Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Extracell Vesicles ; 13(5): e12445, 2024 May.
Article in English | MEDLINE | ID: mdl-38711334

ABSTRACT

Small extracellular vesicles (sEV) derived from various cell sources have been demonstrated to enhance cardiac function in preclinical models of myocardial infarction (MI). The aim of this study was to compare different sources of sEV for cardiac repair and determine the most effective one, which nowadays remains limited. We comprehensively assessed the efficacy of sEV obtained from human primary bone marrow mesenchymal stromal cells (BM-MSC), human immortalized MSC (hTERT-MSC), human embryonic stem cells (ESC), ESC-derived cardiac progenitor cells (CPC), human ESC-derived cardiomyocytes (CM), and human primary ventricular cardiac fibroblasts (VCF), in in vitro models of cardiac repair. ESC-derived sEV (ESC-sEV) exhibited the best pro-angiogenic and anti-fibrotic effects in vitro. Then, we evaluated the functionality of the sEV with the most promising performances in vitro, in a murine model of MI-reperfusion injury (IRI) and analysed their RNA and protein compositions. In vivo, ESC-sEV provided the most favourable outcome after MI by reducing adverse cardiac remodelling through down-regulating fibrosis and increasing angiogenesis. Furthermore, transcriptomic, and proteomic characterizations of sEV derived from hTERT-MSC, ESC, and CPC revealed factors in ESC-sEV that potentially drove the observed functions. In conclusion, ESC-sEV holds great promise as a cell-free treatment for promoting cardiac repair following MI.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Myocardial Infarction , Myocytes, Cardiac , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Humans , Animals , Mice , Myocardial Infarction/therapy , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Male , Myocardial Reperfusion Injury/therapy , Myocardial Reperfusion Injury/metabolism , Disease Models, Animal , Neovascularization, Physiologic , Cells, Cultured
2.
Adv Sci (Weinh) ; 10(12): e2206187, 2023 04.
Article in English | MEDLINE | ID: mdl-36806740

ABSTRACT

Lipid nanoparticles (LNPs) are currently used to transport functional mRNAs, such as COVID-19 mRNA vaccines. The delivery of angiogenic molecules, such as therapeutic VEGF-A mRNA, to ischemic tissues for producing new blood vessels is an emerging strategy for the treatment of cardiovascular diseases. Here, the authors deliver VEGF-A mRNA via LNPs and study stoichiometric quantification of their uptake kinetics and how the transport of exogenous LNP-mRNAs between cells is functionally extended by cells' own vehicles called extracellular vesicles (EVs). The results show that cellular uptake of LNPs and their mRNA molecules occurs quickly, and that the translation of exogenously delivered mRNA begins immediately. Following the VEGF-A mRNA delivery to cells via LNPs, a fraction of internalized VEGF-A mRNA is secreted via EVs. The overexpressed VEGF-A mRNA is detected in EVs secreted from three different cell types. Additionally, RNA-Seq analysis reveals that as cells' response to LNP-VEGF-A mRNA treatment, several overexpressed proangiogenic transcripts are packaged into EVs. EVs are further deployed to deliver VEGF-A mRNA in vitro and in vivo. Upon equal amount of VEGF-A mRNA delivery via three EV types or LNPs in vitro, EVs from cardiac progenitor cells are the most efficient in promoting angiogenesis per amount of VEGF-A protein produced. Intravenous administration of luciferase mRNA shows that EVs could distribute translatable mRNA to different organs with the highest amounts of luciferase detected in the liver. Direct injections of VEGF-A mRNA (via EVs or LNPs) into mice heart result in locally produced VEGF-A protein without spillover to liver and circulation. In addition, EVs from cardiac progenitor cells cause minimal production of inflammatory cytokines in cardiac tissue compared with all other treatment types. Collectively, the data demonstrate that LNPs transform EVs as functional extensions to distribute therapeutic mRNA between cells, where EVs deliver this mRNA differently than LNPs.


