1
|
Crnic A, Rohringer S, Tyschuk T, Holnthoner W. Engineering blood and lymphatic microvascular networks. Atherosclerosis 2024; 393:117458. [PMID: 38320921 DOI: 10.1016/j.atherosclerosis.2024.117458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/18/2023] [Accepted: 01/16/2024] [Indexed: 02/08/2024]
Abstract
The human vasculature plays a crucial role in the blood supply of nearly all organs as well as the drainage of the interstitial fluid. Consequently, if these physiological systems go awry, pathological changes might occur. Hence, the regeneration of existing vessels, as well as approaches to engineer artificial blood and lymphatic structures represent current challenges within the field of vascular research. In this review, we provide an overview of both the vascular blood circulation and the long-time neglected but equally important lymphatic system, with regard to their organotypic vasculature. We summarize the current knowledge within the field of vascular tissue engineering focusing on the design of co-culture systems, thereby mainly discussing suitable cell types, scaffold design and disease models. This review will mainly focus on addressing those subjects concerning atherosclerosis. Moreover, current technological approaches such as vascular organ-on-a-chip models and microfluidic devices will be discussed.
Collapse
Affiliation(s)
- Aldina Crnic
- Ludwig-Boltzmann-Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstraße 13, 1020 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1020 Vienna, Austria
| | - Sabrina Rohringer
- Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1020 Vienna, Austria; Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Tatiana Tyschuk
- Ludwig-Boltzmann-Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstraße 13, 1020 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1020 Vienna, Austria
| | - Wolfgang Holnthoner
- Ludwig-Boltzmann-Institute for Traumatology, The Research Centre in Cooperation with AUVA, Donaueschingenstraße 13, 1020 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1020 Vienna, Austria.
| |
Collapse
|
2
|
Näf L, Miescher I, Pfuderer L, Schweizer TA, Brunner D, Dürig J, Gröninger O, Rieber J, Meier-Buergisser G, Spanaus K, Calcagni M, Bosshard PP, Achermann Y, Stark WJ, Buschmann J. Pro-angiogenic and antibacterial copper containing nanoparticles in PLGA/amorphous calcium phosphate bone nanocomposites. Heliyon 2024; 10:e27267. [PMID: 38486752 PMCID: PMC10937708 DOI: 10.1016/j.heliyon.2024.e27267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
Large bone defects after trauma demand for adequate bone substitutes. Bone void fillers should be antibacterial and pro-angiogenic. One viable option is the use of composite materials like the combination of PLGA and amorphous calcium phosphate (aCaP). Copper stimulates angiogenesis and has antibacterial qualities. Either copper oxide (CuO) nanoparticles (NPs) were therefore added to PLGA/aCaP/CuO in different concentrations (1, 5 and 10 w/w %) or copper-doped tricalcium phosphate NPs (TCP with 2% of copper) were electrospun into PLGA/CuTCP nanocomposites. Bi-layered nanocomposites of PLGA/aCaP with different copper NPs (CuO or TCP) and a second layer of pristine PLGA were fabricated. Two clinical bacterial isolates (Staphylococcus aureus and Staphylococcus epidermidis) were used to assess antibacterial properties of the copper-containing materials. For angiogenesis, the chorioallantoic membrane (CAM) assay of the chicken embryo was performed. The higher the CuO content, the higher were the antibacterial properties, with 10 % CuO reducing bacterial adhesion most effectively. Vessel and cell densities were highest in the 5 % CuO containing scaffolds, while tissue integration was more pronounced at lower CuO content. The PLGA/aCaP/CuO (1 % CuO) behaved similar like PLGA/CuTCP in all angiogenic and antibacterial readouts, based on the same copper fraction. We conclude that CuO NPs or CuTCP NPs are useful components to increase angiogenic properties of nanocomposites and at the same time exhibiting antibacterial characteristics.
Collapse
Affiliation(s)
- Lukas Näf
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Iris Miescher
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Lara Pfuderer
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Tiziano A. Schweizer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - David Brunner
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johannes Dürig
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Olivier Gröninger
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Julia Rieber
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Gabriella Meier-Buergisser
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Katharina Spanaus
- Clinical Chemistry, University Hospital Zurich, 8001, Zurich, Switzerland
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Philipp P. Bosshard
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Yvonne Achermann
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Wendelin J. Stark
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Johanna Buschmann
- Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
3
|
Microfluidic 3D Platform to Evaluate Endothelial Progenitor Cell Recruitment by Bioactive Materials. Acta Biomater 2022; 151:264-277. [PMID: 35981686 DOI: 10.1016/j.actbio.2022.08.019] [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: 02/02/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 12/30/2022]
Abstract
Most of the conventional in vitro models to test biomaterial-driven vascularization are too simplistic to recapitulate the complex interactions taking place in the actual cell microenvironment, which results in a poor prediction of the in vivo performance of the material. However, during the last decade, cell culture models based on microfluidic technology have allowed attaining unprecedented levels of tissue biomimicry. In this work, we propose a microfluidic-based 3D model to evaluate the effect of bioactive biomaterials capable of releasing signalling cues (such as ions or proteins) in the recruitment of endogenous endothelial progenitor cells, a key step in the vascularization process. The usability of the platform is demonstrated using experimentally-validated finite element models and migration and proliferation studies with rat endothelial progenitor cells (rEPCs) and bone marrow-derived rat mesenchymal stromal cells (BM-rMSCs). As a proof of concept of biomaterial evaluation, the response of rEPCs to an electrospun composite made of polylactic acid with calcium phosphates nanoparticles (PLA+CaP) was compared in a co-culture microenvironment with BM-rMSC to a regular PLA control. Our results show a significantly higher rEPCs migration and the upregulation of several pro-inflammatory and proangiogenic proteins in the case of the PLA+CaP. The effects of osteopontin (OPN) on the rEPCs migratory response were also studied using this platform, suggesting its important role in mediating their recruitment to a calcium-rich microenvironment. This new tool could be applied to screen the capacity of a variety of bioactive scaffolds to induce vascularization and accelerate the preclinical testing of biomaterials. STATEMENT OF SIGNIFICANCE: : For many years researchers have used neovascularization models to evaluate bioactive biomaterials both in vitro, with low predictive results due to their poor biomimicry and minimal control over cell cues such as spatiotemporal biomolecule signaling, and in vivo models, presenting drawbacks such as being highly costly, time-consuming, poor human extrapolation, and ethically controversial. We describe a compact microphysiological platform designed for the evaluation of proangiogenesis in biomaterials through the quantification of the level of sprouting in a mimicked endothelium able to react to gradients of biomaterial-released signals in a fibrin-based extracellular matrix. This model is a useful tool to perform preclinical trustworthy studies in tissue regeneration and to better understand the different elements involved in the complex process of vascularization.
