1
|
Wu J, Li J, Mao S, Li B, Zhu L, Jia P, Huang G, Yang X, Xu L, Qiu D, Wang S, Dong Y. Heparin-Functionalized Bioactive Glass to Harvest Endogenous Growth Factors for Pulp Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30715-30727. [PMID: 38833722 DOI: 10.1021/acsami.4c03118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Pulp and periapical diseases can lead to the cessation of tooth development, resulting in compromised tooth structure and functions. Despite numerous efforts to induce pulp regeneration, effective strategies are still lacking. Growth factors (GFs) hold considerable promise in pulp regeneration due to their diverse cellular regulatory properties. However, the limited half-lives and susceptibility to degradation of exogenous GFs necessitate the administration of supra-physiological doses, leading to undesirable side effects. In this research, a heparin-functionalized bioactive glass (CaO-P2O5-SiO2-Heparin, abbreviated as PSC-Heparin) with strong bioactivity and a stable neutral pH is developed as a promising candidate to addressing challenges in pulp regeneration. Fourier transform infrared spectroscopy, X-ray photoelectron spectroscopy, and thermogravimetric analysis reveal the successful synthesis of PSC-Heparin. Scanning electron microscopy and X-ray diffraction show the hydroxyapatite formation can be observed on the surface of PSC-Heparin after soaking in simulated body fluid for 12 h. PSC-Heparin is capable of harvesting various endogenous GFs and sustainably releasing them over an extended duration by the enzyme-linked immunosorbent assay. Cytological experiments show that developed PSC-Heparin can facilitate the adhesion, migration, proliferation, and odontogenic differentiation of stem cells from apical papillae. Notably, the histological analysis of subcutaneous implantation in nude mice demonstrates PSC-Heparin is capable of promoting the odontoblast-like layers and pulp-dentin complex formation without the addition of exogenous GFs, which is vital for clinical applications. This work highlights an effective strategy of harvesting endogenous GFs and avoiding the involvement of exogenous GFs to achieve pulp-dentin complex regeneration, which may open a new horizon for regenerative endodontic therapy.
Collapse
Affiliation(s)
- Jilin Wu
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Jingyi Li
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Sicong Mao
- Department of General Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Baokui Li
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 10090, China
| | - Lin Zhu
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Peipei Jia
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Guibin Huang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Xule Yang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Liju Xu
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 10090, China
| | - Dong Qiu
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 10090, China
| | - Sainan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Yanmei Dong
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| |
Collapse
|
2
|
Xu KL, Mauck RL, Burdick JA. Modeling development using hydrogels. Development 2023; 150:dev201527. [PMID: 37387575 PMCID: PMC10323241 DOI: 10.1242/dev.201527] [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] [Indexed: 07/01/2023]
Abstract
The development of multicellular complex organisms relies on coordinated signaling from the microenvironment, including both biochemical and mechanical interactions. To better understand developmental biology, increasingly sophisticated in vitro systems are needed to mimic these complex extracellular features. In this Primer, we explore how engineered hydrogels can serve as in vitro culture platforms to present such signals in a controlled manner and include examples of how they have been used to advance our understanding of developmental biology.
Collapse
Affiliation(s)
- Karen L. Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
3
|
Wang D, Zhang X, Ng KW, Rao Y, Wang C, Gharaibeh B, Lin S, Abrams G, Safran M, Cheung E, Campbell P, Weiss L, Ker DFE, Yang YP. Growth and differentiation factor-7 immobilized, mechanically strong quadrol-hexamethylene diisocyanate-methacrylic anhydride polyurethane polymer for tendon repair and regeneration. Acta Biomater 2022; 154:108-122. [PMID: 36272687 DOI: 10.1016/j.actbio.2022.10.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022]
Abstract
Biological and mechanical cues are both vital for biomaterial aided tendon repair and regeneration. Here, we fabricated mechanically tendon-like (0 s UV) QHM polyurethane scaffolds (Q: Quadrol, H: Hexamethylene diisocyanate; M: Methacrylic anhydride) and immobilized them with Growth and differentiation factor-7 (GDF-7) to produce mechanically strong and tenogenic scaffolds. In this study, we assessed QHM polymer cytocompatibility, amenability to fibrin-coating, immobilization and persistence of GDF-7, and capability to support GDF-7-mediated tendon differentiation in vitro as well as in vivo in mouse subcutaneous and acute rat rotator cuff tendon resection models. Cytocompatibility studies showed that QHM facilitated cell attachment, proliferation, and viability. Fibrin-coating and GDF-7 retention studies showed that mechanically tendon-like 0 s UV QHM polymer could be immobilized with GDF-7 and retained the growth factor (GF) for at least 1-week ex vivo. In vitro differentiation studies showed that GDF-7 mediated bone marrow-derived human mesenchymal stem cell (hMSC) tendon-like differentiation on 0 s UV QHM. Subcutaneous implantation of GDF-7-immobilized, fibrin-coated, QHM polymer in mice for 2 weeks demonstrated de novo formation of tendon-like tissue while implantation of GDF-7-immobilized, fibrin-coated, QHM polymer in a rat acute rotator cuff resection injury model indicated tendon-like tissue formation in situ and the absence of heterotopic ossification. Together, our work demonstrates a promising synthetic scaffold with human tendon-like biomechanical attributes as well as immobilized tenogenic GDF-7 for tendon repair and regeneration. STATEMENT OF SIGNIFICANCE: Biological activity and mechanical robustness are key features required for tendon-promoting biomaterials. While synthetic biomaterials can be mechanically robust, they often lack bioactivity. To biologically augment synthetic biomaterials, numerous drug and GF delivery strategies exist but the large tissue space within the shoulder is constantly flushed with saline during arthroscopic surgery, hindering efficacious controlled release of therapeutic molecules. Here, we coated QHM polymer (which exhibits human tendon-to-bone-like biomechanical attributes) with fibrin for GF binding. Unlike conventional drug delivery strategies, our approach utilizes immobilized GFs as opposed to released GFs for sustained, localized tissue regeneration. Our data demonstrated that GF immobilization can be broadly applied to synthetic biomaterials for enhancing bioactivity, and GDF-7-immobilized QHM exhibit high clinical translational potential for tendon repair.
Collapse
Affiliation(s)
- Dan Wang
- Department of Orthopaedic Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromuscular Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka Wai Ng
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ying Rao
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chenyang Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Burhan Gharaibeh
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Sien Lin
- Department of Orthopaedic Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA; Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Geoffrey Abrams
- Department of Orthopaedic Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA
| | - Marc Safran
- Department of Orthopaedic Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA
| | - Emilie Cheung
- Department of Orthopaedic Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA
| | - Phil Campbell
- Engineering Research Accelerator, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213, USA; Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213, USA; Robotics Institute, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Lee Weiss
- Robotics Institute, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213, USA; Engineering Research Accelerator, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213, USA; Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Dai Fei Elmer Ker
- Department of Orthopaedic Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromuscular Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China.
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA; Department of Material Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Neubauer VJ, Hüter F, Wittmann J, Trossmann VT, Kleinschrodt C, Alber-Laukant B, Rieg F, Scheibel T. Flow Simulation and Gradient Printing of Fluorapatite- and Cell-Loaded Recombinant Spider Silk Hydrogels. Biomolecules 2022; 12:biom12101413. [PMID: 36291622 PMCID: PMC9599405 DOI: 10.3390/biom12101413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Hierarchical structures are abundant in almost all tissues of the human body. Therefore, it is highly important for tissue engineering approaches to mimic such structures if a gain of function of the new tissue is intended. Here, the hierarchical structures of the so-called enthesis, a gradient tissue located between tendon and bone, were in focus. Bridging the mechanical properties from soft to hard secures a perfect force transmission from the muscle to the skeleton upon locomotion. This study aimed at a novel method of bioprinting to generate gradient biomaterial constructs with a focus on the evaluation of the gradient printing process. First, a numerical approach was used to simulate gradient formation by computational flow as a prerequisite for experimental bioprinting of gradients. Then, hydrogels were printed in a single cartridge printing set-up to transfer the findings to biomedically relevant materials. First, composites of recombinant spider silk hydrogels with fluorapatite rods were used to generate mineralized gradients. Then, fibroblasts were encapsulated in the recombinant spider silk-fluorapatite hydrogels and gradually printed using unloaded spider silk hydrogels as the second component. Thereby, adjustable gradient features were achieved, and multimaterial constructs were generated. The process is suitable for the generation of gradient materials, e.g., for tissue engineering applications such as at the tendon/bone interface.
Collapse
Affiliation(s)
- Vanessa J. Neubauer
- Lehrstuhl Biomaterialien, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany
| | - Florian Hüter
- Lehrstuhl Konstruktionslehre und CAD, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Johannes Wittmann
- Lehrstuhl Konstruktionslehre und CAD, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Vanessa T. Trossmann
- Lehrstuhl Biomaterialien, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany
| | - Claudia Kleinschrodt
- Lehrstuhl Konstruktionslehre und CAD, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Bettina Alber-Laukant
- Lehrstuhl Konstruktionslehre und CAD, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Frank Rieg
- Lehrstuhl Konstruktionslehre und CAD, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuth Engine Research Center (BERC), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Zentrum für Energietechnik (ZET), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Thomas Scheibel
- Lehrstuhl Biomaterialien, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayerisches Polymerinstitut (BPI), Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Materialzentrum (BayMAT), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Correspondence:
| |
Collapse
|
5
|
Uehlin AF, Vines JB, Feldman DS, Nyairo E, Dean DR, Thomas V. Uni-Directionally Oriented Fibro-Porous PLLA/Fibrin Bio-Hybrid Scaffold: Mechano-Morphological and Cell Studies. Pharmaceutics 2022; 14:pharmaceutics14020277. [PMID: 35214010 PMCID: PMC8879164 DOI: 10.3390/pharmaceutics14020277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we report a biohybrid oriented fibrous scaffold based on nanofibers of poly(l-lactic acid) (PLLA)/fibrin produced by electrospinning and subsequent post-treatment. Induced hydrolytic degradation of the fibers in 0.25 M NaOH solution for various time periods followed by the immobilization of fibrin on the hydrolyzed fiber surfaces was shown to significantly affect the mechanical properties, with the tensile strength (40.6 MPa ± 1.3) and strain at failure (38% ± 4.5) attaining a value within the range of human ligaments and ligament-replacement grafts. Unidirectional electrospinning with a mandrel rotational velocity of 26.4 m/s produced highly aligned fibers with an average diameter of 760 ± 96 nm. After a 20-min hydrolysis treatment in NaOH solution, this was further reduced to an average of 457 ± 89 nm, which is within the range of collagen bundles found in ligament tissue. Based on the results presented herein, the authors hypothesize that a combination of fiber orientation/alignment and immobilization of fibrin can result in the mechanical and morphological modification of PLLA tissue scaffolds for ligament-replacement grafts. Further, it was found that treatment with NaOH enhanced the osteogenic differentiation of hMSCs and the additional inclusion of fibrin further enhanced osteogenic differentiation, as demonstrated by decreased proliferative rates and increased ALP activity.