Subject(s)
COVID-19 , Extracellular Vesicles , Mice , Animals , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , COVID-19/metabolism , Extracellular Vesicles/metabolism
3.
Life (Basel) ; 12(5)2022 May 12.
Article in English | MEDLINE | ID: mdl-35629393

ABSTRACT

Cardiac hypertrophy is a condition that may contribute to the development of heart failure. In this study, we compare the gene-expression patterns of our in vitro stem-cell-based cardiac hypertrophy model with the gene expression of biopsies collected from hypertrophic human hearts. Twenty-five differentially expressed genes (DEGs) from both groups were identified and the expression of selected corresponding secreted proteins were validated using ELISA and Western blot. Several biomarkers, including CCN2, THBS1, NPPA, and NPPB, were identified, which showed significant overexpressions in the hypertrophic samples in both the cardiac biopsies and in the endothelin-1-treated cells, both at gene and protein levels. The protein-interaction network analysis revealed CCN2 as a central node among the 25 overlapping DEGs, suggesting that this gene might play an important role in the development of cardiac hypertrophy. GO-enrichment analysis of the 25 DEGs revealed many biological processes associated with cardiac function and the development of cardiac hypertrophy. In conclusion, we identified important similarities between ET-1-stimulated human-stem-cell-derived cardiomyocytes and human hypertrophic cardiac tissue. Novel putative cardiac hypertrophy biomarkers were identified and validated on the protein level, lending support for further investigations to assess their potential for future clinical applications.

4.
J Am Chem Soc ; 144(15): 6734-6741, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35385274

ABSTRACT

The determination of intracellular drug concentrations can provide a better understanding of the drug function and efficacy. Ideally, this should be performed nondestructively, with no modification of either the drug or the target, and with the capability to detect low amounts of the molecule of interest, in many cases in the µM to nM range (pmol to fmol per million cells). Unfortunately, it is currently challenging to have an experimental technique that provides direct quantitative measurements of intracellular drug concentrations that simultaneously satisfies these requirements. Here, we show that magic-angle spinning dynamic nuclear polarization (MAS DNP) can be used to fulfill these requirements. We apply a quantitative 15N MAS DNP approach in combination with 15N labeling to quantify the intracellular amount of the drug [15N]CHIR-98014, an activator of the Wingless and Int-1 signaling pathway, determining intracellular drug amounts in the range of tens to hundreds of picomoles per million cells. This is, to our knowledge, the first time that MAS DNP has been used to successfully estimate intracellular drug amounts.


Subject(s)
Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy/methods
5.
Life (Basel) ; 12(2)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35207580

ABSTRACT

Cardiac hypertrophy is an important and independent risk factor for the development of cardiac myopathy that may lead to heart failure. The mechanisms underlying the development of cardiac hypertrophy are yet not well understood. To increase the knowledge about mechanisms and regulatory pathways involved in the progression of cardiac hypertrophy, we have developed a human induced pluripotent stem cell (hiPSC)-based in vitro model of cardiac hypertrophy and performed extensive characterization using a multi-omics approach. In a series of experiments, hiPSC-derived cardiomyocytes were stimulated with Endothelin-1 for 8, 24, 48, and 72 h, and their transcriptome and secreted proteome were analyzed. The transcriptomic data show many enriched canonical pathways related to cardiac hypertrophy already at the earliest time point, e.g., cardiac hypertrophy signaling. An integrated transcriptome-secretome analysis enabled the identification of multimodal biomarkers that may prove highly relevant for monitoring early cardiac hypertrophy progression. Taken together, the results from this study demonstrate that our in vitro model displays a hypertrophic response on both transcriptomic- and secreted-proteomic levels. The results also shed novel insights into the underlying mechanisms of cardiac hypertrophy, and novel putative early cardiac hypertrophy biomarkers have been identified that warrant further investigation to assess their potential clinical relevance.

6.
Biol Open ; 9(9)2020 09 21.
Article in English | MEDLINE | ID: mdl-32878883

ABSTRACT

Cardiac hypertrophy is an important and independent risk factor for the development of heart failure. To better understand the mechanisms and regulatory pathways involved in cardiac hypertrophy, there is a need for improved in vitro models. In this study, we investigated how hypertrophic stimulation affected human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs). The cells were stimulated with endothelin-1 (ET-1) for 8, 24, 48, 72, or 96 h. Parameters including cell size, ANP-, proBNP-, and lactate concentration were analyzed. Moreover, transcriptional profiling using RNA-sequencing was performed to identify differentially expressed genes following ET-1 stimulation. The results show that the CMs increase in size by approximately 13% when exposed to ET-1 in parallel to increases in ANP and proBNP protein and mRNA levels. Furthermore, the lactate concentration in the media was increased indicating that the CMs consume more glucose, a hallmark of cardiac hypertrophy. Using RNA-seq, a hypertrophic gene expression pattern was also observed in the stimulated CMs. Taken together, these results show that hiPSC-derived CMs stimulated with ET-1 display a hypertrophic response. The results from this study also provide new molecular insights about the underlying mechanisms of cardiac hypertrophy and may help accelerate the development of new drugs against this condition.