Collapse
|
4
|
Fang Y, Sun W, Zhang T, Xiong Z. Recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps: A review. Biomaterials 2021; 280:121298. [PMID: 34864451 DOI: 10.1016/j.biomaterials.2021.121298] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022]
Abstract
The field of cardiac tissue engineering has advanced over the past decades; however, most research progress has been limited to engineered cardiac tissues (ECTs) at the microscale with minimal geometrical complexities such as 3D strips and patches. Although microscale ECTs are advantageous for drug screening applications because of their high-throughput and standardization characteristics, they have limited translational applications in heart repair and the in vitro modeling of cardiac function and diseases. Recently, researchers have made various attempts to construct engineered cardiac pumps (ECPs) such as chambered ventricles, recapitulating the geometrical complexity of the native heart. The transition from microscale ECTs to ECPs at a translatable scale would greatly accelerate their translational applications; however, researchers are confronted with several major hurdles, including geometrical reconstruction, vascularization, and functional maturation. Therefore, the objective of this paper is to review the recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps. We first review the bioengineering approaches to fabricate ECPs, and then emphasize the unmatched potential of 3D bioprinting techniques. We highlight key advances in bioprinting strategies with high cell density as researchers have begun to realize the critical role that the cell density of non-proliferative cardiomyocytes plays in the cell-cell interaction and functional contracting performance. We summarize the current approaches to engineering vasculatures both at micro- and meso-scales, crucial for the survival of thick cardiac tissues and ECPs. We showcase a variety of strategies developed to enable the functional maturation of cardiac tissues, mimicking the in vivo environment during cardiac development. By highlighting state-of-the-art research, this review offers personal perspectives on future opportunities and trends that may bring us closer to the promise of functional ECPs.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| |
Collapse
|
5
|
Schott NG, Stegemann JP. Coculture of Endothelial and Stromal Cells to Promote Concurrent Osteogenesis and Vasculogenesis. Tissue Eng Part A 2021; 27:1376-1386. [PMID: 33599160 PMCID: PMC8827126 DOI: 10.1089/ten.tea.2020.0330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
A key challenge in the treatment of large bone defects is the need to provide an adequate and stable vascular supply as new tissue develops. Bone tissue engineering applies selected biomaterials and cell types to create an environment that promotes tissue formation, maturation, and remodeling. Mesenchymal stromal cells (MSCs) have been widely used in these strategies because of their established effects on bone formation, and their ability to act as stabilizing pericytes that support vascular regeneration by endothelial cells (ECs). However, the creation of vascularized bone tissue in vitro requires coupling of osteogenesis and vasculogenesis in a three-dimensional (3D) biomaterial environment. In the present study, 3D fibrin hydrogels containing MSCs and ECs were prevascularized in vitro for 7 days to create an endothelial network in the matrix, and were subsequently cultured for a further 14 days under either continued vasculogenic stimulus, a combination of vasculogenic and osteogenic (hybrid) stimulus, or only osteogenic stimulus. It was found that ECs produced robust vessel networks in 3D fibrin matrices over 7 days of culture, and these networks continued to expand over the 14-day treatment period under vasculogenic conditions. Culture in hybrid medium resulted in maintenance of vessel networks for 14 days, while osteogenic culture abrogated vessel formation. These trends were mirrored in data representing overall cell viability and cell number in the 3D fibrin constructs. MSCs were found to colocalize with EC networks under vasculogenic and hybrid conditions, suggesting pericyte-like function. The bone marker alkaline phosphatase increased over time in hybrid and osteogenic media, but mineral deposition was evident only under purely osteogenic conditions. These results suggest that hybrid media compositions can support some aspects of multiphase tissue formation, but that alternative strategies are needed to obtain robust, concomitant vascularization, and osteogenesis in engineered tissues in vitro. Impact statement The combined use of mesenchymal stromal cells (MSCs) and endothelial cells to concomitantly produce mature bone and a nourishing vasculature is a promising tissue engineering approach to treating large bone defects. However, it is challenging to create and maintain vascular networks in the presence of osteogenic cues. This study used a 3D fibrin matrix to demonstrate that prevascularization of the construct can lead to maintenance of vessel structures over time, but that osteogenesis is compromised under these conditions. This work illuminates the capacity of MSCs to serve as both supportive pericytes and as osteoprogenitor cells, and motivates new strategies for coupling osteogenesis and vasculogenesis in engineered bone tissues.