Collapse
Affiliation(s)
- Andrew F. Uehlin
- Department of Materials Science and Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA;
| | - Jeremy B. Vines
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (J.B.V.); (D.S.F.)
| | - Dale S. Feldman
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (J.B.V.); (D.S.F.)
| | - Elijah Nyairo
- Biomedical Engineering, Alabama State University, Montgomery, AL 36101, USA;
| | - Derrick R. Dean
- Biomedical Engineering, Alabama State University, Montgomery, AL 36101, USA;
- Correspondence: (D.R.D.); (V.T.)
| | - Vinoy Thomas
- Department of Materials Science and Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA;
- Correspondence: (D.R.D.); (V.T.)
| |
Collapse
|
6
|
Yerneni SS, Adamik J, Weiss LE, Campbell PG. Cell trafficking and regulation of osteoblastogenesis by extracellular vesicle associated bone morphogenetic protein 2. J Extracell Vesicles 2021; 10:e12155. [PMID: 34669267 PMCID: PMC8528095 DOI: 10.1002/jev2.12155] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) are characterized by complex cargo composition and carry a wide array of signalling cargo, including growth factors (GFs). Beyond surface-associated GFs, it is unclear if EV intralumenal growth factors are biologically active. Here, bone morphogenetic protein-2 (BMP2), loaded directly into the lumen of EVs designated engineered BMP2-EVs (eBMP2-EVs), was comprehensively characterized including its regulation of osteoblastogenesis. eBMP2-EVs and non-EV 'free' BMP2 were observed to similarly regulate osteoblastogenesis. Furthermore, cell trafficking experiments suggest rapid BMP2 recycling and its extracellular release as 'free' BMP2 and natural occurring BMP2-EVs (nBMP2-EVs), with both being osteogenic. Interestingly, BMP2 occurs on the EV surface of nBMP2-EVs and is susceptible to proteolysis, inhibition by noggin and complete dissociation from nBMP2-EVs over 3 days. Whereas, within the eBMP2-EVs, BMP2 is protected from proteolysis, inhibition by noggin and is retained in EV lumen at 100% for the first 24 h and ∼80% after 10 days. Similar to 'free' BMP2, bioprinted eBMP2-EV microenvironments induced osteogenesis in vitro and in vivo in spatial registration to the printed patterns. Taken together, BMP2 signalling involves dynamic BMP2 cell trafficking in and out of the cell involving EVs, with distinct differences between these nBMP2-EVs and eBMP2-EVs attributable to the BMP2 cargo location with EVs. Lastly, eBMP2-EVs appear to deliver BMP2 directly into the cytoplasm, initiating BMP2 signalling within the cell, bypassing its cell surface receptors.
Collapse
Affiliation(s)
| | - Juraj Adamik
- Division of Hematology/Oncology, Department of MedicineUPMC Hillman Cancer CenterPittsburghPennsylvaniaUSA
| | - Lee E. Weiss
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- The Robotics InstituteCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- The McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Phil G. Campbell
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- The McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Engineering Research Accelerator, College of EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
7
|
Wang D, Zhang X, Huang S, Liu Y, Fu BSC, Mak KKL, Blocki AM, Yung PSH, Tuan RS, Ker DFE. Engineering multi-tissue units for regenerative Medicine: Bone-tendon-muscle units of the rotator cuff. Biomaterials 2021; 272:120789. [PMID: 33845368 DOI: 10.1016/j.biomaterials.2021.120789] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Our body systems are comprised of numerous multi-tissue units. For the musculoskeletal system, one of the predominant functional units is comprised of bone, tendon/ligament, and muscle tissues working in tandem to facilitate locomotion. To successfully treat musculoskeletal injuries and diseases, critical consideration and thoughtful integration of clinical, biological, and engineering aspects are necessary to achieve translational bench-to-bedside research. In particular, identifying ideal biomaterial design specifications, understanding prior and recent tissue engineering advances, and judicious application of biomaterial and fabrication technologies will be crucial for addressing current clinical challenges in engineering multi-tissue units. Using rotator cuff tears as an example, insights relevant for engineering a bone-tendon-muscle multi-tissue unit are presented. This review highlights the tissue engineering strategies for musculoskeletal repair and regeneration with implications for other bone-tendon-muscle units, their derivatives, and analogous non-musculoskeletal tissue structures.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Yang Liu
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Bruma Sai-Chuen Fu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | | | - Anna Maria Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Patrick Shu-Hang Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
8
|
Daly AC, Prendergast ME, Hughes AJ, Burdick JA. Bioprinting for the Biologist. Cell 2021; 184:18-32. [PMID: 33417859 DOI: 10.1016/j.cell.2020.12.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/29/2020] [Accepted: 12/01/2020] [Indexed: 12/30/2022]
Abstract
Building tissues from scratch to explore entirely new cell configurations could revolutionize fundamental understanding in biology. Bioprinting is an emerging technology to do this. Although typically applied to engineer tissues for therapeutic tissue repair or drug screening, there are many opportunities for bioprinting within biology, such as for exploring cellular crosstalk or cellular morphogenesis. The overall goals of this Primer are to provide an overview of bioprinting with the biologist in mind, outline the steps in extrusion bioprinting (the most widely used and accessible technology), and discuss alternative bioprinting technologies and future opportunities for bioprinting in biology.
Collapse
Affiliation(s)
- Andrew C Daly
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Wang X, Gao B, Chan BP. Multiphoton microfabrication and micropatterning (MMM) - An all-in-one platform for engineering biomimetic soluble cell niches. Biomaterials 2021; 269:120644. [PMID: 33472153 DOI: 10.1016/j.biomaterials.2020.120644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023]
Abstract
Engineered biomimetic cell niches represent a valuable in vitro tool for investigating physiological and pathological cellular activities, while developing an all-in-one technology to engineer cell niches, particularly soluble cell niche factors, with retained bioactivities, remains challenging. Here, we report a mask-free, non-contact and biocompatible multiphoton microfabrication and micropatterning (MMM) technology in engineering a spatially and quantitatively controllable bone morphogenetic protein-2 (BMP-2) soluble niche, by immobilizing optimally biotinylated BMP-2 (bBMP-2) on micro-printed neutravidin (NA) micropatterns. Notably, the micropatterned NA bound-bBMP-2 niche elicited a more sustained and a higher level of the downstream Smad signaling than that by free BMP-2, in C2C12 cells, suggesting the advantages of immobilizing soluble niche factors on engineered micropatterns or scaffold materials. This work reports a universal all-in-one cell niche engineering platform and contributes to reconstituting heterogeneous native soluble cell niches for signal transduction modeling and drug screening studies.
Collapse
Affiliation(s)
- Xinna Wang
- Tissue Engineering Laboratory, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China
| | - Bo Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Barbara P Chan
- Tissue Engineering Laboratory, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region, China.
| |
Collapse
|
10
|
Hamid OA, Eltaher HM, Sottile V, Yang J. 3D bioprinting of a stem cell-laden, multi-material tubular composite: An approach for spinal cord repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111707. [DOI: 10.1016/j.msec.2020.111707] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/14/2020] [Accepted: 11/03/2020] [Indexed: 01/16/2023]
|
11
|
Lowen JM, Leach JK. Functionally graded biomaterials for use as model systems and replacement tissues. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909089. [PMID: 33456431 PMCID: PMC7810245 DOI: 10.1002/adfm.201909089] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 05/03/2023]
Abstract
The heterogeneity of native tissues requires complex materials to provide suitable substitutes for model systems and replacement tissues. Functionally graded materials have the potential to address this challenge by mimicking the gradients in heterogeneous tissues such as porosity, mineralization, and fiber alignment to influence strength, ductility, and cell signaling. Advancements in microfluidics, electrospinning, and 3D printing enable the creation of increasingly complex gradient materials that further our understanding of physiological gradients. The combination of these methods enables rapid prototyping of constructs with high spatial resolution. However, successful translation of these gradients requires both spatial and temporal presentation of cues to model the complexity of native tissues that few materials have demonstrated. This review highlights recent strategies to engineer functionally graded materials for the modeling and repair of heterogeneous tissues, together with a description of how cells interact with various gradients.
Collapse
Affiliation(s)
- Jeremy M. Lowen
- Department of Biomedical Engineering, University of California, Davis, CA, 95616
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, CA, 95616
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817
| |
Collapse
|
12
|
Li X, Liu B, Pei B, Chen J, Zhou D, Peng J, Zhang X, Jia W, Xu T. Inkjet Bioprinting of Biomaterials. Chem Rev 2020; 120:10793-10833. [PMID: 32902959 DOI: 10.1021/acs.chemrev.0c00008] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The inkjet technique has the capability of generating droplets in the picoliter volume range, firing thousands of times in a few seconds and printing in the noncontact manner. Since its emergence, inkjet technology has been widely utilized in the publishing industry for printing of text and pictures. As the technology developed, its applications have been expanded from two-dimensional (2D) to three-dimensional (3D) and even used to fabricate components of electronic devices. At the end of the twentieth century, researchers were aware of the potential value of this technology in life sciences and tissue engineering because its picoliter-level printing unit is suitable for depositing biological components. Currently inkjet technology has been becoming a practical tool in modern medicine serving for drug development, scaffold building, and cell depositing. In this article, we first review the history, principles and different methods of developing this technology. Next, we focus on the recent achievements of inkjet printing in the biological field. Inkjet bioprinting of generic biomaterials, biomacromolecules, DNAs, and cells and their major applications are introduced in order of increasing complexity. The current limitations/challenges and corresponding solutions of this technology are also discussed. A new concept, biopixels, is put forward with a combination of the key characteristics of inkjet printing and basic biological units to bring a comprehensive view on inkjet-based bioprinting. Finally, a roadmap of the entire 3D bioprinting is depicted at the end of this review article, clearly demonstrating the past, present, and future of 3D bioprinting and our current progress in this field.
