36
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Engineering a 3D-Bioprinted Model of Human Heart Valve Disease Using Nanoindentation-Based Biomechanics

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          In calcific aortic valve disease (CAVD), microcalcifications originating from nanoscale calcifying vesicles disrupt the aortic valve (AV) leaflets, which consist of three (biomechanically) distinct layers: the fibrosa, spongiosa, and ventricularis. CAVD has no pharmacotherapy and lacks in vitro models as a result of complex valvular biomechanical features surrounding resident mechanosensitive valvular interstitial cells (VICs). We measured layer-specific mechanical properties of the human AV and engineered a three-dimensional (3D)-bioprinted CAVD model that recapitulates leaflet layer biomechanics for the first time. Human AV leaflet layers were separated by microdissection, and nanoindentation determined layer-specific Young’s moduli. Methacrylated gelatin (GelMA)/methacrylated hyaluronic acid (HAMA) hydrogels were tuned to duplicate layer-specific mechanical characteristics, followed by 3D-printing with encapsulated human VICs. Hydrogels were exposed to osteogenic media (OM) to induce microcalcification, and VIC pathogenesis was assessed by near infrared or immunofluorescence microscopy. Median Young’s moduli of the AV layers were 37.1, 15.4, and 26.9 kPa (fibrosa/spongiosa/ventricularis, respectively). The fibrosa and spongiosa Young’s moduli matched the 3D 5% GelMa/1% HAMA UV-crosslinked hydrogels. OM stimulation of VIC-laden bioprinted hydrogels induced microcalcification without apoptosis. We report the first layer-specific measurements of human AV moduli and a novel 3D-bioprinted CAVD model that potentiates microcalcification by mimicking the native AV mechanical environment. This work sheds light on valvular mechanobiology and could facilitate high-throughput drug-screening in CAVD.

          Related collections

          Most cited references44

          • Record: found
          • Abstract: found
          • Article: not found

          Characterization of the early lesion of 'degenerative' valvular aortic stenosis. Histological and immunohistochemical studies.

          Nonrheumatic stenosis of trileaflet aortic valves, often termed senile or calcific valvular aortic stenosis, is considered a "degenerative" process, but little is known about the cellular or molecular factors that mediate its development. To characterize the developing aortic valvular lesion, we performed histological and immunohistochemical studies on Formalin-fixed and methanol-Carnoy's-fixed paraffin-embedded aortic valve leaflets or on frozen sections obtained at autopsy from 27 adults (age, 46 to 82 years) with normal leaflets (n = 6), mild macroscopic leaflet thickening (n = 15), or clinical aortic stenosis (n = 6). Focal areas of thickening ("early lesions") were characterized by (1) subendothelial thickening on the aortic side of the leaflet, between the basement membrane (PAS-positive) and elastic lamina (Verhoeff-van Gieson), (2) the presence of large amounts of intracellular and extracellular neutral lipids (oil red O) and fine, stippled mineralization (von Kossa), and (3) disruption of the basement membrane overlying the lesion. Regions of the fibrosa adjacent to these lesions were characterized by thickening and by protein, lipid, and calcium accumulation. Control valves showed none of these abnormalities. Immunohistochemical studies were performed using monoclonal antibodies directed against macrophages (anti-CD68 or HAM-56), and contractile proteins of smooth muscle cells or myofibroblasts (anti-alpha-actin and HHF-35) or rabbit polyclonal antiserum against T lymphocytes (anti-CD3). In normal valves, scattered macrophages were present in the fibrosa and ventricularis, and occasional muscle actin-positive cells were detected in the proximal portion of the ventricularis near the leaflet base, but no T lymphocytes were found. In contrast, early lesions were characterized by the presence of an inflammatory infiltrate composed of non-foam cell and foam cell macrophages, occasional T cells, and rare alpha-actin-positive cells. In stenotic aortic valves, a similar but more advanced lesion was seen. The early lesion of "degenerative" aortic stenosis is an active inflammatory process with some similarities (lipid deposition, macrophage and T-cell infiltration, and basement membrane disruption) and some dissimilarities (presence of prominent mineralization and small numbers of smooth muscle cells) to atherosclerosis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Controlled degradation and mechanical behavior of photopolymerized hyaluronic acid networks.

            Hyaluronic acid is a natural polysaccharide found abundantly throughout the body with many desirable properties for application as a biomaterial, including scaffolding for tissue engineering. In this work, hyaluronic acid with molecular weights ranging from 50 to 1100 kDa was modified with methacrylic anhydride and photopolymerized into networks with a wide range of physical properties. With macromer concentrations from 2 to 20 wt %, networks exhibited volumetric swelling ratios ranging from approximately 42 to 8, compressive moduli ranging from approximately 2 to over 100 kPa, and degradation times ranging from less than 1 day up to almost 38 days in the presence of 100 U/mL of hyaluronidase. When 3T3-fibroblasts were photoencapsulated in the hydrogels, cells remained viable with low macromer concentrations but decreased sequentially as the macromer concentration increased. Finally, auricular swine chondrocytes produced neocartilage when photoencapsulated in the hyaluronic acid networks. This work presents a next step toward the development of advanced in vivo curable biomaterials.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The bio in the ink: cartilage regeneration with bioprintable hydrogels and articular cartilage-derived progenitor cells.