Collapse
Affiliation(s)
- Nicholas G. Schott
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Tabatabaei F, Rasoulianboroujeni M, Yadegari A, Tajik S, Moharamzadeh K, Tayebi L. Osteo-mucosal engineered construct: In situ adhesion of hard-soft tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112255. [PMID: 34474817 DOI: 10.1016/j.msec.2021.112255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The aim of this work was to combine engineered hard and soft tissue, adopting a new method for interfacial adhesion of osteo-mucosal construct. We hypothesized that the chemical procedure involved in this method not only adheres the components, but also improves the cell growth inside them. METHODS 3D-printed functionally-graded porous hard-tissue scaffolds were characterized, functionalized by aminolysis and tyrosinase, and accommodated by human osteoblast cells. Introducing amino groups through aminolysis and inducing dopaquinones by tyrosinase can take part in the Michael additions to cause the adhesion. Subsequently, fully-differentiated engineered oral mucosa was formed directly on the surface of hard tissue. Constructs were assessed in term of morphology, structure, chemical composition, histology, and cytocompatibility. Interfacial adhesion was compared to a control group prepared by using a biological glue for the attachment of the soft and hard tissues. RESULTS The data confirmed higher proliferation of osteoblast cells via aminolysis and improved osteoblast cells distribution and differentiation by incorporation of tyrosinase in collagen. There was evidence of multilayered, stratified epithelium on the osteo-mucosal model with viable fibroblasts and osteoblasts within the lamina propria and bone tissue layers. Our method of adhesion resulted in cohesive debonding within the engineered soft tissue; while in the control group, adhesive debonding and complete separation of the oral mucosa from the hard tissue was observed. Although the shear strength of the osteo-mucosal model (157.6 kDa ± 25.1) was slightly higher than that of the control group (149.4 kDa ± 23.1), there was no statistically significant difference between them (p > 0.05). However, the advantage of our in situ adhesion approach is the absence of a barrier like glue which can disrupt direct cellular communications between tissues. SIGNIFICANCE This study provides a novel method of directly combining tissue-engineered human bone with oral mucosa, which has the potential to improve cell-ingrowth and tissue integration. This engineered tissue construct, after further optimization, can be used clinically as a graft material in various oral surgeries and can also be employed as an in vitro model to investigate many aspects of oral diseases and examine dental materials and oral health care products as a replacement of in vivo models.
Collapse
Affiliation(s)
| | | | - Amir Yadegari
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Sanaz Tajik
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine (HBMCDM), Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, United Arab Emirates; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA.
| |
Collapse
|
7
|
Kim HD, Hong X, An YH, Park MJ, Kim DG, Greene AK, Padwa BL, Hwang NS, Lin RZ, Melero-Martin JM. A Biphasic Osteovascular Biomimetic Scaffold for Rapid and Self-Sustained Endochondral Ossification. Adv Healthc Mater 2021; 10:e2100070. [PMID: 33882194 PMCID: PMC8273143 DOI: 10.1002/adhm.202100070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/23/2021] [Indexed: 12/14/2022]
Abstract
Regeneration of large bones remains a challenge in surgery. Recent developmental engineering efforts aim to recapitulate endochondral ossification (EO), a critical step in bone formation. However, this process entails the condensation of mesenchymal stem cells (MSCs) into cartilaginous templates, which requires long-term cultures and is challenging to scale up. Here, a biomimetic scaffold is developed that allows rapid and self-sustained EO without initial hypertrophic chondrogenesis. The design comprises a porous chondroitin sulfate cryogel decorated with whitlockite calcium phosphate nanoparticles, and a soft hydrogel occupying the porous space. This composite scaffold enables human endothelial colony-forming cells (ECFCs) and MSCs to rapidly assemble into osteovascular niches in immunodeficient mice. These niches contain ECFC-lined blood vessels and perivascular MSCs that differentiate into RUNX2+ OSX+ pre-osteoblasts after one week in vivo. Subsequently, multiple ossification centers are formed, leading to de novo bone tissue formation by eight weeks, including mature human OCN+ OPN+ osteoblasts, collagen-rich mineralized extracellular matrix, hydroxyapatite, osteoclast activity, and gradual mechanical competence. The early establishment of blood vessels is essential, and grafts that do not contain ECFCs fail to produce osteovascular niches and ossification centers. The findings suggest a novel bioengineering approach to recapitulate EO in the context of human bone regeneration.
Collapse
Affiliation(s)
- Hwan D. Kim
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea (H.D.K current address)
| | - Xuechong Hong
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Young-Hyeon An
- School of Chemical and Biological Engineering, BioMAX Institute, Institute of Chemical Processes, Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mihn Jeong Park
- School of Chemical and Biological Engineering, BioMAX Institute, Institute of Chemical Processes, Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH 43210, USA
| | - Arin K. Greene
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Bonnie L. Padwa
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Nathaniel S. Hwang
- School of Chemical and Biological Engineering, BioMAX Institute, Institute of Chemical Processes, Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Juan M. Melero-Martin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
8
|
Chakka JL, Acri T, Laird NZ, Zhong L, Shin K, Elangovan S, Salem AK. Polydopamine functionalized VEGF gene-activated 3D printed scaffolds for bone regeneration. RSC Adv 2021; 11:13282-13291. [PMID: 35423856 PMCID: PMC8697638 DOI: 10.1039/d1ra01193f] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/27/2021] [Indexed: 12/20/2022] Open
Abstract
Bone is a highly vascularized organ and the formation of new blood vessels is essential to regenerate large critical bone defects. In this study, polylactic acid (PLA) scaffolds of 20-80% infill were three-dimensionally (3D) printed using a fused deposition modeling based 3D printer. The PLA scaffolds were coated with polydopamine (PDA) and then were surface-functionalized with polyethyleneimine (PEI) and VEGF-encoding plasmid DNA (pVEGF) nanoplexes (PLA-PDA-PEI-pVEGF). The PLA-PDA-PEI-pVEGF scaffolds with 40% infill demonstrated higher encapsulation efficiency and sustained release of pVEGF than scaffolds with 20, 60 and 80% infill and were therefore used for in vitro and in vivo studies. The PLA-PDA-PEI-pVEGF increased the translation and secretion of VEGF and BMP-2. The PLA-PDA-PEI-pVEGF also yielded a 2- and 4.5-fold change in VEGF and osteocalcin gene expression in vitro, respectively. A tube formation assay using human umbilical vascular endothelial cells (HUVECs) showed a significant increase in tube length when exposed to the PLA-PDA-PEI-pVEGF scaffold, in comparison to PLA and PLA-PDA scaffolds. The PLA-PDA-PEI-pVEGF scaffold in an in vivo rat calvarial critical bone defect model demonstrated 1.6-fold higher new bone formation compared to the PLA-PDA scaffold. H&E and Masson's trichrome staining of bone sections also revealed that the PLA-PDA-PEI-pVEGF scaffold facilitated the formation of more blood vessels in the newly formed bone compared to the PLA and PLA-PDA scaffold groups. Thus, PLA-PDA-PEI-pVEGF might be a potential 3D printed gene activated scaffold for bone regeneration in clinical situations.