Collapse
Affiliation(s)
- Xinda Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Boxun Liu
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
| | - Ben Pei
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Jianwei Chen
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China.,East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Jiayi Peng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Xinzhi Zhang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Tao Xu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
| |
Collapse
|
13
|
Shanjani Y, Siebert SM, Ker DFE, Mercado-Pagán AE, Yang YP. Acoustic Patterning of Growth Factor for Three-Dimensional Tissue Engineering. Tissue Eng Part A 2020; 26:602-612. [PMID: 31950880 PMCID: PMC7310194 DOI: 10.1089/ten.tea.2019.0271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
Temporal and spatial presentations of biological cues are critical for tissue engineering. There is a great need in improving the incorporation of bioagent(s) (specifically growth factor(s) [GF(s)]) onto three-dimensional scaffolds. In this study, we developed a process to combine additive manufacturing (AM) technology with acoustic droplet ejection (ADE) technology to control GF distribution. More specifically, we implemented ADE to control the distribution of recombinant human bone morphogenetic protein-2 (rhBMP-2) onto polycaprolactone (PCL)-based tissue engineering constructs (TECs). Three substrates were used in this study: (1) succinimide-terminated PCL (PCL-N-hydroxysuccinimide [NHS]) as model material, (2) alkali-treated PCL (PCL-NaOH) as first control material, and (3) fibrin-coated PCL (PCL-Fibrin) as second control material. It was shown that our process enables a pattern of BMP-2 spots of ∼250 μm in diameter with ∼700 μm center-to-center spacing. An initial concentration of BMP-2 higher than 300 μg/L was required to retain a detectable amount of GF on the substrate after a wash with phosphate-buffered solution. However, to obtain detectable osteogenic differentiation of C2C12 cells, the initial concentration of BMP-2 higher than 750 μg/L was needed. The cells on PCL-NHS samples showed spatial alkaline phosphatase staining correlating with local patterns of BMP-2, although the intensity was lower than the controls (PCL-NaOH and PCL-Fibrin). Our results have demonstrated that the developed AM-ADE process holds great promise in creating TECs with highly controlled GF patterning. Impact statement The combined process of additive manufacturing with acoustic droplet ejection to control growth factor (GF) distribution across three-dimensional (3D) porous scaffolds that is presented in this study enables creating 3D tissue engineering constructs with highly controlled GF patterning. Such constructs enable temporal and spatial presentations of biological cues for enhancing cell migration and differentiation and eventually the formation of targeted tissues in vitro and in vivo.
Collapse
Affiliation(s)
- Yaser Shanjani
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
| | - Sean Michael Siebert
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
| | - Dai Fei Elmer Ker
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, New Territories, Hong Kong SAR
| | - Angel E. Mercado-Pagán
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
| | - Yunzhi Peter Yang
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, California
- Department of Materials Science and Engineering, Stanford University, Stanford, California
- Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
14
|
Sun Y, Wang G, Jing Z, Liang J, Sui J, Fan J, Li J. Microfluidic Pneumatic Printed Sandwiched Microdroplet Array for High-Throughput Enzymatic Reaction and Screening. SLAS Technol 2020; 25:446-454. [PMID: 32406795 DOI: 10.1177/2472630320908248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
High-throughput enzyme screening for desired functionality is highly demanded. This paper utilizes a newly developed microfluidic pneumatic printing platform for high-throughput enzyme screening applications. The novel printing platform can achieve distinct features including a disposable cartridge, which avoids crosstalk; a flexible cartridge design, allowing for integration of multiple channels; and fast printing speed with submicroliter spot size. Moreover, a polydimethylsiloxane (PDMS)-based sandwich structure has been proposed and used during the printing and imaging, which can lead to better results, including reduced evaporation as well as a uniform light path during imaging. Using this microfluidic pneumatic printed PDMS sandwiched microdroplet array platform, we have demonstrated the capability of high-throughput generation of a combinatorial droplet array with concentration and volume gradients. Furthermore, the potential for enzymatic study has been validated by quantified cellulose reaction implemented with the printing platform.
Collapse
Affiliation(s)
- Yang Sun
- Key Laboratory of Straw Biology and Utilization, The Ministry of Education, College of Life Science, Jilin Agricultural University, Chang Chun, Ji Lin, China.,Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Gang Wang
- Key Laboratory of Straw Biology and Utilization, The Ministry of Education, College of Life Science, Jilin Agricultural University, Chang Chun, Ji Lin, China.,Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Zhi Jing
- Key Laboratory of Straw Biology and Utilization, The Ministry of Education, College of Life Science, Jilin Agricultural University, Chang Chun, Ji Lin, China
| | - Jingting Liang
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Jiajie Sui
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Jinzhen Fan
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Jiannan Li
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| |
Collapse
|
15
|
Progress in 3D bioprinting technology for tissue/organ regenerative engineering. Biomaterials 2020; 226:119536. [DOI: 10.1016/j.biomaterials.2019.119536] [Citation(s) in RCA: 359] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/25/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022]
|
16
|
|
17
|
Luo Y, Wei X, Huang P. 3D bioprinting of hydrogel‐based biomimetic microenvironments. J Biomed Mater Res B Appl Biomater 2018; 107:1695-1705. [DOI: 10.1002/jbm.b.34262] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 08/30/2018] [Accepted: 09/23/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Yongxiang Luo
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical Engineering, Health Science Center, Shenzhen University Shenzhen, 518060 China
| | - Xiaoyue Wei
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical Engineering, Health Science Center, Shenzhen University Shenzhen, 518060 China
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingSchool of Biomedical Engineering, Health Science Center, Shenzhen University Shenzhen, 518060 China
| |
Collapse
|
18
|
Bittner SM, Guo JL, Mikos AG. Spatiotemporal Control of Growth Factors in Three-Dimensional Printed Scaffolds. BIOPRINTING (AMSTERDAM, NETHERLANDS) 2018; 12:e00032. [PMID: 31106279 PMCID: PMC6519969 DOI: 10.1016/j.bprint.2018.e00032] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Three-dimensional printing (3DP) has enabled the fabrication of tissue engineering scaffolds that recapitulate the physical, architectural, and biochemical cues of native tissue matrix more effectively than ever before. One key component of biomimetic scaffold fabrication is the patterning of growth factors, whose spatial distribution and temporal release profile should ideally match that seen in native tissue development. Tissue engineers have made significant progress in improving the degree of spatiotemporal control over which growth factors are presented within 3DP scaffolds. However, significant limitations remain in terms in pattern resolution, the fabrication of true gradients, temporal control of growth factor release, the maintenance of growth factor distributions against diffusion, and more. This review summarizes several key areas for advancement of the field in terms of improving spatiotemporal control over growth factor presentation, and additionally highlights several major tissues of interest that have been targeted by 3DP growth factor patterning strategies.
Collapse
Affiliation(s)
- Sean M. Bittner
- Department of Bioengineering, Rice University, Houston, TX, United States
- Center for Engineering Complex Tissues, United States
| | - Jason L. Guo
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Antonios G. Mikos
- Department of Bioengineering, Rice University, Houston, TX, United States
- Center for Engineering Complex Tissues, United States
| |
Collapse
|
19
|
Oberoi G, Nitsch S, Edelmayer M, Janjić K, Müller AS, Agis H. 3D Printing-Encompassing the Facets of Dentistry. Front Bioeng Biotechnol 2018; 6:172. [PMID: 30525032 PMCID: PMC6262086 DOI: 10.3389/fbioe.2018.00172] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
This narrative review presents an overview on the currently available 3D printing technologies and their utilization in experimental, clinical and educational facets, from the perspective of different specialties of dentistry, including oral and maxillofacial surgery, orthodontics, endodontics, prosthodontics, and periodontics. It covers research and innovation, treatment modalities, education and training, employing the rapidly developing 3D printing process. Research-oriented advancement in 3D printing in dentistry is witnessed by the rising number of publications on this topic. Visualization of treatment outcomes makes it a promising clinical tool. Educational programs utilizing 3D-printed models stimulate training of dental skills in students and trainees. 3D printing has enormous potential to ameliorate oral health care in research, clinical treatment, and education in dentistry.
Collapse
Affiliation(s)
- Gunpreet Oberoi
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria
| | - Sophie Nitsch
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Department of Health Sciences, FH Wien, University of Applied Sciences, Vienna, Austria
| | - Michael Edelmayer
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Oral Surgery, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Klara Janjić
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Anna Sonja Müller
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Hermann Agis
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
20
|
Bittner SM, Guo JL, Melchiorri A, Mikos AG. Three-dimensional Printing of Multilayered Tissue Engineering Scaffolds. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2018; 21:861-874. [PMID: 30450010 PMCID: PMC6233733 DOI: 10.1016/j.mattod.2018.02.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The field of tissue engineering has produced new therapies for the repair of damaged tissues and organs, utilizing biomimetic scaffolds that mirror the mechanical and biological properties of host tissue. The emergence of three-dimensional printing (3DP) technologies has enabled the fabrication of highly complex scaffolds which offer a more accurate replication of native tissue properties and architecture than previously possible. Of strong interest to tissue engineers is the construction of multilayered scaffolds that target distinct regions of complex tissues. Musculoskeletal and dental tissues in particular, such as the osteochondral unit and periodontal complex, are composed of multiple interfacing tissue types, and thus benefit from the usage of multilayered scaffold fabrication. Traditional 3DP technologies such as extrusion printing and selective laser sintering have been used for the construction of scaffolds with gradient architectures and mixed material compositions. Additionally, emerging bioprinting strategies have been used for the direct printing and spatial patterning of cells and chemical factors, capturing the complex organization found in the body. To better replicate the varied and gradated properties of larger tissues, researchers have created scaffolds composed of multiple materials spanning natural polymers, synthetic polymers, and ceramics. By utilizing high precision 3DP techniques and judicious material selection, scaffolds can thus be designed to address the regeneration of previously challenging musculoskeletal, dental, and other heterogeneous target tissues. These multilayered 3DP strategies show great promise in the future of tissue engineering.
