1
|
Inal MS, Avci H, Hassan S, Darcan C, Shin SR, Akpek A. Advances in xenogeneic donor decellularized organs: A review on studies with sheep and porcine-derived heart valves. Bioeng Transl Med 2024; 9:e10695. [PMID: 39545084 PMCID: PMC11558188 DOI: 10.1002/btm2.10695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/31/2024] [Accepted: 06/24/2024] [Indexed: 11/17/2024] Open
Abstract
Heart valve replacement surgeries are performed on patients suffering from abnormal heart valve function. In these operations, the problematic tissue is replaced with mechanical valves or with bioprosthetics that are being developed. The thrombotic effect of mechanical valves, reflecting the need for lifelong use of anticoagulation drugs, and the short-lived nature of biological valves make these two types of valves problematic. In addition, they cannot adapt to the somatic growth of young patients. Although decellularized scaffolds have shown some promise, a successful translation has so far evaded. Although decellularized porcine xenografts have been extensively studied in the literature, they have several disadvantages, such as a propensity for calcification in the implant model, a risk of porcine endogenous retrovirus (PERV) infection, and a high xenoantigen density. As seen in clinical data, it is clear that there are biocompatibility problems in almost all studies. However, since decellularized sheep heart valves have not been tried in the clinic, a large data pool could not be established. This review compares and contrasts decellularized porcine and sheep xenografts for heart valve tissue engineering. It reveals that decellularized sheep heart valves can be an alternative to pigs in terms of biocompatibility. In addition, it highlights the potential advantages of bioinks derived from the decellularized extracellular matrix in 3D bioprinting technology, emphasizing that they can be a new alternative for the application. We also outline the future prospects of using sheep xenografts for heart valve tissue engineering.
Collapse
Affiliation(s)
- Muslum Suleyman Inal
- Department of Molecular Biology and GeneticsBilecik Seyh Edebali UniversityBilecikTurkey
| | - Huseyin Avci
- Translational Medicine Research and Clinical Center, Cellular Therapy and Stem Cell Production Application and Research CenterEskisehir Osmangazi UniversityTurkey
| | - Shabir Hassan
- Department of BiologyKhalifa UniversityAbu DhabiUnited Arab Emirates
| | - Cihan Darcan
- Department of Molecular Biology and GeneticsBilecik Seyh Edebali UniversityBilecikTurkey
| | - Su Ryon Shin
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Ali Akpek
- Department of Biomedical EngineeringYildiz Technical UniversityTurkey
| |
Collapse
|
2
|
Nasim S, Abujamra BA, Chaparro D, Nogueira PDS, Riva A, Hutcheson JD, Kos L. Multiple cell types including melanocytes contribute to elastogenesis in the developing murine aortic valve. Sci Rep 2024; 14:25481. [PMID: 39461968 PMCID: PMC11513076 DOI: 10.1038/s41598-024-73673-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Elastic fibers are crucial for aortic valve (AoV) function and are generated and maintained by valvular interstitial cells (VICs). VICs exhibit diverse phenotypes, yet the specific subpopulation responsible for producing and regulating elastic fibers remains unclear. This gap in knowledge is significant, given that elastin (Eln) abnormalities lead to congenital AoV defects and initiate AoV diseases. This study characterizes the timing of Eln expression in murine AoV, revealing it peaks during late embryogenesis and early postnatal stages, decreasing in adulthood. Spatial transcriptomics and RT-qPCR indicate that Eln expression correlates with genes associated to elastogenesis, including Acta2, a smooth muscle cell marker. While Eln expression is not exclusive to a single VIC subpopulation, RNAscope and immunofluorescence demonstrate a population of Eln-expressing VICs that co-express alpha smooth muscle actin and melanocytic markers. As previously reported in adult mice, we show a relationship between AoV pigment and elastic fiber patterning during early postnatal stages and further show that melanocytes may play a critical role in elastogenesis. In summary, Eln is expressed in the AoV during early postnatal stages by cells co-expressing markers of various types, highlighting the complexity of VICs phenotypes and their role in elastic fiber regulation.
Collapse
Affiliation(s)
- Sana Nasim
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Beatriz Abdo Abujamra
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Daniel Chaparro
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Perony Da Silva Nogueira
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | | | - Joshua D Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA.
| | - Lidia Kos
- Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199, USA.
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
3
|
Inflammatory and regenerative processes in bioresorbable synthetic pulmonary valves up to two years in sheep-Spatiotemporal insights augmented by Raman microspectroscopy. Acta Biomater 2021; 135:243-259. [PMID: 34509697 DOI: 10.1016/j.actbio.2021.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/18/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
In situ heart valve tissue engineering is an emerging approach in which resorbable, off-the-shelf available scaffolds are used to induce endogenous heart valve restoration. Such scaffolds are designed to recruit endogenous cells in vivo, which subsequently resorb polymer and produce and remodel new valvular tissue in situ. Recently, preclinical studies using electrospun supramolecular elastomeric valvular grafts have shown that this approach enables in situ regeneration of pulmonary valves with long-term functionality in vivo. However, the evolution and mechanisms of inflammation, polymer absorption and tissue regeneration are largely unknown, and adverse valve remodeling and intra- and inter-valvular variability have been reported. Therefore, the goal of the present study was to gain a mechanistic understanding of the in vivo regenerative processes by combining routine histology and immunohistochemistry, using a comprehensive sheep-specific antibody panel, with Raman microspectroscopy for the spatiotemporal analysis of in situ tissue-engineered pulmonary valves with follow-up to 24 months from a previous preclinical study in sheep. The analyses revealed a strong spatial heterogeneity in the influx of inflammatory cells, graft resorption, and foreign body giant cells. Collagen maturation occurred predominantly between 6 and 12 months after implantation, which was accompanied by a progressive switch to a more quiescent phenotype of infiltrating cells with properties of valvular interstitial cells. Variability among specimens in the extent of tissue remodeling was observed for follow-up times after 6 months. Taken together, these findings advance the understanding of key events and mechanisms in material-driven in situ heart valve tissue engineering. STATEMENT OF SIGNIFICANCE: This study describes for the first time the long-term in vivo inflammatory and regenerative processes that underly in situ heart valve tissue engineering using resorbable synthetic scaffolds. Using a unique combinatorial analysis of immunohistochemistry and Raman microspectroscopy, important spatiotemporal variability in graft resorption and tissue formation was pinpointed in in situ tissue-engineered heart valves, with a follow-up time of up to 24 months in sheep. This variability was correlated to heterogenous regional cellular repopulation, most likely instigated by region-specific differences in surrounding tissue and hemodynamics. The findings of this research contribute to the mechanistic understanding of in situ tissue engineering using resorbable synthetics, which is necessary to enable rational design of improved grafts, and ensure safe and robust clinical translation.
Collapse
|
4
|
Jannasch A, Schnabel C, Galli R, Faak S, Büttner P, Dittfeld C, Tugtekin SM, Koch E, Matschke K. Optical coherence tomography and multiphoton microscopy offer new options for the quantification of fibrotic aortic valve disease in ApoE -/- mice. Sci Rep 2021; 11:5834. [PMID: 33712671 PMCID: PMC7955095 DOI: 10.1038/s41598-021-85142-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/25/2021] [Indexed: 01/31/2023] Open
Abstract
Aortic valve sclerosis is characterized as the thickening of the aortic valve without obstruction of the left ventricular outflow. It has a prevalence of 30% in people over 65 years old. Aortic valve sclerosis represents a cardiovascular risk marker because it may progress to moderate or severe aortic valve stenosis. Thus, the early recognition and management of aortic valve sclerosis are of cardinal importance. We examined the aortic valve geometry and structure from healthy C57Bl6 wild type and age-matched hyperlipidemic ApoE-/- mice with aortic valve sclerosis using optical coherence tomography (OCT) and multiphoton microscopy (MPM) and compared results with histological analyses. Early fibrotic thickening, especially in the tip region of the native aortic valve leaflets from the ApoE-/- mice, was detectable in a precise spatial resolution using OCT. Evaluation of the second harmonic generation signal using MPM demonstrated that collagen content decreased in all aortic valve leaflet regions in the ApoE-/- mice. Lipid droplets and cholesterol crystals were detected using coherent anti-Stokes Raman scattering in the tissue from the ApoE-/- mice. Here, we demonstrated that OCT and MPM, which are fast and precise contactless imaging approaches, are suitable for defining early morphological and structural alterations of sclerotic murine aortic valves.
Collapse
Affiliation(s)
- Anett Jannasch
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstraße 76, 01307, Dresden, Germany.
| | - Christian Schnabel
- Department of Anesthesiology and Intensive Care Medicine and Clinical Sensoring and Monitoring, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Roberta Galli
- Department of Anesthesiology and Intensive Care Medicine and Clinical Sensoring and Monitoring, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Saskia Faak
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstraße 76, 01307, Dresden, Germany
| | - Petra Büttner
- Department of Cardiology, Heart Center Leipzig At University Leipzig, Leipzig, Germany
| | - Claudia Dittfeld
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstraße 76, 01307, Dresden, Germany
| | - Sems Malte Tugtekin
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstraße 76, 01307, Dresden, Germany
| | - Edmund Koch
- Department of Anesthesiology and Intensive Care Medicine and Clinical Sensoring and Monitoring, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Klaus Matschke
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Fetscherstraße 76, 01307, Dresden, Germany
| |
Collapse
|
5
|
Waqanivavalagi SWFR, Bhat S, Ground MB, Milsom PF, Cornish J. Clinical performance of decellularized heart valves versus standard tissue conduits: a systematic review and meta-analysis. J Cardiothorac Surg 2020; 15:260. [PMID: 32948234 PMCID: PMC7501674 DOI: 10.1186/s13019-020-01292-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023] Open
Abstract
Background Valve replacement surgery is the definitive management strategy for patients with severe valvular disease. However, valvular conduits currently in clinical use are associated with significant limitations. Tissue-engineered (decellularized) heart valves are alternative prostheses that have demonstrated promising early results. The purpose of this systematic review and meta-analysis is to perform robust evaluation of the clinical performance of decellularized heart valves implanted in either outflow tract position, in comparison with standard tissue conduits. Methods Systematic searches were conducted in the PubMed, Scopus, and Web of Science databases for articles in which outcomes between decellularized heart valves surgically implanted within either outflow tract position of human subjects and standard tissue conduits were compared. Primary endpoints included postoperative mortality and reoperation rates. Meta-analysis was performed using a random-effects model via the Mantel-Haenszel method. Results Seventeen articles were identified, of which 16 were included in the meta-analysis. In total, 1418 patients underwent outflow tract reconstructions with decellularized heart valves and 2725 patients received standard tissue conduits. Decellularized heart valves were produced from human pulmonary valves and implanted within the right ventricular outflow tract in all cases. Lower postoperative mortality (4.7% vs. 6.1%; RR 0.94, 95% CI: 0.60–1.47; P = 0.77) and reoperation rates (4.8% vs. 7.4%; RR 0.55, 95% CI: 0.36–0.84; P = 0.0057) were observed in patients with decellularized heart valves, although only reoperation rates were statistically significant. There was no statistically significant heterogeneity between the analyzed articles (I2 = 31%, P = 0.13 and I2 = 33%, P = 0.10 respectively). Conclusions Decellularized heart valves implanted within the right ventricular outflow tract have demonstrated significantly lower reoperation rates when compared to standard tissue conduits. However, in order to allow for more accurate conclusions about the clinical performance of decellularized heart valves to be made, there need to be more high-quality studies with greater consistency in the reporting of clinical outcomes.
