less-branching sprout patterns. These results suggest that prolonged mechanical stimulation could represent a cue for angiogenic sprouts to preferentially develop larger lumens over cellular migration and subsequent sprout length.


Subject(s)
Aorta/cytology , Endothelial Cells/cytology , Fibrin/chemistry , Hydrogels/chemistry , Neovascularization, Pathologic , Tissue Engineering/methods , Vasa Vasorum/metabolism , Animals , Bioreactors , Cattle , Fibronectins/chemistry , Microscopy, Confocal/methods , Microscopy, Electron, Transmission/methods , Neovascularization, Physiologic , Stress, Mechanical
14.
J Investig Med ; 58(7): 845-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20683346

ABSTRACT

Vascular tissue engineering should provide more biocompatible and functional conduits than synthetic vascular grafts. Understanding cell-scaffold interactions and developing an efficient delivery system for growth factors and other biomolecules to control the signaling between the cells and the scaffold are fundamental issues in a wide range of tissue engineering research fields. Type 1 collagen is a natural scaffold extensively used in vascular tissue engineering and is a widely used vehicle in biomolecule delivery. In this article, we will discuss type 1 collagen as a vascular tissue engineering scaffold, describe strategies for elucidating the interaction between cells and type 1 collagen scaffolds using various imaging techniques, and summarize our work on the development of a chimeric collagen-binding growth factor-based local delivery system.


Subject(s)
Blood Vessel Prosthesis , Collagen Type I/metabolism , Drug Delivery Systems/methods , Intercellular Signaling Peptides and Proteins/pharmacokinetics , Tissue Scaffolds , Animals , Humans , Protein Binding , Recombinant Fusion Proteins
15.
Biomaterials ; 31(5): 878-85, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19853908

ABSTRACT

We investigated the delivery of R136K-CBD (a collagen-binding mutant chimera of fibroblast growth factor-1) with a type I collagen scaffold as the delivery vehicle to smooth muscle cells (SMCs) for vascular tissue engineering. The binding affinity of R136K-CBD to 3-D collagen scaffolds was investigated both in the presence and absence of cells and/or salts. 2-D and 3-D visualization of delivery of R136K-CBD into SMCs were accomplished by combined fluorescent and reflection confocal microscopy. The mitogenic effect of collagen-immobilized R136K-CBD on SMCs in 3-D collagen was studied by Cyquant assay at different time intervals. In the group devoid of salt and cells, no detectable release of R136K-CBD into overlying culture media was found, compared with burst-and-continuous release of R136K and FGF-1 over a 14-day period in all other groups. The release rate of R136K-CBD was 1.7 and 1.6-fold less than R-136K and FGF-1 when media was supplemented with 2m salt (P<0.0001), and 2.6 and 2.5-fold less in cell-populated collagen hydrogels (P<0.0001), respectively. R136K-CBD showed essentially uniform binding to collagen and its distribution was dependent on that of the collagen scaffold. Internalization of R136K-CBD into SMCs was documented by confocal microscopy. 3-D local delivery of collagen-immobilized R136K-CBD increased the proliferation of SMCs in the collagen matrix to significantly greater levels and for a significantly greater duration than R136K or FGF-1, with 2.0 and 2.1-fold more mitogenicity than R136K and FGF-1 respectively (P<0.0001) at day 7. The results suggest that our collagen-binding fusion protein is an effective strategy for growth factor delivery for vascular tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Collagen/chemistry , Drug Carriers/chemistry , Fibroblast Growth Factor 1/administration & dosage , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Biomimetic Materials/chemistry , Blood Vessels/growth & development , Cell Culture Techniques/methods , Cells, Cultured , Crystallization/methods , Dogs , Fibroblast Growth Factor 1/chemistry , Materials Testing , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Particle Size , Surface Properties , Tissue Engineering/methods
16.
Biomaterials ; 30(11): 2023-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19147225