Collapse
Affiliation(s)
- Sean M Bittner
- Department of Bioengineering, Rice University, Houston, TX
- Center for Engineering Complex Tissues
| | - Jason L Guo
- Department of Bioengineering, Rice University, Houston, TX
| | - Anthony Melchiorri
- Department of Bioengineering, Rice University, Houston, TX
- Center for Engineering Complex Tissues
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX
- Center for Engineering Complex Tissues
| |
Collapse
|
21
|
Gungor-Ozkerim PS, Inci I, Zhang YS, Khademhosseini A, Dokmeci MR. Bioinks for 3D bioprinting: an overview. Biomater Sci 2018; 6:915-946. [PMID: 29492503 PMCID: PMC6439477 DOI: 10.1039/c7bm00765e] [Citation(s) in RCA: 629] [Impact Index Per Article: 104.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bioprinting is an emerging technology with various applications in making functional tissue constructs to replace injured or diseased tissues. It is a relatively new approach that provides high reproducibility and precise control over the fabricated constructs in an automated manner, potentially enabling high-throughput production. During the bioprinting process, a solution of a biomaterial or a mixture of several biomaterials in the hydrogel form, usually encapsulating the desired cell types, termed the bioink, is used for creating tissue constructs. This bioink can be cross-linked or stabilized during or immediately after bioprinting to generate the final shape, structure, and architecture of the designed construct. Bioinks may be made from natural or synthetic biomaterials alone, or a combination of the two as hybrid materials. In certain cases, cell aggregates without any additional biomaterials can also be adopted for use as a bioink for bioprinting processes. An ideal bioink should possess proper mechanical, rheological, and biological properties of the target tissues, which are essential to ensure correct functionality of the bioprinted tissues and organs. In this review, we provide an in-depth discussion of the different bioinks currently employed for bioprinting, and outline some future perspectives in their further development.
Collapse
Affiliation(s)
- P Selcan Gungor-Ozkerim
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | | | | | | | | |
Collapse
|
22
|
Biswas A, Malferrari S, Kalaskar DM, Das AK. Arylboronate esters mediated self-healable and biocompatible dynamic G-quadruplex hydrogels as promising 3D-bioinks. Chem Commun (Camb) 2018; 54:1778-1781. [PMID: 29383339 DOI: 10.1039/c7cc09051j] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Extrudable G-quadruplex hydrogels were prepared at physiological pH. Gels with suitable mechanical properties were explored as 3D-bioinks. The 3D printing process is driven by injectability and the highly thixotropic and self-healable nature of the gel. High cell viability and homogeneous cell distribution within the gel make it a promising material as a 3D bioink.
Collapse
Affiliation(s)
- Ankan Biswas
- Department of Chemistry, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, India.
| | | | | | | |
Collapse
|
23
|
Heher P, Mühleder S, Mittermayr R, Redl H, Slezak P. Fibrin-based delivery strategies for acute and chronic wound healing. Adv Drug Deliv Rev 2018; 129:134-147. [PMID: 29247766 DOI: 10.1016/j.addr.2017.12.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/24/2017] [Accepted: 12/09/2017] [Indexed: 12/17/2022]
Abstract
Fibrin, a natural hydrogel, is the end product of the physiological blood coagulation cascade and naturally involved in wound healing. Beyond its role in hemostasis, it acts as a local reservoir for growth factors and as a provisional matrix for invading cells that drive the regenerative process. Its unique intrinsic features do not only promote wound healing directly via modulation of cell behavior but it can also be fine-tuned to evolve into a delivery system for sustained release of therapeutic biomolecules, cells and gene vectors. To further augment tissue regeneration potential, current strategies exploit and modify the chemical and physical characteristics of fibrin to employ combined incorporation of several factors and their timed release. In this work we show advanced therapeutic approaches employing fibrin matrices in wound healing and cover the many possibilities fibrin offers to the field of regenerative medicine.
Collapse
|
24
|
Daly AC, Freeman FE, Gonzalez-Fernandez T, Critchley SE, Nulty J, Kelly DJ. 3D Bioprinting for Cartilage and Osteochondral Tissue Engineering. Adv Healthc Mater 2017; 6. [PMID: 28804984 DOI: 10.1002/adhm.201700298] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/15/2017] [Indexed: 12/16/2022]
Abstract
Significant progress has been made in the field of cartilage and bone tissue engineering over the last two decades. As a result, there is real promise that strategies to regenerate rather than replace damaged or diseased bones and joints will one day reach the clinic however, a number of major challenges must still be addressed before this becomes a reality. These include vascularization in the context of large bone defect repair, engineering complex gradients for bone-soft tissue interface regeneration and recapitulating the stratified zonal architecture present in many adult tissues such as articular cartilage. Tissue engineered constructs typically lack such spatial complexity in cell types and tissue organization, which may explain their relatively limited success to date. This has led to increased interest in bioprinting technologies in the field of musculoskeletal tissue engineering. The additive, layer by layer nature of such biofabrication strategies makes it possible to generate zonal distributions of cells, matrix and bioactive cues in 3D. The adoption of biofabrication technology in musculoskeletal tissue engineering may therefore make it possible to produce the next generation of biological implants capable of treating a range of conditions. Here, advances in bioprinting for cartilage and osteochondral tissue engineering are reviewed.
Collapse
Affiliation(s)
- Andrew C. Daly
- Trinity Center for Bioengineering; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering; School of Engineering; Trinity College Dublin; Dublin Ireland
- Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Fiona E. Freeman
- Trinity Center for Bioengineering; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering; School of Engineering; Trinity College Dublin; Dublin Ireland
- Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Tomas Gonzalez-Fernandez
- Trinity Center for Bioengineering; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering; School of Engineering; Trinity College Dublin; Dublin Ireland
- Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Susan E. Critchley
- Trinity Center for Bioengineering; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering; School of Engineering; Trinity College Dublin; Dublin Ireland
- Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Jessica Nulty
- Trinity Center for Bioengineering; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering; School of Engineering; Trinity College Dublin; Dublin Ireland
- Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Daniel J. Kelly
- Trinity Center for Bioengineering; Trinity Biomedical Sciences Institute; Trinity College Dublin; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering; School of Engineering; Trinity College Dublin; Dublin Ireland
- Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Advanced Materials and Bioengineering Research Center (AMBER); Royal College of Surgeons in Ireland and Trinity College Dublin; Dublin Ireland
| |
Collapse
|
25
|
Di Luca A, Klein-Gunnewiek M, Vancso JG, van Blitterswijk CA, Benetti EM, Moroni L. Covalent Binding of Bone Morphogenetic Protein-2 and Transforming Growth Factor-β3 to 3D Plotted Scaffolds for Osteochondral Tissue Regeneration. Biotechnol J 2017; 12. [PMID: 28865136 DOI: 10.1002/biot.201700072] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/28/2017] [Indexed: 11/08/2022]
Abstract
Engineering the osteochondral tissue presents some challenges mainly relying in its function of transition from the subchondral bone to articular cartilage and the gradual variation in several biological, mechanical, and structural features. A possible solution for osteochondral regeneration might be the design and fabrication of scaffolds presenting a gradient able to mimic this transition. Covalent binding of biological factors proved to enhance cell adhesion and differentiation in two-dimensional culture substrates. Here, we used polymer brushes as selective linkers of bone morphogenetic protein-2 (BMP-2) and transforming growth factor-β3 (TGF-β3) on the surface of 3D scaffolds fabricated via additive manufacturing (AM) and subsequent controlled radical polymerization. These growth factors (GFs) are known to stimulate the differentiation of human mesenchymal stromal cells (hMSCs) toward the osteogenic and chondrogenic lineages, respectively. BMP-2 and TGF-β3 were covalently bound both homogeneously within a poly(ethylene glycol) (PEG)-based brush-functionalized scaffolds, and following a gradient composition by varying their concentration along the axial section of the 3D constructs. Following an approach previously developed by our group and proved to be successful to generate fibronectin gradients, opposite brush-supported gradients of BMP-2 and TGF-β3 were finally generated and subsequently tested to differentiate cells in a gradient fashion. The brush-supported GFs significantly influenced hMSCs osteochondral differentiation when the scaffolds were homogenously modified, yet no effect was observed in the gradient scaffolds. Therefore, this technique seems promising to maintain the biological activity of growth factors covalently linked to 3D scaffolds, but needs to be further optimized in case biological gradients are desired.