Collapse
Affiliation(s)
- Steve W F R Waqanivavalagi
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand. .,Adult Emergency Department, Auckland City Hospital, Auckland District Health Board, Grafton, Auckland, 1023, New Zealand.
| | - Sameer Bhat
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand.,Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand
| | - Marcus B Ground
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand.,Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, 9054, New Zealand
| | - Paget F Milsom
- Green Lane Cardiothoracic Surgical Unit, Auckland City Hospital, Auckland District Health Board, Grafton, Auckland, 1023, New Zealand
| | - Jillian Cornish
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
6
|
Non-invasive functional molecular phenotyping of human smooth muscle cells utilized in cardiovascular tissue engineering. Acta Biomater 2019; 89:193-205. [PMID: 30878445 DOI: 10.1016/j.actbio.2019.03.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 12/20/2022]
Abstract
Smooth muscle cell (SMC) diversity and plasticity are limiting factors in their characterization and application in cardiovascular tissue engineering. This work aimed to evaluate the potential of Raman microspectroscopy and Raman imaging to distinguish SMCs of different tissue origins and phenotypes. Cultured human SMCs isolated from different vascular and non-vascular tissues as well as fixed human SMC-containing tissues were analyzed. In addition, Raman spectra and images of tissue-engineered SMC constructs were acquired. Routine techniques such as qPCR, histochemistry, histological and immunocytological staining were performed for comparative gene and protein expression analysis. We identified that SMCs of different tissue origins exhibited unique spectral information that allowed a separation of all groups of origin by multivariate data analysis (MVA). We were further able to non-invasively monitor phenotypic switching in cultured SMCs and assess the impact of different culture conditions on extracellular matrix remodeling in the tissue-engineered ring constructs. Interestingly, we identified that the Raman signature of the human SMC-based ring constructs was similar to the one obtained from native aortic tissue. We conclude that Raman microspectroscopic methods are promising tools to characterize cells and define cellular and extracellular matrix components on a molecular level. In this study, in situ measurements were marker-independent, fast, and identified cellular differences that were not detectable by established routine techniques. Perspectively, Raman microspectroscopy and MVA in combination with artificial intelligence can be suitable for automated quality monitoring of (stem) cell and cell-based tissue engineering products. STATEMENT OF SIGNIFICANCE: The accessibility of autologous blood vessels for surgery is limited. Tissue engineering (TE) aims to develop functional vascular replacements; however, no commercially available TE vascular graft (TEVG) exists to date. One limiting factor is the availability of a well-characterized and safe cell source. Smooth muscle cells (SMCs) are generally used for TEVGs. To engineer a TEVG, proliferating SMCs of the synthesizing phenotype are essential, whereas functional, sustainable TEVGs require SMCs of the contractile phenotype. SMC diversity and plasticity are therefore limiting factors, also for their quality monitoring and application in TE. In this study, Raman microspectroscopy and imaging combined with machine learning tools allowed the non-destructive, marker-independent characterization of SMCs, smooth muscle tissues and TE SMC-constructs. The spectral information was specific enough to distinguish for the first time the phenotypic switching in SMCs in real-time, and monitor the impact of culture conditions on ECM remodeling in the TE SMC-constructs.
Collapse
|
7
|
Beziere N, Fuchs K, Maurer A, Reischl G, Brück J, Ghoreschi K, Fehrenbacher B, Berrio DC, Schenke-Layland K, Kohlhofer U, Quintanilla-Martinez L, Gawaz M, Kneilling M, Pichler B. Imaging fibrosis in inflammatory diseases: targeting the exposed extracellular matrix. Theranostics 2019; 9:2868-2881. [PMID: 31244929 PMCID: PMC6568181 DOI: 10.7150/thno.28892] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/10/2018] [Indexed: 01/09/2023] Open
Abstract
In a variety of diseases, from benign to life-threatening ones, inflammation plays a major role. Monitoring the intensity and extent of a multifaceted inflammatory process has become a cornerstone in diagnostics and therapy monitoring. However, the current tools lack the ability to provide insight into one of its most crucial aspects, namely, the alteration of the extracellular matrix (ECM). Using a radiolabeled platelet glycoprotein VI-based ECM-targeting fusion protein (GPVI-Fc), we investigated how binding of GPVI-Fc on fibrous tissue could uncover the progression of several inflammatory disease models at different stages (rheumatoid arthritis, cutaneous delayed-type hypersensitivity, lung inflammation and experimental autoimmune encephalomyelitis). Methods: The fusion protein GPVI-Fc was covalently linked to 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) and subsequently labeled with 64Cu. We analyzed noninvasively in vivo64Cu-GPVI-Fc accumulation in murine cutaneous delayed-type hypersensitivity, anti-glucose-6-phosphate isomerase serum-induced rheumatoid arthritis, lipopolysaccharide-induced lung inflammation and an experimental autoimmune encephalomyelitis model. Static and dynamic Positron Emission Tomography (PET) of the radiotracer distribution was performed in vivo, with ex vivo autoradiography confirmation, yielding quantitative accumulation and a distribution map of 64Cu-GPVI-Fc. Ex vivo tissue histological staining was performed on harvested samples to highlight the fusion protein binding to collagen I, II and III, fibronectin and fibrinogen as well as the morphology of excised tissue. Results:64Cu-GPVI-Fc showed a several-fold increased uptake in inflamed tissue compared to control tissue, particularly in the RA model, with a peak 24 h after radiotracer injection of up to half the injected dose. Blocking and isotype control experiments indicated a target-driven accumulation of the radiotracer in the case of chronic inflammation. Histological analysis confirmed a prolonged accumulation at the inflammation site, with a pronounced colocalization with the different components of the ECM (collagen III and fibronectin notably). Binding of the fusion protein appeared to be specific to the ECM but unspecific to particular components. Conclusion: Imaging of 64Cu-GPVI-Fc accumulation in the ECM matrix appears to be a promising candidate for monitoring chronic inflammation. By binding to exposed fibrous tissue (collagen, fibronectin, etc.) after extravasation, a new insight is provided into the fibrotic events resulting from a prolonged inflammatory state.
Collapse
Affiliation(s)
- Nicolas Beziere
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Kerstin Fuchs
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Jürgen Brück
- Department of Dermatology, University Medical Center, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, University Medical Center, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Birgit Fehrenbacher
- Department of Dermatology, University Medical Center, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Daniel Carvajal Berrio
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
- The Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
- Department of Medicine/ Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, 675 Charles E. Young Drive South, MRL 3645, Los Angeles, CA, USA
| | - Ursula Kohlhofer
- Institute of Pathology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University Hospital Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Department of Dermatology, University Medical Center, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Bernd Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
8
|
Wu J, Brazile B, McMahan SR, Liao J, Hong Y. Heart valve tissue-derived hydrogels: Preparation and characterization of mitral valve chordae, aortic valve, and mitral valve gels. J Biomed Mater Res B Appl Biomater 2018; 107:1732-1740. [PMID: 30419146 DOI: 10.1002/jbm.b.34266] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/30/2018] [Accepted: 09/30/2018] [Indexed: 12/21/2022]
Abstract
Heart valve (HV) diseases are among the leading causes of death and continue to threaten public health worldwide. The current clinical options for HV replacement include mechanical and biological prostheses. However, an ongoing problem with current HV prostheses is their failure to integrate with the host tissue and their inability grow and remodel within the body. Tissue engineered heart valves (TEHVs) are a promising solution to these problems, as they are able to grow and remodel somatically with the rest of the body. Recently, decellularized HVs have demonstrated great potential as valve replacements because they are tissue specific, but recellularization is still a challenge due to the dense HV extracellular matrix (ECM) network. In this proof-of-concept work, we decellularized porcine mitral valve chordae, aortic valve leaflets, and mitral valve leaflets and processed them into injectable hydrogels that could accommodate any geometry. While the three valvular ECMs contained various amounts of collagen, they displayed similar glycosaminoglycan contents. The hydrogels had similar nanofibrous structures and gelation kinetics with various compressive strengths. When encapsulated with NIH 3 T3 fibroblasts, all the hydrogels supported cell survivals up to 7 days. Decellularized HV ECM hydrogels may show promising potential HV tissue engineering applications. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1732-1740, 2019.