Subject(s)
Cardiomegaly/pathology , Myocytes, Cardiac/cytology , Biomarkers , Cell Differentiation , Cell Size , Cells, Cultured , Computational Biology/methods , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Ontology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/pathology , Transcriptome
7.
Drug Discov Today ; 23(11): 1812-1817, 2018 11.
Article in English | MEDLINE | ID: mdl-29883729

ABSTRACT

Mini-organs engineered from decellularized organs repopulated with human stem cells can transform preclinical model strategies in target validation and biomarker discovery. Recellularized organs are whole humanized organs with preserved native architecture, conformity of the organ, composition of extracellular matrix and vascular matrix structures. With mini-organ models further understanding of developmental biology and assessment of potential therapeutic targets can be elucidated utilizing human induced pluripotent stem cells. As a next step, co-cultured mini-organ models could simulate pharmacokinetics and pharmacodynamics in physiological and pathological conditions. By overcoming key challenges, the development of humanized mini-organs as integrated biotechnology can address the translational gaps between in vitro, ex vivo and in vivo systems for an elevated human target validation model.


Subject(s)
Coculture Techniques/methods , Tissue Culture Techniques/methods , Tissue Engineering/methods , Tissue Scaffolds , Translational Research, Biomedical/methods , Humans , Models, Biological
8.
Sci Rep ; 8(1): 7458, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29748585

ABSTRACT

Despite improvements in pre-clinical drug testing models, predictability of clinical outcomes continues to be inadequate and costly due to poor evidence of drug metabolism. Humanized miniature organs integrating decellularized rodent organs with tissue specific cells are translational models that can provide further physiological understanding and evidence. Here, we evaluated 4-Flow cannulated rat hearts as the fundamental humanized organ model for cardiovascular drug validation. Results show clearance of cellular components in all chambers in 4-Flow hearts with efficient perfusion into both coronary arteries and cardiac veins. Furthermore, material characterization depicts preserved organization and content of important matrix proteins such as collagens, laminin, and elastin. With access to the complete vascular network, different human cell types were delivered to show spatial distribution and integration into the matrix under perfusion for up to three weeks. The feature of 4-Flow cannulation is the preservation of whole heart conformity enabling ventricular pacing via the pulmonary vein as demonstrated by noninvasive monitoring with fluid pressure and ultrasound imaging. Consequently, 4-Flow hearts surmounting organ mimicry challenges with intact complexity in vasculature and mechanical compliance of the whole organ providing an ideal platform for improving pre-clinical drug validation in addition to understanding cardiovascular diseases.


Subject(s)
Catheterization/methods , Extracellular Matrix/ultrastructure , Heart/physiology , Myocardium/ultrastructure , Perfusion/methods , Tissue Scaffolds/chemistry , Animals , Collagen/analysis , Drug Evaluation, Preclinical/methods , Elastin/analysis , Extracellular Matrix/chemistry , Extracellular Matrix Proteins/analysis , HEK293 Cells , Humans , Male , Myocardium/chemistry , Myocardium/cytology , Rats , Rats, Sprague-Dawley , Tissue Engineering/methods , Translational Research, Biomedical/methods
9.
J Biomed Mater Res B Appl Biomater ; 103(1): 39-46, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24757041

ABSTRACT

Within the past two decades polylactic-co-glycolic acid (PLGA) has gained considerable attention as a biocompatible and biodegradable polymer that is suitable for tissue engineering and regenerative medicine. In this present study, we have investigated the potential of PLGA, collagen I (ColI), and polyurethane (PU) scaffolds for ligament tissue regeneration. Two different ratios of PLGA (50:50 and 85:15) were used to determine the effects on mechanical tensile properties and cell adhesion. The Young's modulus, tensile stress at yield, and ultimate tensile strain of PLGA(50:50)-ColI-PU scaffolds demonstrated similar tensile properties to that of ligaments found in the knee. Whereas, scaffolds composed of PLGA(85:15)-ColI-PU had lower tensile properties than that of ligaments. Furthermore, we investigated the effect of fiber orientation on mechanical properties and our results indicate that aligned fiber scaffolds demonstrate higher tensile properties than scaffolds with random fiber orientation. Also, human fibroblasts attached and proliferated with no need for additional surface modifications to the presented electrospun scaffolds in both categories. Collectively, our investigation demonstrates the effectiveness of electrospun PLGA scaffolds as a suitable candidate for regenerative medicine, capable of being manipulated and combined with other polymers to create three-dimensional microenvironments with adjustable tensile properties to mimic native tissues.