Collapse
Affiliation(s)
- Jaidev L Chakka
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| | - Timothy Acri
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| | - Noah Z Laird
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| | - Ling Zhong
- Department of Experimental Research, Sun Yat-sen University Guangzhou PR China
| | - Kyungsup Shin
- Department of Orthodontics, College of Dentistry and Dental Clinics, University of Iowa Iowa City IA-52242 USA
| | - Satheesh Elangovan
- Department of Periodontics, College of Dentistry and Dental Clinics, University of Iowa Iowa City IA-52242 USA
| | - Aliasger K Salem
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| |
Collapse
|
9
|
Meijer EM, van Dijk CGM, Kramann R, Verhaar MC, Cheng C. Implementation of Pericytes in Vascular Regeneration Strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1-21. [PMID: 33231500 DOI: 10.1089/ten.teb.2020.0229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For the survival and integration of complex large-sized tissue-engineered (TE) organ constructs that exceed the maximal nutrients and oxygen diffusion distance required for cell survival, graft (pre)vascularization to ensure medium or blood supply is crucial. To achieve this, the morphology and functionality of the microcapillary bed should be mimicked by incorporating vascular cell populations, including endothelium and mural cells. Pericytes play a crucial role in microvascular function, blood vessel stability, angiogenesis, and blood pressure regulation. In addition, tissue-specific pericytes are important in maintaining specific functions in different organs, including vitamin A storage in the liver, renin production in the kidneys and maintenance of the blood-brain-barrier. Together with their multipotential differentiation capacity, this makes pericytes the preferred cell type for application in TE grafts. The use of a tissue-specific pericyte cell population that matches the TE organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)-vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts. Impact statement The use of a tissue-specific pericyte cell population that matches the tissue-engineered (TE) organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts.
Collapse
Affiliation(s)
- Elana M Meijer
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christian G M van Dijk
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rafael Kramann
- Division of Nephrology and Institute of Experimental Medicine and Systems Biology, University Hospital RWTH Aachen, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Experimental Cardiology, Department of Cardiology, Thorax Center Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
10
|
Liu H, Du Y, Yang G, Hu X, Wang L, Liu B, Wang J, Zhang S. Delivering Proangiogenic Factors from 3D-Printed Polycaprolactone Scaffolds for Vascularized Bone Regeneration. Adv Healthc Mater 2020; 9:e2000727. [PMID: 32743958 DOI: 10.1002/adhm.202000727] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/10/2020] [Indexed: 01/04/2023]
Abstract
Natural bone is a highly vascularized tissue that relies on the vasculature for blood and nutrients supply to maintain skeletal integrity. Inadequacy of neovascularization may compromise the tissue ingrowth to the implanted scaffolds, and eventually results in failure for the repair. To tackle this issue and enhance self-vascularized bone regeneration, herein a 3D biomimetic selective lasersintering (SLS) derived scaffold, with an angiogenic growth factor immobilized on its surface, that can be released in a controlled manner is proposed. To this end, a porous polycaprolactone/hydroxyapatite (PCL/HA) scaffold is prepared via the SLS technique, which is further modified with vascular endothelial growth factor (VEGF) by coprecipitation with apatite. The resultant scaffold (PCL/HA/VEGF) has an excellent cytocompatibility, and subcutaneous implantation experiment shows that the VEGF-loaded scaffold significantly enhances the blood vessel formation compared with the VEGF-free control. It is further demonstrated that the PCL/HA/VEGF scaffold is able to enhance the in vivo bone regeneration in a rat cranial defect model. Taken together, the current study provides not only a feasible and promising scaffold candidate to enhance the vascularized bone regeneration, but also a general strategy to overcome the inadequate vascularization issue for the repair of other tissue and organs.