Collapse
Affiliation(s)
- Jinglei Wu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, 76019.,Joint Graduate Biomedical Engineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, 75390
| | - Bryn Brazile
- Department of Biological Engineering, Mississippi State University, Starkville, Mississippi, 39762
| | - Sara R McMahan
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, 76019.,Joint Graduate Biomedical Engineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, 75390
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, 76019.,Joint Graduate Biomedical Engineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, 75390.,Department of Biological Engineering, Mississippi State University, Starkville, Mississippi, 39762
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, 76019.,Joint Graduate Biomedical Engineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, 75390
| |
Collapse
|
9
|
Dekker S, van Geemen D, van den Bogaerdt AJ, Driessen-Mol A, Aikawa E, Smits AIPM. Sheep-Specific Immunohistochemical Panel for the Evaluation of Regenerative and Inflammatory Processes in Tissue-Engineered Heart Valves. Front Cardiovasc Med 2018; 5:105. [PMID: 30159315 PMCID: PMC6104173 DOI: 10.3389/fcvm.2018.00105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
The creation of living heart valve replacements via tissue engineering is actively being pursued by many research groups. Numerous strategies have been described, aimed either at culturing autologous living valves in a bioreactor (in vitro) or inducing endogenous regeneration by the host via resorbable scaffolds (in situ). Whereas a lot of effort is being invested in the optimization of heart valve scaffold parameters and culturing conditions, the pathophysiological in vivo remodeling processes to which tissue-engineered heart valves are subjected upon implantation have been largely under-investigated. This is partly due to the unavailability of suitable immunohistochemical tools specific to sheep, which serves as the gold standard animal model in translational research on heart valve replacements. Therefore, the goal of this study was to comprise and validate a comprehensive sheep-specific panel of antibodies for the immunohistochemical analysis of tissue-engineered heart valve explants. For the selection of our panel we took inspiration from previous histopathological studies describing the morphology, extracellular matrix composition and cellular composition of native human heart valves throughout development and adult stages. Moreover, we included a range of immunological markers, which are particularly relevant to assess the host inflammatory response evoked by the implanted heart valve. The markers specifically identifying extracellular matrix components and cell phenotypes were tested on formalin-fixed paraffin-embedded sections of native sheep aortic valves. Markers for inflammation and apoptosis were tested on ovine spleen and kidney tissues. Taken together, this panel of antibodies could serve as a tool to study the spatiotemporal expression of proteins in remodeling tissue-engineered heart valves after implantation in a sheep model, thereby contributing to our understanding of the in vivo processes which ultimately determine long-term success or failure of tissue-engineered heart valves.
Collapse
Affiliation(s)
- Sylvia Dekker
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Daphne van Geemen
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | | | - Anita Driessen-Mol
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Anthal I. P. M. Smits
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
10
|
Ueda Y, Torrianni MW, Coppin CM, Iwai S, Sawa Y, Matsuda H. Antigen Clearing from Porcine Heart Valves with Preservation of Structural Integrity. Int J Artif Organs 2018; 29:781-9. [PMID: 16969756 DOI: 10.1177/039139880602900808] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bioprostheses currently used for replacement of diseased cardiovascular tissue are preserved and partially protected from immune rejection through chemical fixation. However, after implantation, chemically preserved (fixed) material has limited durability and lacks the ability to revitalize through cellular ingrowth and remodeling. As an alternative to fixation, we aimed at thoroughly removing antigens from tissue, leaving an intact scaffold, suitable for integration and revitalization in the host. Extensive washing of porcine heart valves with a mixture of two detergents (SDS and Triton X-100) yielded an intact matrix devoid of cells and depleted of soluble proteins that was minimally immunogenic in rabbits. A detailed characterization of the biomechanics and durability of the tissue is under way. If the lack of immunogenicity is confirmed in primates, our results would suggest that a detergent-washed, unfixed porcine heart valve can be an attractive non-inflammatory scaffold for heart valve regeneration in humans.
Collapse
Affiliation(s)
- Y Ueda
- Medtronic Heart Valve, Santa Ana, California, USA
| | | | | | | | | | | |
Collapse
|
11
|
Ji W, Li L, Eniola-Adefeso O, Wang Y, Liu C, Feng C. Non-invasively visualizing cell-matrix interactions in two-photon excited supramolecular hydrogels. J Mater Chem B 2017; 5:7790-7795. [PMID: 32264379 DOI: 10.1039/c7tb02274c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Visualizing the role of extracellular matrix (ECM) in cell bioactivities in three-dimensional (3D) view is highly important for in-depth understanding of fundamental physiological issues in various in vitro experiments. Using current designs it is difficult to produce 3D biomimetic ECM with intrinsic fluorescence under non-invasive near-infrared excitation. Herein, we have designed and synthesized a series of non-conventional coumarin-derived hydrogelators, which can self-assemble to form nanofibrous 3D supramolecular hydrogels through C-HO bonds and be excited by two-photon absorption, ensuring the direct and dynamic visualization of cell-matrix interactions with high resolution images in a 3D environment. Real-time monitoring of ECM-regulated dynamic cell behaviours is highly desirable for future basic and applied research.
Collapse
Affiliation(s)
- Wei Ji
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, China.
| | | | | | | | | | | |
Collapse
|
12
|
Roosens A, Puype I, Cornelissen R. Scaffold-free high throughput generation of quiescent valvular microtissues. J Mol Cell Cardiol 2017; 106:45-54. [PMID: 28322869 DOI: 10.1016/j.yjmcc.2017.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022]
Abstract
AIMS The major challenge of working with valvular interstitial cells in vitro is the preservation or recovery of their native quiescent state. In this study, a biomimetic approach is used which aims to engineer small volume, high quality valve microtissues, having a potential in regenerative medicine and as a relevant 3D in vitro model to provide insights into valve (patho)biology. METHODS AND RESULTS To form micro-aggregates, porcine valvular interstitial cells were seeded in agarose micro-wells and cultured in medium supplemented with 250μM Ascorbic Acid 2-phosphate for 22days. Histology showed viable aggregates with normal nuclei and without any signs of calcification. Aggregates stained strongly for GAG and collagen I and reticular fibers were present. ECM formation was quantified and showed a significant increase of GAG, elastin and Col I during aggregate culture. Cultivation of VIC in aggregates also promoted mRNA expression of Col I/III/V, elastin, hyaluronan, biglycan, decorin, versican MMP-1/2/3/9 and TIMP-2 compared to monolayer cultured VIC. Phenotype analysis of aggregates showed a significant decrease in α-SMA expression, and an increase in FSP-1 expression at any time point. Furthermore, VIC aggregates did not show a significant difference in OCN, Egr-1, Sox-9 or Runx2 expression. CONCLUSION In this study high quality valvular interstitial cell aggregates were generated that are able to produce their own ECM, resembling the native valve composition. The applied and completely cell driven 3D approach overcomes the problems of VIC activation in 2D, by downregulating α-SMA expression and stimulating a homeostatic quiescent VIC state.
Collapse
Affiliation(s)
- Annelies Roosens
- Department of Basic Medical Sciences, Tissue Engineering Group, Ghent University, Ghent, Belgium.
| | - Inès Puype
- Department of Basic Medical Sciences, Tissue Engineering Group, Ghent University, Ghent, Belgium.
| | - Ria Cornelissen
- Department of Basic Medical Sciences, Tissue Engineering Group, Ghent University, Ghent, Belgium.
| |
Collapse
|
13
|
Lee PF, Chau E, Cabello R, Yeh AT, Sampaio LC, Gobin AS, Taylor DA. Inverted orientation improves decellularization of whole porcine hearts. Acta Biomater 2017; 49:181-191. [PMID: 27884776 DOI: 10.1016/j.actbio.2016.11.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/14/2016] [Accepted: 11/20/2016] [Indexed: 12/11/2022]
Abstract
In structurally heterogeneous organs, such as heart, it is challenging to retain extracellular matrix integrity in the thinnest regions (eg, valves) during perfusion decellularization and completely remove cellular debris from thicker areas. The high inflow rates necessary to maintain physiologic pressure can distend or damage thin tissues, but lower pressures prolong the process and increase the likelihood of contamination. We examined two novel retrograde decellularization methods for porcine hearts: inverting the heart or venting the apex to decrease inflow rate. We measured flow dynamics through the aorta (Ao) and pulmonary artery (PA) at different Ao pressures and assessed the heart's appearance, turbidity of the outflow solutions, and coronary perfusion efficiency. We used rectangle image fitting of decellularized heart images to obtain a heart shape index. Using nonlinear optical microscopy, we determined the microstructure of collagen and elastin fibers of the aortic valve cusps. DNA, glycosaminoglycan, and residual detergent levels were compared. The inverted method was superior to the vented method, as shown by a higher coronary perfusion efficiency, more cell debris outflow, higher collagen and elastin content inside the aortic valve, lower DNA content, and better retention of the heart shape after decellularization. To our knowledge, this is the first study to use flow dynamics in a whole heart throughout the decellularization procedure to provide real-time information about the success of the process and the integrity of the vulnerable regions of the matrix. Heart orientation was important in optimizing decellularization efficiency and maintaining extracellular matrix integrity. STATEMENT OF SIGNIFICANCE The use of decellularized tissue as a suitable scaffold for engineered tissue has emerged over the past decade as one of the most promising biofabrication platforms. The decellularization process removes all native cells, leaving the natural biopolymers, extracellular matrix materials and native architecture intact. This manuscript describes heart orientation as important in optimizing decellularization efficiency and maintaining extracellular matrix integrity. To our knowledge, this is the first study to assess flow dynamics in a whole heart throughout the decellularization procedure. Our findings compared to currently published methods demonstrate that continuous complex real-time measurements and analyses are required to produce an optimal scaffold for cardiac regeneration.
Collapse
|
14
|
Jahnavi S, Saravanan U, Arthi N, Bhuvaneshwar GS, Kumary TV, Rajan S, Verma RS. Biological and mechanical evaluation of a Bio-Hybrid scaffold for autologous valve tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 73:59-71. [PMID: 28183649 DOI: 10.1016/j.msec.2016.11.116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/10/2016] [Accepted: 11/23/2016] [Indexed: 10/20/2022]
Abstract
Major challenge in heart valve tissue engineering for paediatric patients is the development of an autologous valve with regenerative capacity. Hybrid tissue engineering approach is recently gaining popularity to design scaffolds with desired biological and mechanical properties that can remodel post implantation. In this study, we fabricated aligned nanofibrous Bio-Hybrid scaffold made of decellularized bovine pericardium: polycaprolactone-chitosan with optimized polymer thickness to yield the desired biological and mechanical properties. CD44+, αSMA+, Vimentin+ and CD105- human valve interstitial cells were isolated and seeded on these Bio-Hybrid scaffolds. Subsequent biological evaluation revealed interstitial cell proliferation with dense extra cellular matrix deposition that indicated the viability for growth and proliferation of seeded cells on the scaffolds. Uniaxial mechanical tests along axial direction showed that the Bio-Hybrid scaffolds has at least 20 times the strength of the native valves and its stiffness is nearly 3 times more than that of native valves. Biaxial and uniaxial mechanical studies on valve interstitial cells cultured Bio-Hybrid scaffolds revealed that the response along the axial and circumferential direction was different, similar to native valves. Overall, our findings suggest that Bio-Hybrid scaffold is a promising material for future development of regenerative heart valve constructs in children.