Collapse
Affiliation(s)
- Andrea Di Luca
- University of Twente, Tissue Regeneration Department, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Michel Klein-Gunnewiek
- University of Twente, Materials Science and Technology of Polymers Group, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Julius G Vancso
- University of Twente, Materials Science and Technology of Polymers Group, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Clemens A van Blitterswijk
- University of Twente, Tissue Regeneration Department, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.,Maastricht University, MERLN Institute for Technology Inspired Regenerative Medicine, Complex Tissue Regeneration Department, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Edmondo M Benetti
- University of Twente, Materials Science and Technology of Polymers Group, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.,Laboratory for Surface Science and Technology, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, 8093-CH Zürich, Switzerland
| | - Lorenzo Moroni
- University of Twente, Tissue Regeneration Department, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.,Maastricht University, MERLN Institute for Technology Inspired Regenerative Medicine, Complex Tissue Regeneration Department, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
26
|
Peng W, Datta P, Ayan B, Ozbolat V, Sosnoski D, Ozbolat IT. 3D bioprinting for drug discovery and development in pharmaceutics. Acta Biomater 2017; 57:26-46. [PMID: 28501712 DOI: 10.1016/j.actbio.2017.05.025] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 02/08/2023]
Abstract
Successful launch of a commercial drug requires significant investment of time and financial resources wherein late-stage failures become a reason for catastrophic failures in drug discovery. This calls for infusing constant innovations in technologies, which can give reliable prediction of efficacy, and more importantly, toxicology of the compound early in the drug discovery process before clinical trials. Though computational advances have resulted in more rationale in silico designing, in vitro experimental studies still require gaining industry confidence and improving in vitro-in vivo correlations. In this quest, due to their ability to mimic the spatial and chemical attributes of native tissues, three-dimensional (3D) tissue models have now proven to provide better results for drug screening compared to traditional two-dimensional (2D) models. However, in vitro fabrication of living tissues has remained a bottleneck in realizing the full potential of 3D models. Recent advances in bioprinting provide a valuable tool to fabricate biomimetic constructs, which can be applied in different stages of drug discovery research. This paper presents the first comprehensive review of bioprinting techniques applied for fabrication of 3D tissue models for pharmaceutical studies. A comparative evaluation of different bioprinting modalities is performed to assess the performance and ability of fabricating 3D tissue models for pharmaceutical use as the critical selection of bioprinting modalities indeed plays a crucial role in efficacy and toxicology testing of drugs and accelerates the drug development cycle. In addition, limitations with current tissue models are discussed thoroughly and future prospects of the role of bioprinting in pharmaceutics are provided to the reader. STATEMENT OF SIGNIFICANCE Present advances in tissue biofabrication have crucial role to play in aiding the pharmaceutical development process achieve its objectives. Advent of three-dimensional (3D) models, in particular, is viewed with immense interest by the community due to their ability to mimic in vivo hierarchical tissue architecture and heterogeneous composition. Successful realization of 3D models will not only provide greater in vitro-in vivo correlation compared to the two-dimensional (2D) models, but also eventually replace pre-clinical animal testing, which has their own shortcomings. Amongst all fabrication techniques, bioprinting- comprising all the different modalities (extrusion-, droplet- and laser-based bioprinting), is emerging as the most viable fabrication technique to create the biomimetic tissue constructs. Notwithstanding the interest in bioprinting by the pharmaceutical development researchers, it can be seen that there is a limited availability of comparative literature which can guide the proper selection of bioprinting processes and associated considerations, such as the bioink selection for a particular pharmaceutical study. Thus, this work emphasizes these aspects of bioprinting and presents them in perspective of differential requirements of different pharmaceutical studies like in vitro predictive toxicology, high-throughput screening, drug delivery and tissue-specific efficacies. Moreover, since bioprinting techniques are mostly applied in regenerative medicine and tissue engineering, a comparative analysis of similarities and differences are also expounded to help researchers make informed decisions based on contemporary literature.
Collapse
|
27
|
|
28
|
Zhang W, Gorantla VS, Campbell PG, Li Y, Yang Y, Komatsu C, Weiss LE, Zheng XX, Solari MG. Biopatterned CTLA4/Fc Matrices Facilitate Local Immunomodulation, Engraftment, and Glucose Homeostasis After Pancreatic Islet Transplantation. Diabetes 2016; 65:3660-3666. [PMID: 27650855 DOI: 10.2337/db16-0320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 09/07/2016] [Indexed: 11/13/2022]
Abstract
Pancreatic islet transplantation (PIT) represents a potential therapy to circumvent the need for exogenous insulin in type 1 diabetes. However, PIT remains limited by lack of donor islets and the need for long-term multidrug immunosuppression to prevent alloimmune islet rejection. Our goal was to evaluate a local immunoregulatory strategy that sustains islet allograft survival and restores glucose homeostasis in the absence of systemic immunosuppression. Nanogram quantities of murine CTLA4/Fc fusion protein were controllably delivered within human acellular dermal matrix scaffolds using an inkjet-based biopatterning technology and cotransplanted with allogeneic islets under the renal capsule to create an immunoregulatory microenvironment around the islet allograft. We achieved long-term engraftment of small loads of allogeneic islet cells with 40% of MHC-mismatched mouse recipients maintaining sustained normoglycemia following pancreatic β-cell ablation by streptozotocin. Biopatterned CTLA4/Fc local therapy was associated with expansion of Foxp3+ regulatory T cells and shifts in cytokine production and gene expression from proinflammatory to regulatory profiles, thus substantially benefiting islet allografts survival and function. This study is a new paradigm for targeted therapies in PIT that demonstrates the favorable effects of immune alterations in the transplant milieu and suggests a unique strategy for minimizing systemic immunosuppression and promoting islet allograft survival.
Collapse
Affiliation(s)
- Wensheng Zhang
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Vijay S Gorantla
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Phil G Campbell
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA
| | - Yang Li
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Yang Yang
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Chiaki Komatsu
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Lee E Weiss
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Robotics Institute, Carnegie Mellon University, Pittsburgh, PA
| | - Xin Xiao Zheng
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Transplantation Medical Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Mario G Solari
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
29
|
Abstract
The ability to three-dimensionally interweave biological and functional materials could enable the creation of bionic devices possessing unique and compelling geometries, properties, and functionalities. Indeed, interfacing high performance active devices with biology could impact a variety of fields, including regenerative bioelectronic medicines, smart prosthetics, medical robotics, and human-machine interfaces. Biology, from the molecular scale of DNA and proteins, to the macroscopic scale of tissues and organs, is three-dimensional, often soft and stretchable, and temperature sensitive. This renders most biological platforms incompatible with the fabrication and materials processing methods that have been developed and optimized for functional electronics, which are typically planar, rigid and brittle. A number of strategies have been developed to overcome these dichotomies. One particularly novel approach is the use of extrusion-based multi-material 3D printing, which is an additive manufacturing technology that offers a freeform fabrication strategy. This approach addresses the dichotomies presented above by (1) using 3D printing and imaging for customized, hierarchical, and interwoven device architectures; (2) employing nanotechnology as an enabling route for introducing high performance materials, with the potential for exhibiting properties not found in the bulk; and (3) 3D printing a range of soft and nanoscale materials to enable the integration of a diverse palette of high quality functional nanomaterials with biology. Further, 3D printing is a multi-scale platform, allowing for the incorporation of functional nanoscale inks, the printing of microscale features, and ultimately the creation of macroscale devices. This blending of 3D printing, novel nanomaterial properties, and 'living' platforms may enable next-generation bionic systems. In this review, we highlight this synergistic integration of the unique properties of nanomaterials with the versatility of extrusion-based 3D printing technologies to interweave nanomaterials and fabricate novel bionic devices.
Collapse
Affiliation(s)
- Yong Lin Kong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Maneesh K. Gupta
- Air Force Research Laboratories, Wright-Patterson Air Force Base, Ohio 45433, USA
| | - Blake N. Johnson
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Michael C. McAlpine
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
30
|
Borzenkov M, Määttänen A, Ihalainen P, Collini M, Cabrini E, Dacarro G, Pallavicini P, Chirico G. Fabrication of Inkjet-Printed Gold Nanostar Patterns with Photothermal Properties on Paper Substrate. ACS APPLIED MATERIALS & INTERFACES 2016; 8:9909-16. [PMID: 27031124 DOI: 10.1021/acsami.6b02983] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Inkjet printing technology has brought significant advances in patterning various functional materials that can meet important challenges in personalized medical treatments. Indeed, patterning of photothermal active anisotropic gold nanoparticles is particularly promising for the development of low-cost tools for localized photothermal therapy. In the present work, stable inks containing PEGylated gold nanostars (GNSs) were prepared and inkjet printed on a pigment-coated paper substrate. A significant photothermal effect (ΔT ≅ 20 °C) of the printed patterns was observed under near infrared (NIR) excitation of the localized surface plasmon resonance (LSPR) of the GNS with low laser intensity (I ≅ 0.2 W/cm(2)). Besides the pronounced photothermal effect, we also demonstrated, as an additional valuable effect, the release of a model fluorescent thiol-terminated Bodipy dye (BDP-SH) from the printed gold surface, both under bulk heating and NIR irradiation. These preliminary results suggest the way of the development of a new class of low-cost, disposable, and smart devices for localized thermal treatments combined with temperature-triggered drug release.
Collapse
Affiliation(s)
- Mykola Borzenkov
- Department of Physics "G. Occhialini", Nanomedicine Center, University of Milano-Bicocca , piazza della Scienza 3, 20126 Milan, Italy
| | - Anni Määttänen
- Laboratory of Physical Chemistry, Center for Functional Materials, Åbo Akademi University , Porthaninkatu 3-5, 20500 Turku, Finland
| | - Petri Ihalainen
- Laboratory of Physical Chemistry, Center for Functional Materials, Åbo Akademi University , Porthaninkatu 3-5, 20500 Turku, Finland
| | - Maddalena Collini
- Department of Physics "G. Occhialini", Nanomedicine Center, University of Milano-Bicocca , piazza della Scienza 3, 20126 Milan, Italy
| | - Elisa Cabrini
- Department of Chemistry, University of Pavia , viale Taramelli 12, 27100 Pavia, Italy
| | - Giacomo Dacarro
- Department of Chemistry, University of Pavia , viale Taramelli 12, 27100 Pavia, Italy
| | | | - Giuseppe Chirico
- Department of Physics "G. Occhialini", Nanomedicine Center, University of Milano-Bicocca , piazza della Scienza 3, 20126 Milan, Italy
| |
Collapse
|
31
|
Affiliation(s)
- Falguni Pati
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory KTH – Royal Institute of Technology Stockholm Schweden
| | - Jesper Gantelius
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory KTH – Royal Institute of Technology Stockholm Schweden
| | - Helene Andersson Svahn
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory KTH – Royal Institute of Technology Stockholm Schweden
| |
Collapse
|
32
|
Pati F, Gantelius J, Svahn HA. 3D Bioprinting of Tissue/Organ Models. Angew Chem Int Ed Engl 2016; 55:4650-65. [PMID: 26895542 DOI: 10.1002/anie.201505062] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 12/17/2022]
Abstract
In vitro tissue/organ models are useful platforms that can facilitate systematic, repetitive, and quantitative investigations of drugs/chemicals. The primary objective when developing tissue/organ models is to reproduce physiologically relevant functions that typically require complex culture systems. Bioprinting offers exciting prospects for constructing 3D tissue/organ models, as it enables the reproducible, automated production of complex living tissues. Bioprinted tissues/organs may prove useful for screening novel compounds or predicting toxicity, as the spatial and chemical complexity inherent to native tissues/organs can be recreated. In this Review, we highlight the importance of developing 3D in vitro tissue/organ models by 3D bioprinting techniques, characterization of these models for evaluating their resemblance to native tissue, and their application in the prioritization of lead candidates, toxicity testing, and as disease/tumor models.