Collapse
Affiliation(s)
- Haoming Liu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yingying Du
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Gaojie Yang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xixi Hu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bin Liu
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100037, China
| | - Jianglin Wang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shengmin Zhang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
11
|
Carvalho DJ, Feijão T, Neves MI, da Silva RMP, Barrias C. Directed self-assembly of spheroids into modular vascular beds for engineering large tissue constructs. Biofabrication 2020; 13. [PMID: 33147579 DOI: 10.1088/1758-5090/abc790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/04/2020] [Indexed: 12/12/2022]
Abstract
Spheroids can be used as building-blocks for bottom-up generation of artificial vascular beds, but current biofabrication strategies are often time-consuming and complex. Also, pre-optimization of single spheroid properties is often neglected. Here, we report a simple setup for rapid biomanufacturing of spheroid-based patch-like vascular beds. Prior to patch assembly, spheroids combining mesenchymal stem/stromal cells (MSC) and outgrowth endothelial cells (OEC) at different ratios (10:1; 5:1; 1:1; 1:5) were formed in non-adhesive microwells and monitored along 7 days. Optimal OEC retention and organization was observed at 1:1 MSC/OEC ratio. Dynamic remodelling of spheroids led to changes in both cellular and extracellular matrix components (ECM) over time. Some OEC formed internal clusters, while others organized into a peripheral monolayer, stabilized by ECM and pericyte-like cells, with concomitant increase in surface stiffness. Along spheroid culture, OEC switched from an active to a quiescent state, and their endothelial sprouting potential was significantly abrogated, suggesting that immature spheroids may be more therapeutically relevant. Non-adhesive moulds were subsequently used for triggering rapid, one-step, spheroid formation/fusion into square-shaped patches, with spheroids uniformly interspaced via a thin cell layer. The high surface area, endothelial sprouting potential, and scalability of the developed spheroid-based patches make them stand out as artificial vascular beds for modular engineering of large tissue constructs.
Collapse
Affiliation(s)
- Daniel Jose Carvalho
- Bioengineered 3D microenvironments, Instituto Nacional de Engenharia Biomedica, Porto, Porto, PORTUGAL
| | - Tália Feijão
- Universidade do Porto Instituto de Investigação e Inovação em Saúde, Porto, Porto, PORTUGAL
| | - Mariana Isabel Neves
- Universidade do Porto Instituto de Investigação e Inovação em Saúde, Porto, Porto, PORTUGAL
| | - Ricardo M P da Silva
- Universidade do Porto Instituto de Investigação e Inovação em Saúde, Porto, Porto, PORTUGAL
| | - Cristina Barrias
- Instituto Engenharia Biomedica Laboratorio de Biomaterials, Universidade do Porto, Porto, PORTUGAL
| |
Collapse
|
12
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
13
|
Repair of Bone Defects With Endothelial Progenitor Cells and Bone Marrow-Derived Mesenchymal Stem Cells With Tissue-Engineered Bone in Rabbits. Ann Plast Surg 2020; 85:430-436. [PMID: 32931683 DOI: 10.1097/sap.0000000000002454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE This study aimed to investigate the repair of bone defects in rabbits with tissue-engineered bones using cocultured endothelial progenitor cells (EPCs) and bone marrow mesenchymal stem cells (BMSCs) as seeding cells. METHODS Endothelial progenitor cells and BMSCs were isolated and purified from the peripheral blood and bone marrow, respectively, of New Zealand rabbits. The third passage of BMSCs was cultured alone or with EPCs. Cells were characterized using specific markers and then seeded on partially deproteinized biologic bones from pigs as a scaffold. The engineered bones were used to repair bone defects in rabbits. Hematoxylin and eosin and Masson staining were performed to examine vascularization and osteogenesis in the engineered bone. RESULTS The cocultured EPCs and BMSCs grew well on the surface of the scaffold. Compared with monocultured BMSCs, cocultured EPCs and BMSCs promoted the formation of blood vessels and bone on the scaffold, in addition to accelerating the repair of bone defects. The collagen content was significantly increased in the scaffold with cocultured EPCs and BMSCs, compared with the scaffold seeded with mono-cultured BMSCs. CONCLUSIONS Tissue-engineered bones seeded with cocultured EPCs and BMSCs may be used effectively for the repair of bone defects.
Collapse
|
14
|
Sawyer SW, Zhang K, Horton JA, Soman P. Perfusion-based co-culture model system for bone tissue engineering. AIMS BIOENGINEERING 2020; 7:91-105. [PMID: 33163623 PMCID: PMC7643915 DOI: 10.3934/bioeng.2020009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In this work, we report on a perfusion-based co-culture system that could be used for bone tissue engineering applications. The model system is created using a combination of Primary Human Umbilical Vein Endothelial Cells (HUVECs) and osteoblast-like Saos-2 cells encapsulated within a Gelatin Methacrylate (GelMA)-collagen hydrogel blend contained within 3D printed, perfusable constructs. The constructs contain dual channels, within a custom-built bioreactor, that were perfused with osteogenic media for up to two weeks in order to induce mineral deposition. Mineral deposition in constructs containing only HUVECs, only Saos-2 cells, or a combination thereof was quantified by microCT to determine if the combination of endothelial cells and bone-like cells increased mineral deposition. Histological and fluorescent staining was used to verify mineral deposition and cellular function both along and between the perfused channels. While there was not a quantifiable difference in the amount of mineral deposited in Saos-2 only versus Saos-2 plus HUVEC samples, the location of the deposited mineral differed dramatically between the groups and indicated that the addition of HUVECs within the GelMA matrix allowed Saos-2 cells, in diffusion limited regions of the construct, to deposit bone mineral. This work serves as a model on how to create perfusable bone tissue engineering constructs using a combination of 3D printing and cellular co-cultures.