Collapse
Affiliation(s)
- S Jahnavi
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, TN 600036, India; Tissue Culture Laboratory, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, Kerala 695012, India
| | - U Saravanan
- Department of Civil Engineering, Indian Institute of Technology Madras, Chennai, TN 600036, India
| | - N Arthi
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, TN 600036, India
| | - G S Bhuvaneshwar
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, TN 600036, India
| | - T V Kumary
- Tissue Culture Laboratory, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Trivandrum, Kerala 695012, India
| | - S Rajan
- Madras Medical Mission, Institute of Cardio-Vascular Diseases, Mogappair, Chennai, Tamil Nadu 600037, India
| | - R S Verma
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, TN 600036, India.
| |
Collapse
|
15
|
Wu Y, Grande-Allen KJ, West JL. Adhesive Peptide Sequences Regulate Valve Interstitial Cell Adhesion, Phenotype and Extracellular Matrix Deposition. Cell Mol Bioeng 2016; 9:479-495. [PMID: 28220141 PMCID: PMC5315271 DOI: 10.1007/s12195-016-0451-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 05/30/2016] [Indexed: 12/13/2022] Open
Abstract
Knowledge of how extracellular matrix (ECM) binding impacts valve interstitial cells (VICs) is critical not only to better understanding the etiology of valvular diseases but also to constructing living valve substitutes that can grow and remodel. Use of ECM-mimicking adhesive peptides with specific affinity to different receptors provides insights into adhesion-mediated cell signaling and downstream outcomes. Expression of adhesion receptors by VICs was assessed by flow cytometry and used to guide the choice of peptides studied. The peptide RGDS with affinity to multiple integrin receptors, and specific receptor-targeting peptides DGEA (integrin α2β1), YIGSR (67kDa laminin/elastin receptor; 67LR), and VAPG (67LR) were incorporated into hydrogels to investigate their effects on VICs. DGEA, YIGSR, and VAPG alone were insufficient to induce stable VIC adhesion. As a result, these peptides were studied in combination with 1 mM RGDS. For VICs cultured on two-dimensional hydrogel surfaces, YIGSR and VAPG down-regulated the expression of smooth muscle α-actin (myofibroblast activation marker); DGEA promoted VIC adhesion and VIC-mediated ECM deposition and inhibited the activity of alkaline phosphatase (osteogenic differentiation marker). Further, YIGSR and DGEA in combination promoted ECM deposition while inhibiting both myofibroblastic and osteogenic differentiation. However, VICs behaved differently to adhesive ligands when cultured within three-dimensional hydrogels, with most VICs assuming a healthy, quiescent phenotype under all peptide conditions tested. DGEA promoted ECM deposition by VICs within hydrogels. Overall, we demonstrate that the presentation of defined peptides targeting specific adhesion receptors can be used to regulate VIC adhesion, phenotype and ECM synthesis.
Collapse
Affiliation(s)
- Yan Wu
- Department of Biomedical Engineering, Duke University, Durham, NC
27708
| | | | - Jennifer L. West
- Department of Biomedical Engineering, Duke University, Durham, NC
27708
| |
Collapse
|
16
|
Calcific Aortic Valve Disease Is Associated with Layer-Specific Alterations in Collagen Architecture. PLoS One 2016; 11:e0163858. [PMID: 27685946 PMCID: PMC5042542 DOI: 10.1371/journal.pone.0163858] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/15/2016] [Indexed: 11/20/2022] Open
Abstract
Disorganization of the valve extracellular matrix (ECM) is a hallmark of calcific aortic valve disease (CAVD). However, while microarchitectural features of the ECM can strongly influence the biological and mechanical behavior of tissues, little is known about the ECM microarchitecture in CAVD. In this work, we apply advanced imaging techniques to quantify spatially heterogeneous changes in collagen microarchitecture in CAVD. Human aortic valves were obtained from individuals between 50 and 75 years old with no evidence of valvular disease (healthy) and individuals who underwent valve replacement surgery due to severe stenosis (diseased). Second Harmonic Generation microscopy and subsequent image quantification revealed layer-specific changes in fiber characteristics in healthy and diseased valves. Specifically, the majority of collagen fiber changes in CAVD were found to occur in the spongiosa, where collagen fiber number increased by over 2-fold, and fiber width and density also significantly increased. Relatively few fibrillar changes occurred in the fibrosa in CAVD, where fibers became significantly shorter, but did not otherwise change in terms of number, width, density, or alignment. Immunohistochemical staining for lysyl oxidase showed localized increased expression in the diseased fibrosa. These findings reveal a more complex picture of valvular collagen enrichment and arrangement in CAVD than has previously been described using traditional analysis methods. Changes in fiber architecture may play a role in regulating the pathobiological events and mechanical properties of valves during CAVD. Additionally, characterization of the ECM microarchitecture can inform the design of fibrous scaffolds for heart valve tissue engineering.
Collapse
|
17
|
Hof A, Raschke S, Baier K, Nehrenheim L, Selig JI, Schomaker M, Lichtenberg A, Meyer H, Akhyari P. Challenges in developing a reseeded, tissue-engineered aortic valve prosthesis. Eur J Cardiothorac Surg 2016; 50:446-55. [DOI: 10.1093/ejcts/ezw057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 02/03/2016] [Indexed: 11/13/2022] Open
|
18
|
Aamodt JM, Grainger DW. Extracellular matrix-based biomaterial scaffolds and the host response. Biomaterials 2016; 86:68-82. [PMID: 26890039 DOI: 10.1016/j.biomaterials.2016.02.003] [Citation(s) in RCA: 332] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 01/30/2016] [Accepted: 02/01/2016] [Indexed: 01/08/2023]
Abstract
Extracellular matrix (ECM) collectively represents a class of naturally derived proteinaceous biomaterials purified from harvested organs and tissues with increasing scientific focus and utility in tissue engineering and repair. This interest stems predominantly from the largely unproven concept that processed ECM biomaterials as natural tissue-derived matrices better integrate with host tissue than purely synthetic biomaterials. Nearly every tissue type has been decellularized and processed for re-use as tissue-derived ECM protein implants and scaffolds. To date, however, little consensus exists for defining ECM compositions or sources that best constitute decellularized biomaterials that might better heal, integrate with host tissues and avoid the foreign body response (FBR). Metrics used to assess ECM performance in biomaterial implants are arbitrary and contextually specific by convention. Few comparisons for in vivo host responses to ECM implants from different sources are published. This review discusses current ECM-derived biomaterials characterization methods including relationships between ECM material compositions from different sources, properties and host tissue response as implants. Relevant preclinical in vivo models are compared along with their associated advantages and limitations, and the current state of various metrics used to define material integration and biocompatibility are discussed. Commonly applied applications of these ECM-derived biomaterials as stand-alone implanted matrices and devices are compared with respect to host tissue responses.
Collapse
Affiliation(s)
- Joseph M Aamodt
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112-5820, USA
| | - David W Grainger
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112-5820, USA; Department of Pharmaceutics and Pharmaceutical Chemistry University of Utah, Salt Lake City, UT, 84112-5820, USA.
| |
Collapse
|
19
|
Capulli AK, MacQueen LA, Sheehy SP, Parker KK. Fibrous scaffolds for building hearts and heart parts. Adv Drug Deliv Rev 2016; 96:83-102. [PMID: 26656602 PMCID: PMC4807693 DOI: 10.1016/j.addr.2015.11.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 12/14/2022]
Abstract
Extracellular matrix (ECM) structure and biochemistry provide cell-instructive cues that promote and regulate tissue growth, function, and repair. From a structural perspective, the ECM is a scaffold that guides the self-assembly of cells into distinct functional tissues. The ECM promotes the interaction between individual cells and between different cell types, and increases the strength and resilience of the tissue in mechanically dynamic environments. From a biochemical perspective, factors regulating cell-ECM adhesion have been described and diverse aspects of cell-ECM interactions in health and disease continue to be clarified. Natural ECMs therefore provide excellent design rules for tissue engineering scaffolds. The design of regenerative three-dimensional (3D) engineered scaffolds is informed by the target ECM structure, chemistry, and mechanics, to encourage cell infiltration and tissue genesis. This can be achieved using nanofibrous scaffolds composed of polymers that simultaneously recapitulate 3D ECM architecture, high-fidelity nanoscale topography, and bio-activity. Their high porosity, structural anisotropy, and bio-activity present unique advantages for engineering 3D anisotropic tissues. Here, we use the heart as a case study and examine the potential of ECM-inspired nanofibrous scaffolds for cardiac tissue engineering. We asked: Do we know enough to build a heart? To answer this question, we tabulated structural and functional properties of myocardial and valvular tissues for use as design criteria, reviewed nanofiber manufacturing platforms and assessed their capabilities to produce scaffolds that meet our design criteria. Our knowledge of the anatomy and physiology of the heart, as well as our ability to create synthetic ECM scaffolds have advanced to the point that valve replacement with nanofibrous scaffolds may be achieved in the short term, while myocardial repair requires further study in vitro and in vivo.
Collapse
Affiliation(s)
- A K Capulli
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - L A MacQueen
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Sean P Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - K K Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
20
|
Brugmans MM, Soekhradj-Soechit RS, van Geemen D, Cox M, Bouten CV, Baaijens FP, Driessen-Mol A. Superior Tissue Evolution in Slow-Degrading Scaffolds for Valvular Tissue Engineering. Tissue Eng Part A 2016; 22:123-32. [DOI: 10.1089/ten.tea.2015.0203] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Marieke M.C.P. Brugmans
- Xeltis B.V., Eindhoven, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | - Daphne van Geemen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | - Carlijn V.C. Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Frank P.T. Baaijens
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Anita Driessen-Mol
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
21
|
Thakkar S, Fernandes H, Moroni L. Decellularized Extracellular Matrix Scaffolds for Cartilage Regeneration. Methods Mol Biol 2015; 1340:133-151. [PMID: 26445835 DOI: 10.1007/978-1-4939-2938-2_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Decellularized extracellular matrix (ECM) is gaining a lot of attention as a biomaterial for tissue engineering applications. This chapter describes the processing techniques for decellularization of cell-derived ECM and protocols for the fabrication of ECM-based scaffolds in the form of hydrogels or fibrous polymer meshes by electrospinning. It describes the protocols to analyze the morphology and presence of collagen in fabricated scaffolds using scanning electron microscope and Picrosirius Red staining respectively. Methods to evaluate the metabolic activity and proliferation of cells (resazurin-based assay and DNA assay, respectively) and gene expression are also presented. Furthermore, histological techniques to analyze the presence of sulfated glycosaminoglycans are also described.