Collapse
Affiliation(s)
- Falguni Pati
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Jesper Gantelius
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Helene Andersson Svahn
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
33
|
Migliorini E, Valat A, Picart C, Cavalcanti-Adam EA. Tuning cellular responses to BMP-2 with material surfaces. Cytokine Growth Factor Rev 2015; 27:43-54. [PMID: 26704296 DOI: 10.1016/j.cytogfr.2015.11.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/13/2015] [Indexed: 02/08/2023]
Abstract
Bone morphogenetic protein 2 (BMP-2) has been known for decades as a strong osteoinductive factor and for clinical applications is combined solely with collagen as carrier material. The growing concerns regarding side effects and the importance of BMP-2 in several developmental and physiological processes have raised the need to improve the design of materials by controlling BMP-2 presentation. Inspired by the natural cell environment, new material surfaces have been engineered and tailored to provide both physical and chemical cues that regulate BMP-2 activity. Here we describe surfaces designed to present BMP-2 to cells in a spatially and temporally controlled manner. This is achieved by trapping BMP-2 using physicochemical interactions, either covalently grafted or combined with other extracellular matrix components. In the near future, we anticipate that material science and biology will integrate and further develop tools for in vitro studies and potentially bring some of them toward in vivo applications.
Collapse
Affiliation(s)
- Elisa Migliorini
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, D-70569 Stuttgart, Germany
- Department of Biophysical Chemistry, University of Heidelberg, INF 253, D-69120 Heidelberg, Germany, Tel: +49-6221-54 5064
| | - Anne Valat
- CNRS-UMR 5628, LMGP, 3 parvis L.Néel, F-38 016 Grenoble, France
- University Grenoble Alpes, Grenoble Institute of Technology, LMGP, 3 parvis Louis Néel, F-28016 Grenoble, France
- INSERM U823, ERL CNRS5284, Université de Grenoble Alpes, Institut Albert Bonniot, Site Santé, BP170, 38042 Grenoble cedex 9, France, Tel: +33-04-56529311
| | - Catherine Picart
- CNRS-UMR 5628, LMGP, 3 parvis L.Néel, F-38 016 Grenoble, France
- University Grenoble Alpes, Grenoble Institute of Technology, LMGP, 3 parvis Louis Néel, F-28016 Grenoble, France
| | - Elisabetta Ada Cavalcanti-Adam
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, D-70569 Stuttgart, Germany
- Department of Biophysical Chemistry, University of Heidelberg, INF 253, D-69120 Heidelberg, Germany, Tel: +49-6221-54 5064
| |
Collapse
|
34
|
Lagunas A, Martínez E, Samitier J. Surface-Bound Molecular Gradients for the High-Throughput Screening of Cell Responses. Front Bioeng Biotechnol 2015; 3:132. [PMID: 26380260 PMCID: PMC4553394 DOI: 10.3389/fbioe.2015.00132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/17/2015] [Indexed: 11/24/2022] Open
Abstract
Chemical gradient surfaces are described as surfaces with a gradually varying composition along their length. Continuous chemical gradients have recently been proposed as an alternative to discrete microarrays for the high-throughput screening of the effects of ligand concentration in cells. Here, we review some of the most recent examples in which gradients have been used to evaluate the effect of a varying ligand concentration in cell adhesion, morphology, growth, and differentiation of cells, including some of our recent findings. They show the importance of the organization of ligands at the nanoscale, which is highlighted by abrupt changes in cell behavior at critical concentration thresholds.
Collapse
Affiliation(s)
- Anna Lagunas
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) , Madrid , Spain ; Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC) , Barcelona , Spain
| | - Elena Martínez
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) , Madrid , Spain ; Biomimetic Systems for Cell Engineering Group, Institute for Bioengineering of Catalonia (IBEC) , Barcelona , Spain ; Department of Electronics, University of Barcelona (UB) , Barcelona , Spain
| | - Josep Samitier
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) , Madrid , Spain ; Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC) , Barcelona , Spain ; Department of Electronics, University of Barcelona (UB) , Barcelona , Spain
| |
Collapse
|
35
|
Tao H, Marelli B, Yang M, An B, Onses MS, Rogers JA, Kaplan DL, Omenetto FG. Inkjet Printing of Regenerated Silk Fibroin: From Printable Forms to Printable Functions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:4273-4279. [PMID: 26079217 DOI: 10.1002/adma.201501425] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/13/2015] [Indexed: 06/04/2023]
Abstract
A formulation of regenerated silk fibroin solution that can be easily functionalized and inkjet printed on numerous surfaces is developed. As an example, the inks can be printed on laboratory gloves that change color when exposed to bacteria.
Collapse
Affiliation(s)
- Hu Tao
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, CAS, Shanghai, 200050, PR China
| | - Benedetto Marelli
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Miaomiao Yang
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Bo An
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - M Serdar Onses
- Department of Materials Science and Engineering, Beckman Institute for Advanced Science and Technology, Urbana, IL, 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - John A Rogers
- Department of Materials Science and Engineering, Beckman Institute for Advanced Science and Technology, Urbana, IL, 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
| | - Fiorenzo G Omenetto
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Physics, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
36
|
Herberg S, Kondrikova G, Periyasamy-Thandavan S, Howie RN, Elsalanty ME, Weiss L, Campbell P, Hill WD, Cray JJ. Inkjet-based biopatterning of SDF-1β augments BMP-2-induced repair of critical size calvarial bone defects in mice. Bone 2014; 67:95-103. [PMID: 25016095 PMCID: PMC4149833 DOI: 10.1016/j.bone.2014.07.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/08/2014] [Accepted: 07/03/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND A major problem in craniofacial surgery is non-healing bone defects. Autologous reconstruction remains the standard of care for these cases. Bone morphogenetic protein-2 (BMP-2) therapy has proven its clinical utility, although non-targeted adverse events occur due to the high milligram-level doses used. Ongoing efforts explore the use of different growth factors, cytokines, or chemokines, as well as co-therapy to augment healing. METHODS Here we utilize inkjet-based biopatterning to load acellular DermaMatrix delivery matrices with nanogram-level doses of BMP-2, stromal cell-derived factor-1β (SDF-1β), transforming growth factor-β1 (TGF-β1), or co-therapies thereof. We tested the hypothesis that bioprinted SDF-1β co-delivery enhances BMP-2 and TGF-β1-driven osteogenesis both in-vitro and in-vivo using a mouse calvarial critical size defect (CSD) model. RESULTS Our data showed that BMP-2 bioprinted in low-doses induced significant new bone formation by four weeks post-operation. TGF-β1 was less effective compared to BMP-2, and SDF-1β therapy did not enhance osteogenesis above control levels. However, co-delivery of BMP-2+SDF-1β was shown to augment BMP-2-induced bone formation compared to BMP-2 alone. In contrast, co-delivery of TGF-β1+SDF-1β decreased bone healing compared to TGF-β1 alone. This was further confirmed in vitro by osteogenic differentiation studies using MC3T3-E1 pre-osteoblasts. CONCLUSIONS Our data indicates that sustained release delivery of a low-dose growth factor therapy using biopatterning technology can aid in healing CSD injuries. SDF-1β augments the ability for BMP-2 to drive healing, a result confirmed in vivo and in vitro; however, because SDF-1β is detrimental to TGF-β1-driven osteogenesis, its effect on osteogenesis is not universal.
Collapse
Affiliation(s)
- Samuel Herberg
- Department of Cellular Biology and Anatomy, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA
| | - Galina Kondrikova
- Department of Cellular Biology and Anatomy, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA
| | | | - R Nicole Howie
- Department of Oral Biology, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA
| | - Mohammed E Elsalanty
- Department of Oral Biology, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA; The Institute for Regenerative and Reparative Medicine, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA
| | - Lee Weiss
- The Robotics Institute, Carnegie Mellon University, 5000 Forbes Ave., Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, 450 Technology Drive, Pittsburgh, PA, USA
| | - Phil Campbell
- The Institute for Complex Engineered Systems, Carnegie Mellon University, 5000 Forbes Ave., Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, 450 Technology Drive, Pittsburgh, PA, USA
| | - William D Hill
- Department of Cellular Biology and Anatomy, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA; Department of Orthopaedic Surgery, Georgia Regents University, 1120 15th St., Augusta, GA, USA; The Institute for Regenerative and Reparative Medicine, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - James J Cray
- Department of Cellular Biology and Anatomy, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA; Department of Oral Biology, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA; Department of Orthopaedic Surgery, Georgia Regents University, 1120 15th St., Augusta, GA, USA; Department of Orthodontics and Surgery, Division of Plastic Surgery, Georgia Regents University, 1120 15th St., Augusta, GA, USA; The Institute for Regenerative and Reparative Medicine, Georgia Regents University, 1459 Laney Walker Blvd., Augusta, GA, USA.
| |
Collapse
|
37
|
Gurkan UA, El Assal R, Yildiz SE, Sung Y, Trachtenberg AJ, Kuo WP, Demirci U. Engineering anisotropic biomimetic fibrocartilage microenvironment by bioprinting mesenchymal stem cells in nanoliter gel droplets. Mol Pharm 2014; 11:2151-9. [PMID: 24495169 PMCID: PMC4096228 DOI: 10.1021/mp400573g] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past decade, bioprinting has emerged as a promising patterning strategy to organize cells and extracellular components both in two and three dimensions (2D and 3D) to engineer functional tissue mimicking constructs. So far, tissue printing has neither been used for 3D patterning of mesenchymal stem cells (MSCs) in multiphase growth factor embedded 3D hydrogels nor been investigated phenotypically in terms of simultaneous differentiation into different cell types within the same micropatterned 3D tissue constructs. Accordingly, we demonstrated a biochemical gradient by bioprinting nanoliter droplets encapsulating human MSCs, bone morphogenetic protein 2 (BMP-2), and transforming growth factor β1 (TGF- β1), engineering an anisotropic biomimetic fibrocartilage microenvironment. Assessment of the model tissue construct displayed multiphasic anisotropy of the incorporated biochemical factors after patterning. Quantitative real time polymerase chain reaction (qRT-PCR) results suggested genomic expression patterns leading to simultaneous differentiation of MSC populations into osteogenic and chondrogenic phenotype within the multiphasic construct, evidenced by upregulation of osteogenesis and condrogenesis related genes during in vitro culture. Comprehensive phenotypic network and pathway analysis results, which were based on genomic expression data, indicated activation of differentiation related mechanisms, via signaling pathways, including TGF, BMP, and vascular endothelial growth factor.