Collapse
Affiliation(s)
- Stephen W. Sawyer
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
| | - Kairui Zhang
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
| | - Jason A. Horton
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Pranav Soman
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
| |
Collapse
|
15
|
Shpichka AI, Konarev PV, Efremov YM, Kryukova AE, Aksenova NA, Kotova SL, Frolova AA, Kosheleva NV, Zhigalina OM, Yusupov VI, Khmelenin DN, Koroleva A, Volkov VV, Asadchikov VE, Timashev PS. Digging deeper: structural background of PEGylated fibrin gels in cell migration and lumenogenesis. RSC Adv 2020; 10:4190-4200. [PMID: 35495227 PMCID: PMC9049040 DOI: 10.1039/c9ra08169k] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/19/2019] [Indexed: 12/02/2022] Open
Abstract
Fibrin is a well-known tool in tissue engineering, but the structure of its modifications created to improve its properties remains undiscussed despite its importance, e.g. in designing biomaterials that ensure cell migration and lumenogenesis. We sought to uncover the structural aspects of PEGylated fibrin hydrogels shown to contribute to angiogenesis. The analysis of the small-angle X-ray scattering (SAXS) data and ab initio modeling revealed that the PEGylation of fibrinogen led to the formation of oligomeric species, which are larger at a higher PEG : fibrinogen molar ratio. The improvement of optical properties was provided by the decrease in aggregates' sizes and also by retaining the bound water. Compared to the native fibrin, the structure of the 5 : 1 PEGylated fibrin gel consisted of homogenously distributed flexible fibrils with a smaller space between them. Moreover, as arginylglycylaspartic acid (RGD) sites may be partly bound to PEG-NHS or masked because of the oligomerization, the number of adhesion sites may be slightly reduced that may provide the better cell migration and formation of continuous capillary-like structures.
Collapse
Affiliation(s)
- A I Shpichka
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
| | - P V Konarev
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
- National Research Center "Kurchatov Institute" Moscow Russia
| | - Yu M Efremov
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
| | - A E Kryukova
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
- National Research Center "Kurchatov Institute" Moscow Russia
| | - N A Aksenova
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
- Department of Polymers and Composites, N. N. Semenov Institute of Chemical Physics Moscow Russia
| | - S L Kotova
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
- Department of Polymers and Composites, N. N. Semenov Institute of Chemical Physics Moscow Russia
| | - A A Frolova
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
| | - N V Kosheleva
- FSBSI 'Institute of General Pathology and Pathophysiology' Moscow Russia
- Faculty of Biology, Lomonosov Moscow State University Moscow Russia
| | - O M Zhigalina
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
- Bauman Moscow State Technical University Moscow Russia
| | - V I Yusupov
- Institute of Photon Technologies, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - D N Khmelenin
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - A Koroleva
- Laser Zentrum Hannover e. V. Hannover Germany
| | - V V Volkov
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - V E Asadchikov
- A. V. Shubnikov Institute of Crystallography, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| | - P S Timashev
- Institute for Regenerative Medicine, Sechenov University 2-8 Trubetskaya St. Moscow Russia 119991 +7 495 6091400 ext. 3638
- Department of Polymers and Composites, N. N. Semenov Institute of Chemical Physics Moscow Russia
- Institute of Photon Technologies, Federal Scientific Research Center "Crystallography and Photonics" RAS Moscow Russia
| |
Collapse
|
16
|
Yang G, Mahadik B, Choi JY, Fisher JP. Vascularization in tissue engineering: fundamentals and state-of-art. ACTA ACUST UNITED AC 2020; 2. [PMID: 34308105 DOI: 10.1088/2516-1091/ab5637] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vascularization is among the top challenges that impede the clinical application of engineered tissues. This challenge has spurred tremendous research endeavor, defined as vascular tissue engineering (VTE) in this article, to establish a pre-existing vascular network inside the tissue engineered graft prior to implantation. Ideally, the engineered vasculature can be integrated into the host vasculature via anastomosis to supply nutrient to all cells instantaneously after surgery. Moreover, sufficient vascularization is of great significance in regenerative medicine from many other perspectives. Due to the critical role of vascularization in successful tissue engineering, we aim to provide an up-to-date overview of the fundamentals and VTE strategies in this article, including angiogenic cells, biomaterial/bio-scaffold design and bio-fabrication approaches, along with the reported utility of vascularized tissue complex in regenerative medicine. We will also share our opinion on the future perspective of this field.
Collapse
Affiliation(s)
- Guang Yang
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America.,Center for Engineering Complex Tissues, University of Maryland, College Park, MD, United States of America
| | - Bhushan Mahadik
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America.,Center for Engineering Complex Tissues, University of Maryland, College Park, MD, United States of America
| | - Ji Young Choi
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America
| | - John P Fisher
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States of America.,Center for Engineering Complex Tissues, University of Maryland, College Park, MD, United States of America
| |
Collapse
|
17
|
Functionalization of Silk Fibers by PDGF and Bioceramics for Bone Tissue Regeneration. COATINGS 2019. [DOI: 10.3390/coatings10010008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bone regeneration is a complex, well-organized physiological process of bone formation observed during normal fracture healing and involved in continuous remodeling throughout adult life. An ideal medical device for bone regeneration requires interconnected pores within the device to allow for penetration of blood vessels and cells, enabling material biodegradation and bone ingrowth. Additional mandatory characteristics include an excellent resorption rate, a 3D structure similar to natural bone, biocompatibility, and customizability to multiple patient-specific geometries combined with adequate mechanical strength. Therefore, endless silk fibers were spun from native silk solution isolated from silkworm larvae and functionalized with osteoconductive bioceramic materials. In addition, transgenic silkworms were generated to functionalize silk proteins with human platelet-derived growth factor (hPDGF). Both, PDGF-silk and bioceramic modified silk were then assembled into 3D textile implants using an additive manufacturing approach. Textile implants were characterized in terms of porosity, compressive strength, and cyclic load. In addition, osteogenic differentiation of mesenchymal stem cells was evaluated. Silk fiber-based 3D textile implants showed good cytocompatibility and stem cells cultured on bioceramic material functionalized silk implants were differentiating into bone cells. Thus, functionalized 3D interconnected porous textile scaffolds were shown to be promising biomaterials for bone regeneration.