Subject(s)
Collagen Type I/chemistry , Lactic Acid/chemistry , Ligaments , Polyglycolic Acid/chemistry , Polyurethanes/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry , Elastic Modulus , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Materials Testing , Polylactic Acid-Polyglycolic Acid Copolymer
10.
J Biomed Mater Res B Appl Biomater ; 102(8): 1730-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24687591

ABSTRACT

We examined the effects of the microenvironment on vascular differentiation of murine cardiovascular progenitor cells (CPCs). We isolated CPCs and seeded them in culture exposed to the various extracellular matrix (ECM) proteins in both two-dimensional (2D) and 3D culture systems. To better understand the contribution of the microenvironment to vascular differentiation, we analyzed endothelial and smooth muscle cell differentiation at both day 7 and day 14. We found that laminin and vitronectin enhanced vascular endothelial cell differentiation while fibronectin enhanced vascular smooth muscle cell differentiation. We also observed that the effects of the 3D electrospun scaffolds were delayed and not noticeable until the later time point (day 14), which may be due to the amount of time necessary for the cells to migrate to the interior of the scaffold. The study characterized the contributions of both ECM proteins and the addition of a 3D culture system to continued vascular differentiation. Additionally, we demonstrated the capability bioengineer a CPC-derived vascular graft.


Subject(s)
Cell Differentiation , Cellular Microenvironment , Endothelial Cells/metabolism , Myocardium/metabolism , Myocytes, Smooth Muscle/metabolism , Stem Cells/metabolism , Animals , Blood Vessel Prosthesis , Cells, Cultured , Endothelial Cells/cytology , Extracellular Matrix/chemistry , Mice , Myocardium/cytology , Myocytes, Smooth Muscle/cytology , Stem Cells/cytology , Tissue Scaffolds/chemistry
11.
Stem Cells Transl Med ; 3(2): 161-71, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24396033

ABSTRACT

White mature adipocytes give rise to so-called dedifferentiated fat (DFAT) cells that spontaneously undergo multilineage differentiation. In this study, we defined stem cell characteristics of DFAT cells as they are generated from adipocytes and the relationship between these characteristics and lineage differentiation. Both mouse and human DFAT cells, prepared from adipose tissue and lipoaspirate, respectively, showed evidence of pluripotency, with a maximum 5-7 days after adipocyte isolation. The DFAT cells spontaneously formed clusters in culture, which transiently expressed multiple stem cell markers, including stage-specific embryonic antigens, and Sca-1 (mouse) and CD105 (human), as determined by real-time polymerase chain reaction, fluorescence-activated cell sorting, and immunostaining. As the stem cell markers decreased, markers characteristic of the three germ layers and specific lineage differentiation, such as α-fetoprotein (endoderm, hepatic), Neurofilament-66 (ectoderm, neurogenic), and Troponin I (mesoderm, cardiomyogenic), increased. However, no teratoma formation was detected after injection in immunodeficient mice. A novel modification of the adipocyte isolation aimed at ensuring the initial purity of the adipocytes and avoiding ceiling culture allowed isolation of DFAT cells with pluripotent characteristics. Thus, the adipocyte-derived DFAT cells represent a plastic stem cell population that is highly responsive to changes in culture conditions and may benefit cell-based therapies.