Collapse
Affiliation(s)
- Shraddha Thakkar
- Department of Biomedical Engineering, Eindhoven University of Technology, 513, 5600MB, Eindhoven, The Netherlands.
| | - Hugo Fernandes
- Tissue Regeneration Department, University of Maastricht - MERLN Institute for Technology Inspired Regenerative Medicine Complex, Tissue Regeneration Department Universiteitsingel 40, 6229 ER, Maastricht, The Netherlands.
| | - Lorenzo Moroni
- Tissue Regeneration Department, University of Maastricht - MERLN Institute for Technology Inspired Regenerative Medicine Complex, Tissue Regeneration Department Universiteitsingel 40, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
22
|
Tremblay C, Ruel J, Bourget JM, Laterreur V, Vallières K, Tondreau MY, Lacroix D, Germain L, Auger FA. A new construction technique for tissue-engineered heart valves using the self-assembly method. Tissue Eng Part C Methods 2014; 20:905-15. [PMID: 24576074 DOI: 10.1089/ten.tec.2013.0698] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Tissue engineering appears as a promising option to create new heart valve substitutes able to overcome the serious drawbacks encountered with mechanical substitutes or tissue valves. The objective of this article is to present the construction method of a new entirely biological stentless aortic valve using the self-assembly method and also a first assessment of its behavior in a bioreactor when exposed to a pulsatile flow. A thick tissue was created by stacking several fibroblast sheets produced with the self-assembly technique. Different sets of custom-made templates were designed to confer to the thick tissue a three-dimensional (3D) shape similar to that of a native aortic valve. The construction of the valve was divided in two sequential steps. The first step was the installation of the thick tissue in a flat preshaping template followed by a 4-week maturation period. The second step was the actual cylindrical 3D forming of the valve. The microscopic tissue structure was assessed using histological cross sections stained with Masson's Trichrome and Picrosirius Red. The thick tissue remained uniformly populated with cells throughout the construction steps and the dense extracellular matrix presented corrugated fibers of collagen. This first prototype of tissue-engineered heart valve was installed in a bioreactor to assess its capacity to sustain a light pulsatile flow at a frequency of 0.5 Hz. Under the light pulsed flow, it was observed that the leaflets opened and closed according to the flow variations. This study demonstrates that the self-assembly method is a viable option for the construction of complex 3D shapes, such as heart valves, with an entirely biological material.
Collapse
Affiliation(s)
- Catherine Tremblay
- 1 Département de génie mécanique, Faculté des sciences et de génie, Université Laval , Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Le Huu A, Shum-Tim D. Tissue engineering of autologous heart valves: a focused update. Future Cardiol 2013; 10:93-104. [PMID: 24344666 DOI: 10.2217/fca.13.96] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The prevalence of valvular heart disease is expected to increase in the coming decades, with an associated rise in valve-related surgeries. Current options for valve prostheses remain limited, essentially confined to mechanical or biological valves. Neither selection provides an optimal balance between structural integrity and associated morbidity. Mechanical valves offer exceptional durability coupled with a considerable risk of thrombogenesis. Conversely, a biological prosthesis affords freedom from anticoagulation, but with a truncated valve lifespan. Tissue-engineered heart valves have been touted as a solution to this dilemma, by offering an immunopriviledged prosthesis combined with resistance from degeneration and the potential to grow. Although the reality of commercially available tissue-engineered heart valves remains distant, this article will highlight the cellular and clinical advancements in recent years.
Collapse
Affiliation(s)
- Alice Le Huu
- Division of Cardiac Surgery & Surgical Research, Department of Surgery, The Royal Victoria Hospital, McGill University Health Center, 687 Pine Avenue West, Suite S8.73b, Montreal, Quebec, H3A 1A1, Canada
| | | |
Collapse
|
24
|
Engineering of a bio-functionalized hybrid off-the-shelf heart valve. Biomaterials 2013; 35:2130-9. [PMID: 24333025 DOI: 10.1016/j.biomaterials.2013.10.080] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/31/2013] [Indexed: 11/20/2022]
Abstract
Currently available heart valve replacements are limited in long-term performance or fail due to leaflet thickening, lack of growth or remodeling potential. In order to address these issues, it is necessary to mimic multiple factors of the native valvular extracellular matrix (ECM) such as architecture, mechanical behavior and biochemical signals. Here, we successfully generated an electrospun PEGdma-PLA scaffold adapted to the structure and mechanical properties of native valve leaflets. Valvular interstitial cells (VICs) and valvular endothelial cells (VECs) were seeded on the scaffold and when cultured under physiological conditions in a bioreactor, the construct performed like a native leaflet. Atomic force microscopy (AFM) was employed to obtain detailed mechanical information from the leaflets, which enabled the first layer-specific measurement of the Young's modulus. Interestingly, spongiosa stiffness was much lower compared to the fibrosa and ventricularis. Moreover, investigations into human fetal heart valve development identified collagen type I and versican as important structural proteins. As a proof of principle, these proteins were introduced to the scaffold, demonstrating the ability to bio-functionalize the hybrid valve based on natures' blueprint.
Collapse
|
25
|
Abstract
Much has been made about the potential for stem cells in regenerative medicine but the reality is that the development of actual therapies has been slow. Adult stem cells rely heavily on the assortment of biochemical and biophysical elements that constitute the local microenvironment in which they exist. One goal of biomedicine is to create an artificial yet biofunctional niche to support multipotency, differentiation and proliferation. Such tools would facilitate more conclusive experimentation by biologists, pharmaceutical scientists and tissue engineers. While many bioengineering techniques and platforms are already in use, technological innovations now allow this to be done at a higher resolution and specificity. Ultimately, the multidisciplinary integration of engineering and biology will allow the niche to be generated at a scale that can be clinically exploited. Using the systems that constitute the intestinal, hematopoietic and epidermal tissues, this article summarizes the various approaches and tools currently employed to recreate stem cell niches and also explores recent advances in the field.
Collapse
Affiliation(s)
- Shawna Tan
- A*STAR Institute of Medical Biology , 8A Biomedical Grove, 06-06 Immunos , Singapore
| | | |
Collapse
|
26
|
Quinn RW, McFall C. Methods for analyzing the density, distribution, origin, and phenotypes of restored cell populations in vivo and post-explant. PROGRESS IN PEDIATRIC CARDIOLOGY 2013. [DOI: 10.1016/j.ppedcard.2013.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
27
|
Votteler M, Berrio DAC, Horke A, Sabatier L, Reinhardt DP, Nsair A, Aikawa E, Schenke-Layland K. Elastogenesis at the onset of human cardiac valve development. Development 2013; 140:2345-53. [PMID: 23637335 PMCID: PMC3912871 DOI: 10.1242/dev.093500] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Semilunar valve leaflets have a well-described trilaminar histoarchitecture, with a sophisticated elastic fiber network. It was previously proposed that elastin-containing fibers play a subordinate role in early human cardiac valve development; however, this assumption was based on data obtained from mouse models and human second and third trimester tissues. Here, we systematically analyzed tissues from human fetal first (4-12 weeks) and second (13-18 weeks) trimester, adolescent (14-19 years) and adult (50-55 years) hearts to monitor the temporal and spatial distribution of elastic fibers, focusing on semilunar valves. Global expression analyses revealed that the transcription of genes essential for elastic fiber formation starts early within the first trimester. These data were confirmed by quantitative PCR and immunohistochemistry employing antibodies that recognize fibronectin, fibrillin 1, 2 and 3, EMILIN1 and fibulin 4 and 5, which were all expressed at the onset of cardiac cushion formation (~week 4 of development). Tropoelastin/elastin protein expression was first detectable in leaflets of 7-week hearts. We revealed that immature elastic fibers are organized in early human cardiovascular development and that mature elastin-containing fibers first evolve in semilunar valves when blood pressure and heartbeat accelerate. Our findings provide a conceptual framework with the potential to offer novel insights into human cardiac valve development and disease.
Collapse
Affiliation(s)
- Miriam Votteler
- University Women's Hospital Tübingen and Inter-University Centre for Medical Technology Stuttgart-Tübingen (IZST), Eberhard Karls University, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Arjunon S, Rathan S, Jo H, Yoganathan AP. Aortic valve: mechanical environment and mechanobiology. Ann Biomed Eng 2013; 41:1331-46. [PMID: 23515935 DOI: 10.1007/s10439-013-0785-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 03/02/2013] [Indexed: 01/11/2023]
Abstract
The aortic valve (AV) experiences a complex mechanical environment, which includes tension, flexure, pressure, and shear stress forces due to blood flow during each cardiac cycle. This mechanical environment regulates AV tissue structure by constantly renewing and remodeling the phenotype. In vitro, ex vivo and in vivo studies have shown that pathological states such as hypertension and congenital defect like bicuspid AV (BAV) can potentially alter the AV's mechanical environment, triggering a cascade of remodeling, inflammation, and calcification activities in AV tissue. Alteration in mechanical environment is first sensed by the endothelium, which in turn induces changes in the extracellular matrix, and triggers cell differentiation and activation. However, the molecular mechanism of this process is not understood very well. Understanding these mechanisms is critical for advancing the development of effective medical based therapies. Recently, there have been some interesting studies on characterizing the hemodynamics associated with AV, especially in pathologies like BAV, using different experimental and numerical methods. Here, we review the current knowledge of the local AV mechanical environment and its effect on valve biology, focusing on in vitro and ex vivo approaches.