Collapse
|
18
|
Andrew TW, Kanapathy M, Murugesan L, Muneer A, Kalaskar D, Atala A. Towards clinical application of tissue engineering for erectile penile regeneration. Nat Rev Urol 2019; 16:734-744. [PMID: 31649327 DOI: 10.1038/s41585-019-0246-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2019] [Indexed: 11/09/2022]
Abstract
Penile wounds after traumatic and surgical amputation require reconstruction in the form of autologous tissue transfers. However, currently used techniques are associated with high infection rates, implant erosion and donor site morbidity. The use of tissue-engineered neocorpora provides an alternative treatment option. Contemporary tissue-engineering strategies enable the seeding of a biomaterial scaffold and subsequent implantation to construct a neocorpus. Tissue engineering of penile tissue should focus on two main strategies: first, correcting the volume deficit for structural integrity in order to enable urinary voiding in the standing position and second, achieving erectile function for sexual activity. The functional outcomes of the neocorpus can be addressed by optimizing the use of stem cells and scaffolds, or alternatively, the use of gene therapy. Current research in penile tissue engineering is largely restricted to rodent and rabbit models, but the use of larger animal models should be considered as a better representation of the anatomical and physiological function in humans. The development of a cell-seeded scaffold to achieve and maintain erection continues to be a considerable challenge in humans. However, advances in penile tissue engineering show great promise and, in combination with gene therapy and surgical techniques, have the potential to substantially improve patient outcomes.
Collapse
Affiliation(s)
- Tom W Andrew
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK.
| | - Muholan Kanapathy
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK
| | - Log Murugesan
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK
| | - Asif Muneer
- Department of Urology, University College London Hospital, London, UK
| | - Deepak Kalaskar
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, USA
| |
Collapse
|
19
|
Tam WL, Luyten FP, Roberts SJ. From skeletal development to the creation of pluripotent stem cell-derived bone-forming progenitors. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0218. [PMID: 29786553 DOI: 10.1098/rstb.2017.0218] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Bone has many functions. It is responsible for protecting the underlying soft organs, it allows locomotion, houses the bone marrow and stores minerals such as calcium and phosphate. Upon damage, bone tissue can efficiently repair itself. However, healing is hampered if the defect exceeds a critical size and/or is in compromised conditions. The isolation or generation of bone-forming progenitors has applicability to skeletal repair and may be used in tissue engineering approaches. Traditionally, bone engineering uses osteochondrogenic stem cells, which are combined with scaffold materials and growth factors. Despite promising preclinical data, limited translation towards the clinic has been observed to date. There may be several reasons for this including the lack of robust cell populations with favourable proliferative and differentiation capacities. However, perhaps the most pertinent reason is the failure to produce an implant that can replicate the developmental programme that is observed during skeletal repair. Pluripotent stem cells (PSCs) can potentially offer a solution for bone tissue engineering by providing unlimited cell sources at various stages of differentiation. In this review, we summarize key embryonic signalling pathways in bone formation coupled with PSC differentiation strategies for the derivation of bone-forming progenitors.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Wai Long Tam
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Frank P Luyten
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Scott J Roberts
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium .,Bone Therapeutic Area, UCB Pharma, 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| |
Collapse
|
20
|
Liu X, Jakus AE, Kural M, Qian H, Engler A, Ghaedi M, Shah R, Steinbacher DM, Niklason LE. Vascularization of Natural and Synthetic Bone Scaffolds. Cell Transplant 2018; 27:1269-1280. [PMID: 30008231 PMCID: PMC6434463 DOI: 10.1177/0963689718782452] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Vascularization of engineered bone tissue is critical for ensuring its survival after implantation. In vitro pre-vascularization of bone grafts with endothelial cells is a promising strategy to improve implant survival. In this study, we pre-cultured human smooth muscle cells (hSMCs) on bone scaffolds for 3 weeks followed by seeding of human umbilical vein endothelial cells (HUVECs), which produced a desirable environment for microvasculature formation. The sequential cell-seeding protocol was successfully applied to both natural (decellularized native bone, or DB) and synthetic (3D-printed Hyperelastic "Bone" scaffolds, or HB) scaffolds, demonstrating a comprehensive platform for developing natural and synthetic-based in vitro vascularized bone grafts. Using this sequential cell-seeding process, the HUVECs formed lumen structures throughout the DB scaffolds as well as vascular tissue bridging 3D-printed fibers within the HB. The pre-cultured hSMCs were essential for endothelial cell (EC) lumen formation within DB scaffolds, as well as for upregulating EC-specific gene expression of HUVECs grown on HB scaffolds. We further applied this co-culture protocol to DB scaffolds using a perfusion bioreactor, to overcome the limitations of diffusive mass transport into the interiors of the scaffolds. Compared with static culture, panoramic histological sections of DB scaffolds cultured in bioreactors showed improved cellular density, as well as a nominal increase in the number of lumen structures formed by ECs in the interior regions of the scaffolds. In conclusion, we have demonstrated that the sequential seeding of hSMCs and HUVECs can serve to generate early microvascular networks that could further support the in vitro tissue engineering of naturally or synthetically derived bone grafts and in both random (DB) and ordered (HB) pore networks. Combined with the preliminary bioreactor study, this process also shows potential to generate clinically sized, vascularized bone scaffolds for tissue and regenerative engineering.