Subject(s)
Adipocytes, White/cytology , Cell Dedifferentiation/physiology , Myocardial Infarction/pathology , Myocardium/cytology , Pluripotent Stem Cells/cytology , Teratoma/pathology , Animals , Biomarkers/metabolism , Cell Culture Techniques/methods , Cells, Cultured , Disease Models, Animal , Green Fluorescent Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Teratoma/etiology
12.
J Vis Exp ; (79): e50585, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24121366

ABSTRACT

In 2001, researchers at the University of California, Los Angeles, described the isolation of a new population of adult stem cells from liposuctioned adipose tissue that they initially termed Processed Lipoaspirate Cells or PLA cells. Since then, these stem cells have been renamed as Adipose-derived Stem Cells or ASCs and have gone on to become one of the most popular adult stem cells populations in the fields of stem cell research and regenerative medicine. Thousands of articles now describe the use of ASCs in a variety of regenerative animal models, including bone regeneration, peripheral nerve repair and cardiovascular engineering. Recent articles have begun to describe the myriad of uses for ASCs in the clinic. The protocol shown in this article outlines the basic procedure for manually and enzymatically isolating ASCs from large amounts of lipoaspirates obtained from cosmetic procedures. This protocol can easily be scaled up or down to accommodate the volume of lipoaspirate and can be adapted to isolate ASCs from fat tissue obtained through abdominoplasties and other similar procedures.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/cytology , Cytological Techniques/methods , Tissue and Organ Harvesting/methods , Centrifugation/methods , Humans , Lipectomy
13.
ISRN Tissue Eng ; 20132013.
Article in English | MEDLINE | ID: mdl-33426049

ABSTRACT

The relationship between stem cell niches in vivo and their surrounding microenvironment is still relatively unknown. Recent advances have indicated that extrinsic factors within the cardiovascular progenitor cell niche influence maintenance of a multipotent state as well as drive cell-fate decisions. We have previously shown the direct effects of extracellular matrix (ECM) proteins and have now investigated the effects of dimension on the induction of a cardiovascular progenitor cell (CPC) population. We have shown here that the three-dimensionality of a hyaluronan-based hydrogel greatly induces a CPC population, as marked by Flk-1. We have compared the effects of a 3D microenvironment to those of conventional 2D cell culture practices and have found that the 3D microenvironment potently induces a progenitor cell state.

14.
J Mol Cell Cardiol ; 53(6): 790-800, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22999861

ABSTRACT

White mature adipocytes give rise to multipotent cells, so-called de-differentiated fat (DFAT) cells, when losing their fat in culture. The objective of this study was to examine the ability of DFAT cells to give rise to endothelial cells (ECs) in vitro and vivo. We demonstrate that mouse and human DFAT cells, derived from adipose tissue and lipospirate, respectively, initially lack expression of CD34, CD31, CD146, CD45 and pericyte markers, distinguishing them from progenitor cells previously identified in adipose stroma. The DFAT cells spontaneously differentiate into vascular ECs in vitro, as determined by real-time PCR, fluorescence activated cell sorting, immunostaining, and formation of tube structures. Treatment with bone morphogenetic protein (BMP)4 and BMP9, important in regulating angiogenesis, significantly enhances the EC differentiation. Furthermore, adipocyte-derived cells from Green Fluorescent Protein-transgenic mice were detected in the vasculature of infarcted myocardium up to 6 weeks after ligation of the left anterior descending artery in mice. We conclude that adipocyte-derived multipotent cells are able to spontaneously give rise to ECs, a process that is promoted by BMPs and may be important in cardiovascular regeneration and in physiological and pathological changes in fat and other tissues.


Subject(s)
Adipocytes, White/cytology , Cell Dedifferentiation/physiology , Cell Differentiation/physiology , Multipotent Stem Cells/cytology , Adipocytes, White/drug effects , Animals , Bone Morphogenetic Protein 4/pharmacology , Cell Dedifferentiation/drug effects , Cell Differentiation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression , Growth Differentiation Factors/pharmacology , Humans , Male , Mice , Mice, Transgenic , Multipotent Stem Cells/drug effects , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Stem Cell Transplantation
15.
Biomaterials ; 33(7): 2032-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22169822

ABSTRACT

While stem cell niches in vivo are complex three-dimensional (3D) microenvironments, the relationship between the dimensionality of the niche to its function is unknown. We have created a 3D microenvironment through electrospinning to study the impact of geometry and different extracellular proteins on the development of cardiac progenitor cells (Flk-1(+)) from resident stem cells and their differentiation into functional cardiovascular cells. We have investigated the effect of collagen IV, fibronectin, laminin and vitronectin on the adhesion and proliferation of murine ES cells as well as the effects of these proteins on the number of Flk-1(+) cells cultured in 2D conditions compared to 3D system in a feeder free condition. We found that the number of Flk-1(+) cells was significantly higher in 3D scaffolds coated with laminin or vitronectin compared to colIV-coated scaffolds. Our results show the importance of defined culture systems in vitro for studying the guided differentiation of pluripotent embryonic stem cells in the field of cardiovascular tissue engineering and regenerative medicine.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Vitronectin/metabolism , Animals , Biocompatible Materials/metabolism , Cell Proliferation , Cells, Cultured , Collagen Type IV/metabolism , Embryonic Stem Cells/cytology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Heart/embryology , Humans , Laminin/metabolism , Mice , Myocardium/cytology , Tissue Engineering/methods , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
J Biomed Mater Res B Appl Biomater ; 99(1): 180-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21732530