ABSTRACT

Smooth muscle cells (SMCs) and collagen scaffolds are widely used in vascular tissue engineering but their interactions in remodeling at the microscale level remained unclear. We characterized microscale morphologic alterations of collagen remodeled by SMCs in six dimensions: three spatial, time, multichannel and multi-position dimensions. In live imaging assays, computer-assisted cell tracking showed locomotion characteristics of SMCs; reflection and fluorescent confocal microscopy and spatial reconstruction images of each time point showed detailed morphologic changes of collagen fibers and spatial collagen-SMC interactions. The density of the collagen around the SMCs was changed dynamically by the leading edges of the cells. The density of the collagen following 24h of cell-induced remodeling increased 51.61+/-9.73% compared to unremodeled collagen containing cells for 1h (P<0.0001, n=40) (NS vs. collagen without cells). Fast Fourier transform analysis showed that the collagen fibers' orientation changed from random (alignment index=0.047+/-0.029, n=40) after 1h into concordant with that of the SMCs (alignment index=0.379+/-0.098, P<0.0001, n=40) after 24h. Mosaic imaging extended the visual field from a single cell to a group of cells in one image without loss of optical resolution. Direct visualization of alignment of actin fibers and collagen fibers showed the molecular machinery of the process of scaffold remodeling. This is a new approach to better understanding the mechanism of scaffold remodeling and our techniques represent effective tools to investigate the interactions between cells and scaffold in detail at the microscale level.


Subject(s)
Collagen/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Tissue Engineering/methods , Animals , Cells, Cultured , Dogs , Microscopy, Confocal
17.
Article in Chinese | MEDLINE | ID: mdl-16108350

ABSTRACT

OBJECTIVE: To investigate the possibility of establishing the human bone marrow mesenchymal stem cells (hMSCs) bank as to provide an alternative source for the seed cells of tissue engineering. METHODS: The cell surface antigens of the purified, expanded hMSCs and the ones following cryopreservation were detected by flow cytometry, cultured in a special medium to induce the osteogenic and chondrocytic differentiation. Morphology was studied by light and electronic microscopes. The detection of alkaline phosphatase, collagen type I, osteocalcin, and collagen type II were also performed by immunochemistry and molecular biology. RESULTS: The phenotype and expansion possibility of hMSCs after cryopreservation were remained. It could expand for 10 generations. The doubling time was 40 h. CONCLUSION: The bank of hMSCs is incipiently established and can provide eligible seed cells for tissue engineering.


Subject(s)
Biological Specimen Banks , Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Alkaline Phosphatase/metabolism , Antigens, Surface/metabolism , Bone Marrow Cells/metabolism , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type II/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteocalcin/genetics , Osteocalcin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Engineering
18.
Arch Facial Plast Surg ; 7(1): 7-11, 2005.
Article in English | MEDLINE | ID: mdl-15655167

ABSTRACT

OBJECTIVES: To assess the possibility of cartilage tissue engineering using human mesenchymal stem cells (hMSCs) and to investigate the quantitative relationship between hMSCs and engineered cartilage. DESIGN: Human mesenchymal stem cells were cultured, cryopreserved, and expanded in vitro. Surface antigens were detected by flow cytometry. In vitro chondrogenesis of hMSCs and cryopreserved hMSCs was performed. The chondrogenesis-induced hMSCs were seeded onto polyglycolic acid scaffolds, cultured in vitro for 3 weeks in chondrogenic medium, and then implanted into nude mice. The implants were harvested after 10 weeks and examined with histologic and immunochemical staining. RESULTS: The construction of cartilages was identified grossly and histologically: 1.9 to 2.5 x 10(7) nucleated cells were obtained from 1 mL of bone marrow, and about 1 to 2 x 10(6) hMSCs were obtained from the primary culture. The number of hMSCs tripled at every passage and reached 1.4 to 2.8 x 10(12) at passage 15. The purity of hMSCs was 95% and 98% at the primary and the fourth passages, respectively. Twenty-one days was the optimal (induction rate, 95%) induction time, with no apparent differences in induction rates among different passages. Based on our findings, hMSCs from 0.07 to 0.14 mL of bone marrow, expanded during 4 passages and induced for 21 days, would be sufficient to engineer 1 cm(2) of cartilage, 3-mm thick. CONCLUSION: Quantitative standards of hMSCs as seed cells for cartilage tissue engineering were established and may have value for later clinical work.