Collapse
Affiliation(s)
- Sivakkumar Arjunon
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Room 2119 U. A. Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332-0535, USA
| | | | | | | |
Collapse
|
29
|
Non-Destructive Analysis of Extracellular Matrix Development in Cardiovascular Tissue-Engineered Constructs. Ann Biomed Eng 2013; 41:883-93. [DOI: 10.1007/s10439-012-0734-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/19/2012] [Indexed: 10/27/2022]
|
30
|
Quinlan AMT, Billiar KL. Investigating the role of substrate stiffness in the persistence of valvular interstitial cell activation. J Biomed Mater Res A 2012; 100:2474-82. [PMID: 22581728 PMCID: PMC3880130 DOI: 10.1002/jbm.a.34162] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 02/26/2012] [Accepted: 02/28/2012] [Indexed: 01/13/2023]
Abstract
During heart valve remodeling and in many disease states, valvular interstitial cells (VICs) shift to an activated myofibroblast phenotype characterized by enhanced synthetic and contractile activity. Pronounced alpha smooth muscle actin (αSMA)-positive stress fibers, the hallmark of activated myofibroblasts, are also observed in VICs cultured on stiff substrates especially in the presence of transforming growth factor-beta1 (TGF-β1), however, the detailed relationship between stiffness and VIC phenotype has not been explored. The goal of this study was to characterize VIC activation as a function of substrate stiffness over a wide range of stiffness levels including that of diseased valves (stiff), normal valves (compliant), and hydrogels for heart valve tissue engineering (very soft). VICs obtained from porcine aortic valves were cultured on stiff tissue culture plastic to activate them, then, cultured on collagen-coated polyacrylamide substrates of predefined stiffness in a high-throughput culture system to assess the persistence of activation. Metrics extracted from regression analysis demonstrate that relative to a compliant substrate, stiff substrates result in higher cell numbers, more pronounced expression of αSMA-positive stress fibers, and larger spread area which is in qualitative agreement with previous studies. Our data also indicate that VICs require a much lower substrate stiffness level to "deactivate" them than previously thought. The high sensitivity of VICs to substrate stiffness demonstrates the importance of the mechanical properties of materials used for valve repair or for engineering valve tissue.
Collapse
Affiliation(s)
- Angela M. Throm Quinlan
- Dept of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA
| | - Kristen L. Billiar
- Dept of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA
- Dept of Surgery, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
31
|
Kusuma S, Zhao S, Gerecht S. The extracellular matrix is a novel attribute of endothelial progenitors and of hypoxic mature endothelial cells. FASEB J 2012; 26:4925-36. [PMID: 22919069 DOI: 10.1096/fj.12-209296] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Extracellular matrix (ECM) production is critical to preserve the function and integrity of mature blood vessels. Toward the engineering of blood vessels, studies have centered on ECM production by supporting cells, whereas few studies implicate endothelial cells (ECs) with ECM synthesis. Here, we elucidate variations between cultured human arterial, venous, and progenitor ECs with respect to ECM deposition assembly, composition, and response to biomolecular and physiological factors. Our studies reveal that progenitor ECs, endothelial colony-forming cells (ECFCs), deposit collagen IV, fibronectin, and laminin that assemble to an organized weblike structure, as confirmed by decellularized cultures. Mature ECs only express these ECM proteins intracellularly. ECFC-derived ECM is abrogated in response to TGFβ signaling inhibition and actin cytoskeleton disruption. Hypoxic (1%) and physiological (5%) O(2) tension stimulate ECM deposition from mature ECs. Interestingly, deposition of collagen I is observed only under 5% O(2) tension. ECM production from all ECs is found to be regulated by hypoxia-inducible factors 1α and 2α but differentially in the different cell lines. Collectively, we suggest that ECM deposition and assembly by ECs is dependent on maturation stage and oxygen supply and that these findings can be harnessed to advance engineered vascular therapeutics.
Collapse
Affiliation(s)
- Sravanti Kusuma
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
32
|
Choi YC, Choi JS, Kim BS, Kim JD, Yoon HI, Cho YW. Decellularized extracellular matrix derived from porcine adipose tissue as a xenogeneic biomaterial for tissue engineering. Tissue Eng Part C Methods 2012; 18:866-76. [PMID: 22559904 DOI: 10.1089/ten.tec.2012.0009] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cells in tissues are surrounded by the extracellular matrix (ECM), a gel-like material of proteins and polysaccharides that are synthesized and secreted by cells. Here we propose that the ECM can be isolated from porcine adipose tissue and holds great promise as a xenogeneic biomaterial for tissue engineering and regenerative medicine. Porcine adipose tissue is easily obtained in large quantities from commonly discarded food waste. Decellularization protocols have been developed for extracting an intact ECM while effectively eliminating xenogeneic epitopes and minimally disrupting the ECM composition. Porcine adipose tissue was defatted by homogenization and centrifugation. It was then decellularized via chemical (1.5 M sodium chloride and 0.5% sodium dodecyl sulfate) and enzymatic treatments (DNase and RNase) with temperature control. After decellularization, immunogenic components such as nucleic acids and α-Gal were significantly reduced. However, abundant ECM components, such as collagen (332.9±12.1 μg/mg ECM dry weight), sulfated glycosaminoglycan (GAG, 85±0.7 μg/mg ECM dry weight), and elastin (152.6±4.5 μg/mg ECM dry weight), were well preserved in the decellularized material. The biochemical and mechanical features of a decellularized ECM supported the adhesion and growth of human cells in vitro. Moreover, the decellularized ECM exhibited biocompatibility, long-term stability, and bioinductivity in vivo. The overall results suggest that the decellularized ECM derived from porcine adipose tissue could be useful as an alternative biomaterial for xenograft tissue engineering.
Collapse
Affiliation(s)
- Young Chan Choi
- Departments of Chemical Engineering and Bionanotechnology, Hanyang University, Ansan, Republic of Korea
| | | | | | | | | | | |
Collapse
|
33
|
The adhering junctions of valvular interstitial cells: molecular composition in fetal and adult hearts and the comings and goings of plakophilin-2 in situ, in cell culture and upon re-association with scaffolds. Cell Tissue Res 2012; 348:295-307. [DOI: 10.1007/s00441-011-1315-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 12/20/2011] [Indexed: 10/14/2022]
|
34
|
Gerson CJ, Elkins RC, Goldstein S, Heacox AE. Structural integrity of collagen and elastin in SynerGraft® decellularized-cryopreserved human heart valves. Cryobiology 2011; 64:33-42. [PMID: 22119932 DOI: 10.1016/j.cryobiol.2011.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 11/08/2011] [Accepted: 11/09/2011] [Indexed: 10/15/2022]
Abstract
SynerGraft® (SG) decellularized-cryopreserved cardiac valve allografts have been developed to provide a valve replacement option that has reduced antigenicity, retained structural integrity, and the ability to be stored long-term until needed for implantation. However, it is critical to ensure that both the SG processing and cryopreservation of these allografts do not detrimentally affect the extracellular matrix architecture within the tissue. This study evaluates the effects of SG decellularization and subsequent cryopreservation on the extracellular matrix integrity of allograft heart valves. Human aortic and pulmonary valves were trisected, with one-third of each either left fresh (no further processing after dissection), decellularized, or decellularized and cryopreserved. Two-photon laser scanning confocal microscopy was used to visualize collagen and elastin in leaflets and conduits. The optimized percent laser transmission (OPLT) required for full dynamic range imaging of each site was determined, and changes in OPLT were used to infer changes in collagen and elastin signal intensity. Collagen fiber crimp period and collagen and elastin fiber diameter were measured in leaflet tissue. Statistically significant differences in OPLT and the dimensional characteristics of collagen and elastin in study groups were determined through single factor ANOVA. The majority of donor-aggregated average OPLT observations showed no statistically significant differences among all groups, indicating no difference in collagen or elastin signal strength. Morphometric analysis of collagen and elastin fibers revealed no significant alterations in treated leaflet tissues relative to fresh tissues. Collagen and elastin structural integrity within allograft heart valves are maintained through SynerGraft® decellularization and subsequent cryopreservation.
Collapse
|
35
|
Crawford B, Koshy ST, Jhamb G, Woodford C, Thompson CM, Levy AS, Rush JWE, Guillemette JG, Lillicrap D, Jervis E. Cardiac decellularisation with long-term storage and repopulation with canine peripheral blood progenitor cells. CAN J CHEM ENG 2011. [DOI: 10.1002/cjce.20670] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
36
|
Fritze O, Schleicher M, König K, Schenke-Layland K, Stock U, Harasztosi C. Facilitated noninvasive visualization of collagen and elastin in blood vessels. Tissue Eng Part C Methods 2010; 16:705-10. [PMID: 19803792 DOI: 10.1089/ten.tec.2009.0309] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiphoton imaging is a powerful tool for three-dimensional visualization of extracellular matrix components such as collagen and elastin in fresh, nonfixed, and nonembedded tissues. We have previously published data on the induction of the second harmonic generation signal of collagen and autofluorescence of elastin using a tunable multiphoton laser system. Without staining, a second harmonic generation signal was detected for collagen when excited at wavelength lambda(ex) = 840 nm. Switching the excitation wavelength to 760 nm enabled visualization of elastic fiber structures. A limitation of this technology is the laser-tuning process that requires calibration of the system in between the studies. Now we have developed a facilitated method for studying tissues and tissue equivalents that enables simultaneous visualization of collagen and elastin structures using only a single excitation wavelength of 840 nm in combination with two different band-pass filters. This facilitated method will expand the range of application by reducing required time and expenses for the laser system without reducing its capability.
Collapse
Affiliation(s)
- Olaf Fritze
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen, Tuebingen, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Dickinson LE, Kusuma S, Gerecht S. Reconstructing the differentiation niche of embryonic stem cells using biomaterials. Macromol Biosci 2010; 11:36-49. [PMID: 20967797 DOI: 10.1002/mabi.201000245] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 07/30/2010] [Indexed: 01/14/2023]
Abstract
The biochemical cues and topographical architecture of the extracellular environment extensively influence ES cell fate. The microenvironment surrounding the developing embryo presents these instructive cues in a complex and interactive manner in order to guide cell fate decisions. Current stem cell research aims to reconstruct this multifaceted embryonic niche to recapitulate development in vitro. This review focuses on 2D and 3D differentiation niches created from natural and synthetic biomaterials to guide the differentiation of ES cells toward specific lineages. Biomaterials engineered to present specific physical constraints are also reviewed for their role in differentiation.