ABSTRACT

Electrospinning using synthetic and natural polymers is a promising technique for the fabrication of scaffolds for tissue engineering. Numerous synthetic polymers are available to maximize durability and mechanical properties (polyurethane) versus degradability and cell adhesion (polycaprolactone). In this study, we explored the feasibility of creating scaffolds made of bicomponent nanofibers from both polymers using a coaxial electrospinning system. We used a core of poly(urethane) and a sheath of a mixture of poly(ε-caprolactone) and gelatin, all dissolved in 1,1,1,3,3,3-hexafluror-2-propanol. These nanofibrous scaffolds were then evaluated to confirm their core-sheath nature and characterize their morphology and mechanical properties under static and dynamic conditions. Furthermore, the antigenicity of the scaffolds was studied to confirm that there is no significant foreign body response to the scaffold itself that would preclude its use in vivo. The results show the advantages of combining both natural and synethic polymers to create a coaxial scaffold capable of withstanding dynamic culture conditions and encourage cellular migration to the interior of the scaffold for tissue-engineering applications. Also, the results show that there is no significant immunoreactivity in vivo to the components of the scaffolds.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Foreign-Body Reaction/immunology , Nanofibers/chemistry , Tissue Engineering/instrumentation , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Electrochemical Techniques/methods , Gelatin/chemistry , Implants, Experimental , Materials Testing , Mice , NIH 3T3 Cells , Nanofibers/ultrastructure , Polyesters/chemistry , Polymers/chemical synthesis , Polymers/chemistry , Polymers/metabolism , Polyurethanes/chemistry , Porosity , Stress, Mechanical
17.
Biomaterials ; 30(27): 4665-75, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19524289

ABSTRACT

Synthetic polymers or naturally-derived extracellular matrix (ECM) proteins have been used to create tissue engineering scaffolds; however, the need for surface modification in order to achieve polymer biocompatibility and the lack of biomechanical strength of constructs built using proteins alone remain major limitations. To overcome these obstacles, we developed novel hybrid constructs composed of both strong biosynthetic materials and natural human ECM proteins. Taking advantage of the ability of cells to produce their own ECM, human foreskin fibroblasts were grown on silicon-based nanostructures exhibiting various surface topographies that significantly enhanced ECM protein production. After 4 weeks, cell-derived sheets were harvested and histology, immunochemistry, biochemistry and multiphoton imaging revealed the presence of collagens, tropoelastin, fibronectin and glycosaminoglycans. Following decellularization, purified sheet-derived ECM proteins were mixed with poly(epsilon-caprolactone) to create fibrous scaffolds using electrospinning. These hybrid scaffolds exhibited excellent biomechanical properties with fiber and pore sizes that allowed attachment and migration of adipose tissue-derived stem cells. Our study represents an innovative approach to generate strong, non-cytotoxic scaffolds that could have broad applications in tissue regeneration strategies.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/metabolism , Nanostructures/chemistry , Regenerative Medicine/methods , Adipose Tissue/cytology , Biological Assay , Extracellular Matrix/drug effects , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fluorescence , Humans , Photons , Polyesters/pharmacology , Porosity/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/ultrastructure , Tissue Scaffolds
18.
J Biomed Mater Res A ; 89(3): 804-17, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18523950

ABSTRACT

Cells in vivo encounter with and react to the extracellular matrix materials on a nanometer scale. Recent advances in nanofabrication technologies allowing the precise control of a nanostructure's pattern, periodicity, shape, and height have enabled a systematic study of cell interactions with three-dimensional nanotopographies. In this report, we examined the behavior of human foreskin fibroblasts on well-ordered dense arrays (post and grate patterns with a 230-nm pitch) of sharp-tip nanostructures with varying three-dimensionalities (from 50 to 600 nm in structural height) over time-until a cell sheet was formed. Although cells started out smaller and proliferated slower on tall nanostructures (both posts and grates) than on smooth surfaces, they became confluent to form a sheet in 3 weeks. On grate patterns, significant cell elongation in alignment with the underlying pattern was observed and maintained over time. On tall nanostructures, cells grew while raised on sharp tips, resulting in a weak total adherence to the solid surface. A sheet of cells was easily peeled off from such surfaces, suggesting that nanoscale topographies can be used as the basis for cell-sheet tissue engineering.