Subject(s)
Bone Marrow Cells/cytology , Cartilage/cytology , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Animals , Cell Culture Techniques , Chondrogenesis/physiology , Humans , Mice , Mice, Nude
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 20(3): 306-9, 2004 May.
Article in Chinese | MEDLINE | ID: mdl-15193224

ABSTRACT

AIM: To investigate the feasibility of human bone marrow mesenchymal stem cells(hMSCs) as the seed cells for bone and cartilage tissue engineering. METHODS: Purified hMSCs were cultured in-vitro and induced to differentiate into osteoblasts and chondrocytes. Cellular morphologies were observed under inverted and electron microscopes. The specific markers of the osteoblasts and chondrocytes were detected by histochemical staining, immunohistochemical staining, and RT-PCR. RESULTS: After the hMSCs were passaged for 15 generations, the choractenistic morphology and cell surface antigens of hMSCs remained unchanged. The level of alkaline phosphatase(ALP) in the culture supernatant of the osteoinduction groups was higher than those in the control groups (P<0.05). The morphology of the cells in the osteoinduction and chondroinduction groups changed from spindle-shaped cells into polygon-shaped cells. A large number of the dilated rough endoplasmic reticulua, Golgi apparatus and mitochondria could be seen under transmission electron microscope. Calcium deposition was detected on the surfaces of the hMSCs after osteoinduction. Collagen(COL)-like processes were detected under scanning electron microscope. The staining of the ALP, calcium nudis, COL-I and osteocalcin(OC) were positive, and expressions of the COL-I and OC mRNAs were detected after osteoinduction. The expression of COL-II was detected by immunohistochemical staining and RT-PCR and a lot of the metachromatic-staining matrix around the cells was observed with toluidine blue staining after chondroinduction. CONCLUSION: hMSCs from human bone marrow can be purified, expanded and differentiated into osteoblasts and chondrocytes in-vitro, providing an alternative source for bone and cartilage tissue engineering.


Subject(s)
Bone Marrow Cells/cytology , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Alkaline Phosphatase/metabolism , Ascorbic Acid/pharmacology , Bone Marrow Cells/ultrastructure , Calcium/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/ultrastructure , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type II/biosynthesis , Collagen Type II/genetics , Dexamethasone/pharmacology , Humans , Mesenchymal Stem Cells/ultrastructure , Osteoblasts/metabolism , Osteoblasts/ultrastructure , Osteocalcin/biosynthesis , Osteocalcin/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tissue Engineering , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
20.
Article in Chinese | MEDLINE | ID: mdl-15211841

ABSTRACT

OBJECTIVE: To optimize the culture conditions of the human bone marrow mesenchymal stem cells (hMSCs). METHODS: The influence of the primary culture method, planting density, the time of the first medium changing, culture medium and serum concentration on growth of the hMSCs were analyzed. RESULTS: When the other conditions were the same, the density gradient isolation was better than whole-marrow isolation; 2.5 x 10(5) cells/cm2 was the best planting density; the best first medium changing was the fifth day at primary culture, DMEM medium was better than alpha-MEM, serum C was the best of four serums compared, 10% was the suitable serum concentration. The hMSCs under the optimal conditions could expand over 15 passages, remaining their normal modality and differentiation potentials. CONCLUSION: The optimal culture condition of the hMSCs is established and it is a new investigation on application of hMSCs to tissue engineering.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Cell Differentiation , Cell Proliferation , Cells, Cultured , Culture Media , Humans , Serum , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...