Collapse
Affiliation(s)
- Laura E Dickinson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, 3400 North Charles Street, Baltimore, MD 21210, USA
| | | | | |
Collapse
|
38
|
Metzner A, Stock UA, Iino K, Fischer G, Huemme T, Boldt J, Braesen JH, Bein B, Renner J, Cremer J, Lutter G. Percutaneous pulmonary valve replacement: autologous tissue-engineered valved stents. Cardiovasc Res 2010; 88:453-61. [DOI: 10.1093/cvr/cvq212] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
39
|
Wang BG, König K, Halbhuber KJ. Two-photon microscopy of deep intravital tissues and its merits in clinical research. J Microsc 2010; 238:1-20. [PMID: 20384833 DOI: 10.1111/j.1365-2818.2009.03330.x] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiphoton excitation laser scanning microscopy, relying on the simultaneous absorption of two or more photons by a molecule, is one of the most exciting recent developments in biomedical imaging. Thanks to its superior imaging capability of deeper tissue penetration and efficient light detection, this system becomes more and more an inspiring tool for intravital bulk tissue imaging. Two-photon excitation microscopy including 2-photon fluorescence and second harmonic generated signal microscopy is the most common multiphoton microscopic application. In the present review we take diverse ocular tissues as intravital samples to demonstrate the advantages of this approach. Experiments with registration of intracellular 2-photon fluorescence and extracellular collagen second harmonic generated signal microscopy in native ocular tissues are focused. Data show that the in-tandem combination of 2-photon fluorescence and second harmonic generated signal microscopy as two-modality microscopy allows for in situ co-localization imaging of various microstructural components in the whole-mount deep intravital tissues. New applications and recent developments of this high technology in clinical studies such as 2-photon-controlled drug release, in vivo drug screening and administration in skin and kidney, as well as its uses in tumourous tissues such as melanoma and glioma, in diseased lung, brain and heart are additionally reviewed. Intrinsic emission two-modal 2-photon microscopy/tomography, acting as an efficient and sensitive non-injurious imaging approach featured by high contrast and subcellular spatial resolution, has been proved to be a promising tool for intravital deep tissue imaging and clinical studies. Given the level of its performance, we believe that the non-linear optical imaging technique has tremendous potentials to find more applications in biomedical fundamental and clinical research in the near future.
Collapse
Affiliation(s)
- B-G Wang
- Laser Microscopy Research Unit, Institute of Microscopic Anatomy, Faculty of Medicine, Friedrich Schiller University of Jena. Teichgraben 7, 07743 Jena, Germany.
| | | | | |
Collapse
|
40
|
Lutter G, Metzner A, Jahnke T, Bombien R, Boldt J, Iino K, Cremer J, Stock UA. Percutaneous tissue-engineered pulmonary valved stent implantation. Ann Thorac Surg 2010; 89:259-63. [PMID: 20103248 DOI: 10.1016/j.athoracsur.2009.06.048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 06/11/2009] [Accepted: 06/12/2009] [Indexed: 11/19/2022]
Abstract
PURPOSE The purpose of this study was to evaluate the feasibility of percutaneously implanted tissue-engineered valved stents in the ovine pulmonary valve position. DESCRIPTION Porcine pulmonary heart valves and small intestinal submucosa were obtained from a slaughterhouse, and the intestinal submucosa used to cover the inside of the porcine pulmonary valved stents. Endothelial cells and autologous myofibroblasts were obtained from carotid artery segments of juvenile sheep. After myofibroblast seeding, constructs were placed in a dynamic bioreactor system and were cultured for 16 days. After Endothelial cell seeding, the tissue-engineered valved stents were deployed into the pulmonary valve annular site. Angiography was performed at implantation and explantation (4 weeks). Constructs were analyzed macroscopically and microscopically. EVALUATION Orthotopic positioning of the stents (n = 3) at the time of implantation and explantation, as well as normal valve function, was observed through angiography. Gross morphology confirmed excellent opening and closing of all leaflets. Strong expression of alpha-smooth muscle actin in neointerstitial cells and of von-Willebrand-Factor in endothelial cells was revealed by immunocytochemistry. CONCLUSIONS This study demonstrates successful merging of two novel technologies: (1) percutaneous valved stent implantation and (2) tissue engineering of autologous heart valves.
Collapse
Affiliation(s)
- Georg Lutter
- Department of Cardiovascular Surgery, Christian-Albrechts-University of Kiel, School of Medicine, Kiel, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
De Cock LJ, De Koker S, De Vos F, Vervaet C, Remon JP, De Geest BG. Layer-by-Layer Incorporation of Growth Factors in Decellularized Aortic Heart Valve Leaflets. Biomacromolecules 2010; 11:1002-8. [DOI: 10.1021/bm9014649] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Liesbeth J. De Cock
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium, Department of Molecular Biomedical Research, Ghent University, Technologiepark Zwijnaarde 927, 9052 Ghent, Belgium, and Laboratory of Radiopharmacy, Department of Pharmaceutical Analysis, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Stefaan De Koker
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium, Department of Molecular Biomedical Research, Ghent University, Technologiepark Zwijnaarde 927, 9052 Ghent, Belgium, and Laboratory of Radiopharmacy, Department of Pharmaceutical Analysis, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Filip De Vos
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium, Department of Molecular Biomedical Research, Ghent University, Technologiepark Zwijnaarde 927, 9052 Ghent, Belgium, and Laboratory of Radiopharmacy, Department of Pharmaceutical Analysis, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Chris Vervaet
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium, Department of Molecular Biomedical Research, Ghent University, Technologiepark Zwijnaarde 927, 9052 Ghent, Belgium, and Laboratory of Radiopharmacy, Department of Pharmaceutical Analysis, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Jean-Paul Remon
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium, Department of Molecular Biomedical Research, Ghent University, Technologiepark Zwijnaarde 927, 9052 Ghent, Belgium, and Laboratory of Radiopharmacy, Department of Pharmaceutical Analysis, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - Bruno G. De Geest
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium, Department of Molecular Biomedical Research, Ghent University, Technologiepark Zwijnaarde 927, 9052 Ghent, Belgium, and Laboratory of Radiopharmacy, Department of Pharmaceutical Analysis, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| |
Collapse
|
42
|
Zhou J, Fritze O, Schleicher M, Wendel HP, Schenke-Layland K, Harasztosi C, Hu S, Stock UA. Impact of heart valve decellularization on 3-D ultrastructure, immunogenicity and thrombogenicity. Biomaterials 2010; 31:2549-54. [PMID: 20061016 DOI: 10.1016/j.biomaterials.2009.11.088] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 11/25/2009] [Indexed: 10/20/2022]
Abstract
Decellularized xenogeneic tissue represents an interesting material for heart valve tissue engineering. The prospect objective is removal of all viable cells while preserving extracellular matrix (ECM) integrity. The major concerns of all decellularization protocols remain ECM disruption, immunogenicity and thrombogenicity. Accordingly the aim of this study was visualization of ultrastructural ECM disruption and human immune response and thrombogenicity using different decellularization protocols of porcine heart valves. Porcine pulmonary leaflets were decellularized with four different protocols: sodium deoxycholate, sodium dedecylsulfate, trypsin/EDTA, and trypsin-detergent-nuclease. Then the tissues were processed for histology and two-photon laser scanning microscopy (LSM). For thrombogenicity and immunogenicity testing tissues were incubated with human blood. The histological examination revealed no remaining cells and no significant differences in the ECM histoarchitecture in any group. LSM detected significant ECM alterations in all groups except sodium deoxycholate group with an almost completely preserved ECM. There was no increased immunogenicity between fresh and decellularized tissue. Compared to GA-fixed tissue however significantly increased immune responses and thrombogenicity was observed in all protocols. From our experiment, sodium deoxycholate enables cell removal with almost complete preservation of ECM structures. And all of these four decellularization protocols affected human immunological response and increased thrombogenicity.
Collapse
Affiliation(s)
- Jianye Zhou
- Thoracic, Cardiac and Vascular Surgery, University Hospital Tübingen, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
43
|
On the multiscale modeling of heart valve biomechanics in health and disease. Biomech Model Mechanobiol 2010; 9:373-87. [PMID: 20066464 DOI: 10.1007/s10237-009-0181-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 11/26/2009] [Indexed: 01/10/2023]
Abstract
Theoretical models of the human heart valves are useful tools for understanding and characterizing the dynamics of healthy and diseased valves. Enabled by advances in numerical modeling and in a range of disciplines within experimental biomechanics, recent models of the heart valves have become increasingly comprehensive and accurate. In this paper, we first review the fundamentals of native heart valve physiology, composition and mechanics in health and disease. We will then furnish an overview of the development of theoretical and experimental methods in modeling heart valve biomechanics over the past three decades. Next, we will emphasize the necessity of using multiscale modeling approaches in order to provide a comprehensive description of heart valve biomechanics able to capture general heart valve behavior. Finally, we will offer an outlook for the future of valve multiscale modeling, the potential directions for further developments and the challenges involved.