Collapse
Affiliation(s)
- Umut A Gurkan
- Case Biomanufacturing and Microfabrication Laboratory, Mechanical and Aerospace Engineering Department, Department of Orthopaedics, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | | | | | | | | | | | | |
Collapse
|
38
|
Almodóvar J, Guillot R, Monge C, Vollaire J, Selimović S, Coll JL, Khademhosseini A, Picart C. Spatial patterning of BMP-2 and BMP-7 on biopolymeric films and the guidance of muscle cell fate. Biomaterials 2014; 35:3975-85. [PMID: 24485790 DOI: 10.1016/j.biomaterials.2014.01.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/07/2014] [Indexed: 10/25/2022]
Abstract
In the cellular microenvironment, growth factor gradients are crucial in dictating cell fate. Towards developing materials that capture the native microenvironment we engineered biomimetic films that present gradients of matrix-bound bone morphogenetic proteins (BMP-2 and BMP-7). To this end layer-by-layer films composed of poly(L-lysine) and hyaluronan were combined in a simple microfluidic device enabling spatially controlled growth factor diffusion along the film. Linear long-range gradients of both BMPs induced the trans-differentiation of C2C12 myoblasts towards the osteogenic lineage in a dose dependent manner with a different signature for each BMP. The osteogenic marker alkaline phosphatase (ALP) increased in a linear manner for BMP-7 and non-linearly for BMP-2. Moreover, an increased expression of the myogenic marker troponin T was observed with decreasing matrix-bound BMP concentration, providing a substrate that it is both osteo- and myo-inductive. Lastly, dual parallel matrix-bound gradients of BMP-2 and -7 revealed a complete saturation of the ALP signal. This suggested an additive or synergistic effect of the two BMPs. This simple technology allows for determining quickly and efficiently the optimal concentration of matrix-bound growth factors, as well as for investigating the presentation of multiple growth factors in their solid-phase and in a spatially controlled manner.
Collapse
Affiliation(s)
- Jorge Almodóvar
- CNRS UMR 5628 (LMGP), MINATEC, 3 parvis Louis Néel, 38016 Grenoble, France; Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Raphaël Guillot
- CNRS UMR 5628 (LMGP), MINATEC, 3 parvis Louis Néel, 38016 Grenoble, France; Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Claire Monge
- CNRS UMR 5628 (LMGP), MINATEC, 3 parvis Louis Néel, 38016 Grenoble, France; Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, 38016 Grenoble, France
| | | | - Seila Selimović
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Catherine Picart
- CNRS UMR 5628 (LMGP), MINATEC, 3 parvis Louis Néel, 38016 Grenoble, France; Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, 38016 Grenoble, France.
| |
Collapse
|
39
|
Shi X, Ostrovidov S, Shu Y, Liang X, Nakajima K, Wu H, Khademhosseini A. Microfluidic generation of polydopamine gradients on hydrophobic surfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:832-8. [PMID: 24358938 DOI: 10.1021/la4041216] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Engineered surface-bound molecular gradients are of great importance for a range of biological applications. In this paper, we fabricated a polydopamine gradient on a hydrophobic surface. A microfluidic device was used to generate a covalently conjugated gradient of polydopamine (PDA), which changed the wettabilty and the surface energy of the substrate. The gradient was subsequently used to enable the spatial deposition of adhesive proteins on the surface. When seeded with human adipose mesenchymal stem cells, the PDA-graded surface induced a gradient of cell adhesion and spreading. The PDA gradient developed in this study is a promising tool for controlling cellular behavior and may be useful in various biological applications.
Collapse
Affiliation(s)
- Xuetao Shi
- WPI-Advanced Institute for Materials Research, Tohoku University , Sendai 980-8578, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Almodóvar J, Crouzier T, Selimović Š, Boudou T, Khademhosseini A, Picart C. Gradients of physical and biochemical cues on polyelectrolyte multilayer films generated via microfluidics. LAB ON A CHIP 2013; 13:1562-70. [PMID: 23440074 PMCID: PMC4155072 DOI: 10.1039/c3lc41407h] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The cell microenvironment is a complex and anisotropic matrix composed of a number of physical and biochemical cues that control cellular processes. A current challenge in biomaterials is the engineering of biomimetic materials which present spatially controlled physical and biochemical cues. The layer-by-layer assembly of polyelectrolyte multilayers (PEM) has been demonstrated to be a promising candidate for a biomaterial mimicking the native extracellular matrix. In this work, gradients of biochemical and physical cues were generated on PEM films composed of hyaluronan (HA) and poly(l-lysine) (PLL) using a microfluidic device. As a proof of concept, four different types of surface concentration gradients adsorbed onto the films were generated. These included surface concentration gradients of fluorescent PLL, fluorescent microbeads, a cross-linker, and one consisting of a polyelectrolyte grafted with a cell adhesive peptide. In all cases, reproducible centimeter-long linear gradients were obtained. Fluorescence microscopy, Fourier transform infrared spectroscopy and atomic force microscopy were used to characterize these gradients. Cell responses to the stiffness gradient and to the peptide gradient were studied. Pre-osteoblastic cells were found to adhere and spread more along the stiffness gradient, which varied linearly from 200 kPa-600 kPa. Myoblast cell spreading also increased throughout the length of the increasing RGD-peptide gradient. This work demonstrates a simple method to modify PEM films with concentration gradients of non-covalently bound biomolecules and with gradients in stiffness. These results highlight the potential of this technique to efficiently and quickly determine the optimal biochemical and mechanical cues necessary for specific cellular processes.
Collapse
Affiliation(s)
- Jorge Almodóvar
- LMGP, UMR 5628, CNRS and Grenoble Institute of Technology, MINATEC, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Thomas Crouzier
- LMGP, UMR 5628, CNRS and Grenoble Institute of Technology, MINATEC, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Šeila Selimović
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Thomas Boudou
- LMGP, UMR 5628, CNRS and Grenoble Institute of Technology, MINATEC, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Catherine Picart
- LMGP, UMR 5628, CNRS and Grenoble Institute of Technology, MINATEC, 3 parvis Louis Néel, 38016, Grenoble, France
| |
Collapse
|
41
|
Ross AM, Lahann J. Surface engineering the cellular microenvironment via patterning and gradients. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/polb.23275] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
42
|
Lagunas A, Comelles J, Oberhansl S, Hortigüela V, Martínez E, Samitier J. Continuous bone morphogenetic protein-2 gradients for concentration effect studies on C2C12 osteogenic fate. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:694-701. [PMID: 23313904 DOI: 10.1016/j.nano.2012.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 12/26/2012] [Indexed: 11/20/2022]
Abstract
UNLABELLED Cells can respond to small changes in a varying concentration of exogenous signaling molecules. Here we propose the use of continuous surface chemical gradients for the in-depth study of dose-dependent effects on cells. A continuous surface gradient of bone morphogenetic protein-2 (BMP-2) is presented. The gradient covers a narrow range of surface densities (from 1.4 to 2.3 pmol/cm(2)) with a shallow slope (0.9 pmol/cm(3)). These characteristics represent a quasi-homogeneous surface concentration at the cell scale, which is crucial for cell screening studies. Cell fate evaluation at early stages of osteogenesis in C2C12 cells, indicates the potential of continuous gradients for in vitro screening applications. FROM THE CLINICAL EDITOR The authors propose the use of surface-applied continuous chemical gradients for in-depth study of dose-dependent effects on cells. The method is demonstrated using BMP-2 proteins on C2C12 cells as a model system.
Collapse
Affiliation(s)
- Anna Lagunas
- Centro de Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (Ciber-bbn), C/ María de Luna 11, Edificio CEEI, 50018 Zaragoza, Spain.
| | | | | | | | | | | |
Collapse
|
43
|
Tasoglu S, Demirci U. Bioprinting for stem cell research. Trends Biotechnol 2012; 31:10-9. [PMID: 23260439 DOI: 10.1016/j.tibtech.2012.10.005] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 10/15/2012] [Accepted: 10/19/2012] [Indexed: 12/18/2022]
Abstract
Recently, there has been growing interest in applying bioprinting techniques to stem cell research. Several bioprinting methods have been developed utilizing acoustics, piezoelectricity, and lasers to deposit living cells onto receiving substrates. Using these technologies, spatially defined gradients of immobilized biomolecules can be engineered to direct stem cell differentiation into multiple subpopulations of different lineages. Stem cells can also be patterned in a high-throughput manner onto flexible implementation patches for tissue regeneration or onto substrates with the goal of accessing encapsulated stem cells of interest for genomic analysis. Here, we review recent achievements with bioprinting technologies in stem cell research, and identify future challenges and potential applications including tissue engineering and regenerative medicine, wound healing, and genomics.