Subject(s)
Fibroblasts/cytology , Nanostructures/chemistry , Cell Adhesion , Cell Proliferation , Cell Shape , Cells, Cultured , Fibroblasts/ultrastructure , Humans , Image Processing, Computer-Assisted , Male
19.
Stem Cells ; 26(6): 1537-46, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18450826

ABSTRACT

Forced expression of the four transcription factors Oct4, Sox2, c-Myc, and Klf4 is sufficient to confer a pluripotent state upon the murine fibroblast genome, generating induced pluripotent stem (iPS) cells. Although the differentiation potential of these cells is thought to be equivalent to that of embryonic stem (ES) cells, it has not been rigorously determined. In this study, we sought to identify the capacity of iPS cells to differentiate into Flk1-positive progenitors and their mesodermal progeny, including cells of the cardiovascular and hematopoietic lineages. Immunostaining of tissues from iPS cell-derived chimeric mice demonstrated that iPS cells could contribute in vivo to cardiomyocytes, smooth muscle cells, endothelial cells, and hematopoietic cells. To compare the in vitro differentiation potential of murine ES and iPS cells, we either induced embryoid body (EB) formation of each cell type or cultured the cells on collagen type IV (ColIV), an extracellular matrix protein that had been reported to direct murine ES cell differentiation to mesodermal lineages. EB formation and exposure to ColIV both induced iPS cell differentiation into cells that expressed cardiovascular and hematopoietic markers. To determine whether ColIV-differentiated iPS cells contained a progenitor cell with cardiovascular and hematopoietic differentiation potential, Flk1-positive cells were isolated by magnetic cell sorting and exposed to specific differentiation conditions, which induced differentiation into functional cardiomyocytes, smooth muscle cells, endothelial cells, and hematopoietic cells. Our data demonstrate that murine iPS cells, like ES cells, can differentiate into cells of the cardiovascular and hematopoietic lineages and therefore may represent a valuable cell source for applications in regenerative medicine. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Cell Differentiation/physiology , Endothelium, Vascular/cytology , Fibroblasts/cytology , Hematopoietic Stem Cells/cytology , Muscle, Smooth/cytology , Myocytes, Cardiac/cytology , Animals , Cell Culture Techniques/methods , Genes, Reporter , Genome , Green Fluorescent Proteins/genetics , Kruppel-Like Factor 4 , Mice , Transcription Factors/genetics , Transcription Factors/physiology
20.
Biomaterials ; 29(19): 2907-14, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403012

ABSTRACT

Electrospinning using natural proteins or synthetic polymers is a promising technique for the fabrication of fibrous scaffolds for various tissue engineering applications. However, one limitation of scaffolds electrospun from natural proteins is the need to cross-link with glutaraldehyde for stability, which has been postulated to lead to many complications in vivo including graft failure. In this study, we determined the characteristics of hybrid scaffolds composed of natural proteins including collagen and elastin, as well as gelatin, and the synthetic polymer poly(epsilon-caprolactone) (PCL), so to avoid chemical cross-linking. Fiber size increased proportionally with increasing protein and polymer concentrations, whereas pore size decreased. Electrospun gelatin/PCL scaffolds showed a higher tensile strength when compared to collagen/elastin/PCL constructs. To determine the effects of pore size on cell attachment and migration, both hybrid scaffolds were seeded with adipose-derived stem cells. Scanning electron microscopy and nuclei staining of cell-seeded scaffolds demonstrated the complete cell attachment to the surfaces of both hybrid scaffolds, although cell migration into the scaffold was predominantly seen in the gelatin/PCL hybrid. The combination of natural proteins and synthetic polymers to create electrospun fibrous structures resulted in scaffolds with favorable mechanical and biological properties.


Subject(s)
Cardiovascular System , Extracellular Matrix , Tissue Engineering , Adipose Tissue/cytology , Humans , Microscopy, Electron , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...