Collapse
|
44
|
Schenke-Layland K, Xie J, Magnusson M, Angelis E, Li X, Wu K, Reinhardt DP, Maclellan WR, Hamm-Alvarez SF. Lymphocytic infiltration leads to degradation of lacrimal gland extracellular matrix structures in NOD mice exhibiting a Sjögren's syndrome-like exocrinopathy. Exp Eye Res 2009; 90:223-37. [PMID: 19852957 DOI: 10.1016/j.exer.2009.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 09/30/2009] [Accepted: 10/14/2009] [Indexed: 01/29/2023]
Abstract
We previously reported that lacrimal glands (LGs) of male non-obese diabetic (NOD) mice, an established mouse model of autoimmune inflammatory LG disease that displays many features of human LGs in patients afflicted with Sjögren's syndrome (SjS), exhibit significant degradation of extracellular matrix (ECM) structures as well as increased expression of matrix metalloproteinases (MMPs). The purpose of the current study was to expand the spectrum of proteases identified, to clarify their probable origin as well as to identify the contribution of these changes to disease pathogenesis. We explored in depth the changes in ECM structures and ECM protease expression at the onset of disease (6 weeks) versus late stage disease (18 weeks) in male NOD mouse LGs, relative to LGs of age-matched male NODscid, a severely immunocompromised congenic strain, and healthy BALB/c mice. LG tissues were examined using routine histological, immunohistochemical, Western Blot and gene expression analyses novel multiphoton imaging technologies. We further characterized the profile of infiltrating immune cells under each condition using flow cytometry. Our results show that the initial infiltrating cells at 6 weeks of age are responsible for increased MMP and cathepsin H expression and therefore initiate the LG ECM degradation in NOD mice. More importantly, NODscid mice exhibited normal LG ECM structures, indicating the lymphocytes seen in the LGs of NOD mice are responsible for the degradation of the LG ECM. The disease-related remodeling of LG ECM structures may play a crucial role in altering the acinar signaling environment, disrupting the signaling scaffolds within the cells, which are required to mobilize the exocytotic trafficking machinery, ultimately leading to a loss of LG function in patients afflicted with SjS.
Collapse
Affiliation(s)
- Katja Schenke-Layland
- Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1760, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mansfield J, Yu J, Attenburrow D, Moger J, Tirlapur U, Urban J, Cui Z, Winlove P. The elastin network: its relationship with collagen and cells in articular cartilage as visualized by multiphoton microscopy. J Anat 2009; 215:682-91. [PMID: 19796069 DOI: 10.1111/j.1469-7580.2009.01149.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
A combination of two-photon fluorescence (TPF), second harmonic generation (SHG) and coherent anti-Stokes Raman scattering (CARS) imaging has been used to investigate the elastin fibre network in healthy equine articular cartilage from the metacarpophalangeal joint. The elastin fibres were identified using their intrinsic two-photon fluorescence and immuno-staining was used to confirm the identity of these fibres. SHG was used to reveal the collagen matrix and the collagen fibre orientations were determined from their SHG polarization sensitivity, while CARS was used to clearly delineate the cell boundaries. Extensive elastin fibre networks were found in all the joint regions investigated. The elastin was found predominantly in the superficial zone (upper 50 microm) and was aligned parallel to the articular surface. Elastin was also detected in the pericellular matrix surrounding the superficial chondrocytes; however, individual fibres could not be resolved in this region. Variations in the density and organization of the fibres were observed in different regions on the joint surface.
Collapse
Affiliation(s)
- Jessica Mansfield
- Biophysics, School of Physics, University of Exeter, Exeter EX4 4QL, UK.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Schenke-Layland K, Stock UA, Nsair A, Xie J, Angelis E, Fonseca CG, Larbig R, Mahajan A, Shivkumar K, Fishbein MC, MacLellan WR. Cardiomyopathy is associated with structural remodelling of heart valve extracellular matrix. Eur Heart J 2009; 30:2254-65. [PMID: 19561339 DOI: 10.1093/eurheartj/ehp267] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS To increase the supply, many countries harvest allograft valves from explanted hearts of transplant recipients with ischaemic (ICM) or dilated cardiomyopathy (DCM). This study determines the structural integrity of valves from cardiomyopathic hearts. METHODS AND RESULTS Extracellular matrix (ECM) was examined in human valves obtained from normal, ICM, and DCM hearts. To confirm if ECM changes were directly related to the cardiomyopathy, we developed a porcine model of chronic ICM. Histology and immunohistostaining, as well as non-invasive multiphoton and second harmonic generation (SHG) imaging revealed marked disruption of ECM structures in human valves from ICM and DCM hearts. The ECM was unaffected in valves from normal and acute ICM pigs, whereas chronic ICM specimens showed ECM alterations similar to those seen in ICM and DCM patients. Proteins and proteinases implicated in ECM remodelling, including Tenascin C, TGFbeta1, Cathepsin B, MMP2, were upregulated in human ICM and DCM, and porcine chronic ICM specimens. CONCLUSION Valves from cardiomyopathic hearts showed significant ECM deterioration with a disrupted collagen and elastic fibre network. It will be important to determine the impact of this ECM damage on valve durability and calcification in vivo if allografts are to be used from these donors.
Collapse
Affiliation(s)
- Katja Schenke-Layland
- Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Schenke-Layland K, Rofail F, Heydarkhan S, Gluck JM, Ingle NP, Angelis E, Choi CH, MacLellan WR, Beygui RE, Shemin RJ, Heydarkhan-Hagvall S. The use of three-dimensional nanostructures to instruct cells to produce extracellular matrix for regenerative medicine strategies. Biomaterials 2009; 30:4665-75. [PMID: 19524289 DOI: 10.1016/j.biomaterials.2009.05.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 05/17/2009] [Indexed: 11/27/2022]
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.
Collapse
Affiliation(s)
- Katja Schenke-Layland
- Cardiovascular Research Laboratories, Department of Medicine/Cardiology, David Geffen School of Medicine, 675 Charles E Young Dr. South, MRL-3579, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Iop L, Renier V, Naso F, Piccoli M, Bonetti A, Gandaglia A, Pozzobon M, Paolin A, Ortolani F, Marchini M, Spina M, De Coppi P, Sartore S, Gerosa G. The influence of heart valve leaflet matrix characteristics on the interaction between human mesenchymal stem cells and decellularized scaffolds. Biomaterials 2009; 30:4104-16. [PMID: 19481252 DOI: 10.1016/j.biomaterials.2009.04.031] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 04/21/2009] [Indexed: 10/20/2022]
Abstract
The potential for in vitro colonization of decellularized valves by human bone marrow mesenchymal stem cells (hBM-MSCs) towards the anisotropic layers ventricularis and fibrosa and in homo- vs. heterotypic cell-ECM interactions has never been investigated. hBM-MSCs were expanded and characterized by immunofluorescence and FACS analysis. Porcine and human pulmonary valve leaflets (p- and hPVLs, respectively) underwent decellularization with Triton X100-sodium cholate treatment (TRICOL), followed by nuclear fragment removal. hBM-MSCs (2x10(6) cells/cm(2)) were seeded onto fibrosa (FS) or ventricularis (VS) of decellularized PVLs, precoated with FBS and fibronectin, and statically cultured for 30 days. Bioengineered PVLs revealed no histopathological features but a reconstructed endothelium lining and the presence of fibroblasts, myofibroblasts and SMCs, as in the corresponding native leaflet. The two valve layers behaved differently as regards hBM-MSC repopulation potential, however, with a higher degree of 3D spreading and differentiation in VS than in FS samples, and with enhanced cell survival and colonization effects in the homotypic ventricularis matrix, suggesting that hBM-MSC phenotypic conversion is strongly influenced in vitro by the anisotropic valve microstructure and species-specific matching between extracellular matrix and donor cells. These findings are of particular relevance to in vivo future applications of valve tissue engineering.
Collapse
Affiliation(s)
- Laura Iop
- Department of Cardiologic, Thoracic and Vascular Sciences, School of Medicine, University of Padua, Via Giustiniani, 2 - I-35128 Padua, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Barth M, Schumacher H, Kuhn C, Akhyari P, Lichtenberg A, Franke WW. Cordial connections: molecular ensembles and structures of adhering junctions connecting interstitial cells of cardiac valves in situ and in cell culture. Cell Tissue Res 2009; 337:63-77. [PMID: 19475424 DOI: 10.1007/s00441-009-0806-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Accepted: 04/06/2009] [Indexed: 01/19/2023]
Abstract
Remarkable efforts have recently been made in the tissue engineering of heart valves to improve the results of valve transplantations and replacements, including the design of artificial valves. However, knowledge of the cell and molecular biology of valves and, specifically, of valvular interstitial cells (VICs) remains limited. Therefore, our aim has been to determine and localize the molecules forming the adhering junctions (AJs) that connect VICs in situ and in cell culture. Using biochemical and immunolocalization methods at the light- and electron-microscopic levels, we have identified, in man, cow, sheep and rat, the components of VIC-connecting AJs in situ and in cell culture. These AJs contain, in addition to the transmembrane glycoproteins N-cadherin and cadherin-11, the typical plaque proteins alpha- and beta-catenin as well as plakoglobin and p120, together with minor amounts of protein p0071, i.e. a total of five plaque proteins of the armadillo family. While we can exclude the occurrence of desmogleins, desmocollins and desmoplakin, we have noted with surprise that AJs of VICs in cell cultures, but not those growing in the valve tissue, contain substantial amounts of the desmosomal plaque protein, plakophilin-2. Clusters of AJs occur not only on the main VIC cell bodies but are also found widely dispersed on their long filopodia thus forming, in the tissue, a meshwork that, together with filopodial attachments to paracrystalline collagen fiber bundles, establishes a three-dimensional suprastructure, the role of which is discussed with respect to valve formation, regeneration and function.
Collapse
Affiliation(s)
- Mareike Barth
- Helmholtz Group/Cell Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Schenke-Layland K. Non-invasive multiphoton imaging of extracellular matrix structures. JOURNAL OF BIOPHOTONICS 2008; 1:451-62. [PMID: 19343671 PMCID: PMC4350994 DOI: 10.1002/jbio.200810045] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Multiphoton microscopy has become a powerful method for the artifact-free, nondestructive evaluation of deep-tissue cells and extracellular matrix (ECM) structures in their native environment. By interacting with highly non-centrosymmetric molecular assemblies such as fibrillar collagen, the non-linear process called second harmonic generation (SHG) has also proven to be an important diagnostic tool for the visualization of ECM compartments in situ with submicron resolution without the need for tissue processing. This review reports on applications of multiphoton-induced autofluorescence and SHG microscopy to identify collagen and elastic fiber orientation in native, tissue-engineered and processed, as well as healthy and diseased, tissues and organs. SHG signal profiling was used to quantify ECM damage in various cardiovascular and exocrine tissues, as well as cartilage. These novel imaging modalities open the general possibility of high-resolution in situ and more important in vivo imaging of ECM structures, cells and intracellular organelles in living intact tissues.
Collapse
Affiliation(s)
- Katja Schenke-Layland
- Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California, Los Angeles, UCLA, 675 Charles E. Young Drive South, MRL 3-579, Los Angeles, CA 90095, USA.
| |
Collapse
|