Collapse
Affiliation(s)
- Savas Tasoglu
- Brigham and Women's Hospital, Bio-Acoustic MEMS in Medicine Lab, Division of Biomedical Engineering, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
44
|
KHATIWALA CHIRAG, LAW RICHARD, SHEPHERD BENJAMIN, DORFMAN SCOTT, CSETE MARIE. 3D CELL BIOPRINTING FOR REGENERATIVE MEDICINE RESEARCH AND THERAPIES. ACTA ACUST UNITED AC 2012. [DOI: 10.1142/s1568558611000301] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
45
|
Zhu X, Zheng Q, Yang H, Cai J, Huang L, Duan Y, Xu Z, Cen P. Recent advances in inkjet dispensing technologies: applications in drug discovery. Expert Opin Drug Discov 2012; 7:761-70. [DOI: 10.1517/17460441.2012.697892] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
46
|
Ker DFE, Weiss LE, Junkers SN, Chen M, Yin Z, Sandbothe MF, Huh SI, Eom S, Bise R, Osuna-Highley E, Kanade T, Campbell PG. An engineered approach to stem cell culture: automating the decision process for real-time adaptive subculture of stem cells. PLoS One 2011; 6:e27672. [PMID: 22110715 PMCID: PMC3218005 DOI: 10.1371/journal.pone.0027672] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 10/21/2011] [Indexed: 11/18/2022] Open
Abstract
Current cell culture practices are dependent upon human operators and remain laborious and highly subjective, resulting in large variations and inconsistent outcomes, especially when using visual assessments of cell confluency to determine the appropriate time to subculture cells. Although efforts to automate cell culture with robotic systems are underway, the majority of such systems still require human intervention to determine when to subculture. Thus, it is necessary to accurately and objectively determine the appropriate time for cell passaging. Optimal stem cell culturing that maintains cell pluripotency while maximizing cell yields will be especially important for efficient, cost-effective stem cell-based therapies. Toward this goal we developed a real-time computer vision-based system that monitors the degree of cell confluency with a precision of 0.791±0.031 and recall of 0.559±0.043. The system consists of an automated phase-contrast time-lapse microscope and a server. Multiple dishes are sequentially imaged and the data is uploaded to the server that performs computer vision processing, predicts when cells will exceed a pre-defined threshold for optimal cell confluency, and provides a Web-based interface for remote cell culture monitoring. Human operators are also notified via text messaging and e-mail 4 hours prior to reaching this threshold and immediately upon reaching this threshold. This system was successfully used to direct the expansion of a paradigm stem cell population, C2C12 cells. Computer-directed and human-directed control subcultures required 3 serial cultures to achieve the theoretical target cell yield of 50 million C2C12 cells and showed no difference for myogenic and osteogenic differentiation. This automated vision-based system has potential as a tool toward adaptive real-time control of subculturing, cell culture optimization and quality assurance/quality control, and it could be integrated with current and developing robotic cell cultures systems to achieve complete automation.
Collapse
Affiliation(s)
- Dai Fei Elmer Ker
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ker EDF, Nain AS, Weiss LE, Wang J, Suhan J, Amon CH, Campbell PG. Bioprinting of growth factors onto aligned sub-micron fibrous scaffolds for simultaneous control of cell differentiation and alignment. Biomaterials 2011; 32:8097-107. [PMID: 21820736 DOI: 10.1016/j.biomaterials.2011.07.025] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/08/2011] [Indexed: 11/30/2022]
Abstract
The capability to spatially control stem cell orientation and differentiation simultaneously using a combination of geometric cues that mimic structural aspects of native extracellular matrix (ECM) and biochemical cues such as ECM-bound growth factors (GFs) is important for understanding the organization and function of musculoskeletal tissues. Herein, oriented sub-micron fibers, which are morphologically similar to musculoskeletal ECM, were spatially patterned with GFs using an inkjet-based bioprinter to create geometric and biochemical cues that direct musculoskeletal cell alignment and differentiation in vitro in registration with fiber orientation and printed patterns, respectively. Sub-micron polystyrene fibers (diameter ~ 655 nm) were fabricated using a Spinneret-based Tunable Engineered Parameters (STEP) technique and coated with serum or fibrin. The fibers were subsequently patterned with tendon-promoting fibroblast growth factor-2 (FGF-2) or bone-promoting bone morphogenetic protein-2 (BMP-2) prior to seeding with mouse C2C12 myoblasts or C3H10T1/2 mesenchymal fibroblasts. Unprinted regions of STEP fibers showed myocyte differentiation while printed FGF-2 and BMP-2 patterns promoted tenocyte and osteoblast fates, respectively, and inhibited myocyte differentiation. Additionally, cells aligned along the fiber length. Functionalizing oriented sub-micron fibers with printed GFs provides instructive cues to spatially control cell fate and alignment to mimic native tissue organization and may have applications in regenerative medicine.
Collapse
Affiliation(s)
- Elmer D F Ker
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Chirieleison SM, Bissell TA, Scelfo CC, Anderson JE, Li Y, Koebler DJ, Deasy BM. Automated live cell imaging systems reveal dynamic cell behavior. Biotechnol Prog 2011; 27:913-24. [PMID: 21692197 DOI: 10.1002/btpr.629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 03/11/2011] [Indexed: 11/11/2022]
Abstract
Automated time-lapsed microscopy provides unique research opportunities to visualize cells and subcellular components in experiments with time-dependent parameters. As accessibility to these systems is increasing, we review here their use in cell science with a focus on stem cell research. Although the use of time-lapsed imaging to answer biological questions dates back nearly 150 years, only recently have the use of an environmentally controlled chamber and robotic stage controllers allowed for high-throughput continuous imaging over long periods at the cell and subcellular levels. Numerous automated imaging systems are now available from both companies that specialize in live cell imaging and from major microscope manufacturers. We discuss the key components of robots used for time-lapsed live microscopic imaging, and the unique data that can be obtained from image analysis. We show how automated features enhance experimentation by providing examples of uniquely quantified proliferation and migration live cell imaging data. In addition to providing an efficient system that drastically reduces man-hours and consumes fewer laboratory resources, this technology greatly enhances cell science by providing a unique dataset of temporal changes in cell activity.
Collapse
|
49
|
Liberski AR, Delaney JT, Schubert US. "One cell-one well": a new approach to inkjet printing single cell microarrays. ACS COMBINATORIAL SCIENCE 2011; 13:190-5. [PMID: 21395345 DOI: 10.1021/co100061c] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A new approach to prepare arrays of sessile droplets of living single cell cultures using a liquid hydrophobic barrier prevents the samples from dehydrating, and allows for spatially addressable arrays for statistical quantitative single cell studies. By carefully advancing a thin layer of mineral oil on the substrate over the droplets during the printing, dehydration of the droplets can be prevented, and the vitality of the cells can be maintained. The net result of this confluence of submerged cell culturing and inkjet printing is facile access to spatially addressable arrays of isolated single cells on surfaces. Such single cell arrays may be particularly useful as high-throughput tools in the rapidly emerging "omics" fields of cell biology.
Collapse
Affiliation(s)
- Albert R. Liberski
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich-Schiller-University Jena, Humboldtstrasse 10, Jena D-07743, Germany
- Dutch Polymer Institute (DPI), Post Office Box 902, Eindhoven 5600 AX, The Netherlands
| | - Joseph T. Delaney
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich-Schiller-University Jena, Humboldtstrasse 10, Jena D-07743, Germany
- Laboratory of Macromolecular Chemistry and Nanoscience, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
- Dutch Polymer Institute (DPI), Post Office Box 902, Eindhoven 5600 AX, The Netherlands
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich-Schiller-University Jena, Humboldtstrasse 10, Jena D-07743, Germany
- Laboratory of Macromolecular Chemistry and Nanoscience, Eindhoven University of Technology, P.O. Box 513, Eindhoven 5600 MB, The Netherlands
- Center for Nanoscience (CeNS), Ludwigs-Maximilians-University München, Amalienstrasse 54, München D-80799, Germany
- Dutch Polymer Institute (DPI), Post Office Box 902, Eindhoven 5600 AX, The Netherlands
| |
Collapse
|
50
|
Ker EDF, Chu B, Phillippi JA, Gharaibeh B, Huard J, Weiss LE, Campbell PG. Engineering spatial control of multiple differentiation fates within a stem cell population. Biomaterials 2011; 32:3413-22. [PMID: 21316755 DOI: 10.1016/j.biomaterials.2011.01.036] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Accepted: 01/13/2011] [Indexed: 12/22/2022]
Abstract
The capability to engineer microenvironmental cues to direct a stem cell population toward multiple fates, simultaneously, in spatially defined regions is important for understanding the maintenance and repair of multi-tissue units. We have previously developed an inkjet-based bioprinter to create patterns of solid-phase growth factors (GFs) immobilized to an extracellular matrix (ECM) substrate, and applied this approach to drive muscle-derived stem cells toward osteoblasts 'on-pattern' and myocytes 'off-pattern' simultaneously. Here this technology is extended to spatially control osteoblast, tenocyte and myocyte differentiation simultaneously. Utilizing immunofluorescence staining to identify tendon-promoting GFs, fibroblast growth factor-2 (FGF-2) was shown to upregulate the tendon marker Scleraxis (Scx) in C3H10T1/2 mesenchymal fibroblasts, C2C12 myoblasts and primary muscle-derived stem cells, while downregulating the myofibroblast marker α-smooth muscle actin (α-SMA). Quantitative PCR studies indicated that FGF-2 may direct stem cells toward a tendon fate via the Ets family members of transcription factors such as pea3 and erm. Neighboring patterns of FGF-2 and bone morphogenetic protein-2 (BMP-2) printed onto a single fibrin-coated coverslip upregulated Scx and the osteoblast marker ALP, respectively, while non-printed regions showed spontaneous myotube differentiation. This work illustrates spatial control of multi-phenotype differentiation and may have potential in the regeneration of multi-tissue units.
Collapse
Affiliation(s)
- Elmer D F Ker
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|