              Cell-laden hydrogels are the primary building blocks for bioprinting, and, also termed bioinks, are the foundations for creating structures that can potentially recapitulate the architecture of articular cartilage. To be functional, hydrogel constructs need to unlock the regenerative capacity of encapsulated cells. The recent identification of multipotent articular cartilage-resident chondroprogenitor cells (ACPCs), which share important traits with adult stem cells, represents a new opportunity for cartilage regeneration. However, little is known about the suitability of ACPCs for tissue engineering, especially in combination with biomaterials. This study aimed to investigate the potential of ACPCs in hydrogels for cartilage regeneration and biofabrication, and to evaluate their ability for zone-specific matrix production. Gelatin methacryloyl (gelMA)-based hydrogels were used to culture ACPCs, bone marrow mesenchymal stromal cells (MSCs) and chondrocytes, and as bioinks for printing. Our data shows ACPCs outperformed chondrocytes in terms of neo-cartilage production and unlike MSCs, ACPCs had the lowest gene expression levels of hypertrophy marker collagen type X, and the highest expression of PRG4, a key factor in joint lubrication. Co-cultures of the cell types in multi-compartment hydrogels allowed generating constructs with a layered distribution of collagens and glycosaminoglycans. By combining ACPC- and MSC-laden bioinks, a bioprinted model of articular cartilage was generated, consisting of defined superficial and deep regions, each with distinct cellular and extracellular matrix composition. Taken together, these results provide important information for the use of ACPC-laden hydrogels in regenerative medicine, and pave the way to the biofabrication of 3D constructs with multiple cell types for cartilage regeneration or in vitro tissue models.
                Bookmark

                Author and article information

                Journal
                Nanomaterials (Basel)
                Nanomaterials (Basel)
                nanomaterials
                Nanomaterials
                MDPI
                2079-4991
                03 May 2018
                May 2018
                : 8
                : 5
                : 296
                Affiliations
                [1 ]Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; dewyvdvalk@ 123456gmail.com (D.C.v.d.V.); ponnie.wu@ 123456gmail.com (P.-J.W.); lhlee@ 123456bwh.harvard.edu (L.H.L.); jwen4@ 123456bwh.harvard.edu (J.R.W.); ahha@ 123456bwh.harvard.edu (A.H.H.); fbuffolo@ 123456bwh.harvard.edu (F.B.); maikawa@ 123456bwh.harvard.edu (M.A.)
                [2 ]Center of Excellence in Cardiovascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA 02115, USA; c.f.t.vanderven@ 123456umcutrecht.nl
                [3 ]David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; osfenton@ 123456mit.edu (O.S.F.); mtibbitt@ 123456ethz.ch (M.W.T.); jandrese@ 123456mit.edu (J.L.A.); rlanger@ 123456mit.edu (R.L.)
                [4 ]Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA; grolman@ 123456fas.harvard.edu (J.M.G.); mooneyd@ 123456seas.harvard.edu (D.J.M.)
                [5 ]John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
                [6 ]Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zürich, Switzerland
                [7 ]Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, the Netherlands; a.vanmil@ 123456umcutrecht.nl (A.v.M.); j.sluijter@ 123456umcutrecht.nl (J.P.G.S.); jhjortna@ 123456umcutrecht.nl (J.H.)
                [8 ]Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
                [9 ]Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands; c.v.c.bouten@ 123456tue.nl
                [10 ]Center for Perioperative Genomics, Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; sbody@ 123456bwh.harvard.edu
                [11 ]Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
                [12 ]Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
                Author notes
                [* ]Correspondence: mblaser@ 123456bwh.harvard.edu (M.C.B.); eaikawa@ 123456bwh.harvard.edu (E.A.); Tel.: +617-730-7755 (M.C.B.); Tel.: +617-730-7755 (E.A.)
                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0003-1035-5094
                https://orcid.org/0000-0003-2088-9102
                Article
                nanomaterials-08-00296
                10.3390/nano8050296
                5977310
                29751516
                2f27a751-cfec-48e5-9f9d-891112cd09e0
                © 2018 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 02 April 2018
                : 24 April 2018
                Categories
                Article

                aortic valve,calcific aortic valve disease,calcification,mechanobiology,bioprinting,3d printing,microdissection,nanoindentation

                Comments

                Comment on this article