Collapse
Affiliation(s)
- Xi Liu
- 1 Plastic and Reconstructive Surgery, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Adam E Jakus
- 2 Department of Materials Science and Engineering, McCormick School of Engineering, Northwestern University, Chicago, IL, USA.,3 Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - Mehmet Kural
- 4 Department of Anesthesiology, Yale University, New Haven, CT, USA.,5 Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Hong Qian
- 4 Department of Anesthesiology, Yale University, New Haven, CT, USA.,5 Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Alexander Engler
- 4 Department of Anesthesiology, Yale University, New Haven, CT, USA.,5 Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Mahboobe Ghaedi
- 4 Department of Anesthesiology, Yale University, New Haven, CT, USA.,5 Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ramille Shah
- 2 Department of Materials Science and Engineering, McCormick School of Engineering, Northwestern University, Chicago, IL, USA.,3 Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA.,6 Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Chicago, IL, USA.,7 Division of Organ Transplantation, Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Derek M Steinbacher
- 1 Plastic and Reconstructive Surgery, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Laura E Niklason
- 4 Department of Anesthesiology, Yale University, New Haven, CT, USA.,5 Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
21
|
Kawecki F, Clafshenkel WP, Auger FA, Bourget JM, Fradette J, Devillard R. Self-assembled human osseous cell sheets as living biopapers for the laser-assisted bioprinting of human endothelial cells. Biofabrication 2018; 10:035006. [PMID: 29638221 DOI: 10.1088/1758-5090/aabd5b] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A major challenge during the engineering of voluminous bone tissues is to maintain cell viability in the central regions of the construct. In vitro prevascularization of bone substitutes relying on endothelial cell bioprinting has the potential to resolve this issue and to replicate the native bone microvasculature. Laser-assisted bioprinting (LAB) commonly uses biological layers of hydrogel, called 'biopapers', to support patterns of printed cells and constitute the basic units of the construct. The self-assembly approach of tissue engineering allows the production of biomimetic cell-derived bone extracellular matrix including living cells. We hypothesized that self-assembled osseous sheets can serve as living biopapers to support the LAB of human endothelial cells and thus guide tubule-like structure formation. Human umbilical vein endothelial cells were bioprinted on the surface of the biopapers following a predefined pattern of lines. The osseous biopapers showed relevant matrix mineralization and pro-angiogenic hallmarks. Our results revealed that formation of tubule-like structures was favored when the cellular orientation within the biopaper was parallel to the printed lines. Altogether, we validated that human osseous cell sheets can be used as biopapers for LAB, allowing the production of human prevascularized cell-based osseous constructs that can be relevant for autologous bone repair applications.
Collapse
Affiliation(s)
- F Kawecki
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center-Université Laval, QC, Canada. Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | | | | | | | | | | |
Collapse
|
22
|
Pirosa A, Gottardi R, Alexander PG, Tuan RS. Engineering in-vitro stem cell-based vascularized bone models for drug screening and predictive toxicology. Stem Cell Res Ther 2018; 9:112. [PMID: 29678192 PMCID: PMC5910611 DOI: 10.1186/s13287-018-0847-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The production of veritable in-vitro models of bone tissue is essential to understand the biology of bone and its surrounding environment, to analyze the pathogenesis of bone diseases (e.g., osteoporosis, osteoarthritis, osteomyelitis, etc.), to develop effective therapeutic drug screening, and to test potential therapeutic strategies. Dysregulated interactions between vasculature and bone cells are often related to the aforementioned pathologies, underscoring the need for a bone model that contains engineered vasculature. Due to ethical restraints and limited prediction power of animal models, human stem cell-based tissue engineering has gained increasing relevance as a candidate approach to overcome the limitations of animals and to serve as preclinical models for drug testing. Since bone is a highly vascularized tissue, the concomitant development of vasculature and mineralized matrix requires a synergistic interaction between osteogenic and endothelial precursors. A number of experimental approaches have been used to achieve this goal, such as the combination of angiogenic factors and three-dimensional scaffolds, prevascularization strategies, and coculture systems. In this review, we present an overview of the current models and approaches to generate in-vitro stem cell-based vascularized bone, with emphasis on the main challenges of vasculature engineering. These challenges are related to the choice of biomaterials, scaffold fabrication techniques, and cells, as well as the type of culturing conditions required, and specifically the application of dynamic culture systems using bioreactors.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
- Ri.MED Foundation, Via Bandiera 11, Palermo, 90133 Italy
| | - Peter G. Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| |
Collapse
|
23
|
Goonoo N. Vascularization and angiogenesis in electrospun tissue engineered constructs: towards the creation of long-term functional networks. Biomed Phys Eng Express 2018. [DOI: 10.1088/2057-1976/aaab03] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Kawecki F, Clafshenkel WP, Fortin M, Auger FA, Fradette J. Biomimetic Tissue-Engineered Bone Substitutes for Maxillofacial and Craniofacial Repair: The Potential of Cell Sheet Technologies. Adv Healthc Mater 2018; 7:e1700919. [PMID: 29280323 DOI: 10.1002/adhm.201700919] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/02/2017] [Indexed: 12/21/2022]
Abstract
Maxillofacial defects are complex lesions stemming from various etiologies: accidental, congenital, pathological, or surgical. A bone graft may be required when the normal regenerative capacity of the bone is exceeded or insufficient. Surgeons have many options available for bone grafting including the "gold standard" autologous bone graft. However, this approach is not without drawbacks such as the morbidity associated with harvesting bone from a donor site, pain, infection, or a poor quantity and quality of bone in some patient populations. This review discusses the various bone graft substitutes used for maxillofacial and craniofacial repair: allografts, xenografts, synthetic biomaterials, and tissue-engineered substitutes. A brief overview of bone tissue engineering evolution including the use of mesenchymal stem cells is exposed, highlighting the first clinical applications of adipose-derived stem/stromal cells in craniofacial reconstruction. The importance of prevascularization strategies for bone tissue engineering is also discussed, with an emphasis on recent work describing substitutes produced using cell sheet-based technologies, including the use of thermo-responsive plates and the self-assembly approach of tissue engineering. Indeed, considering their entirely cell-based design, these natural bone-like substitutes have the potential to closely mimic the osteogenicity, osteoconductivity, osteoinduction, and osseointegration properties of autogenous bone for maxillofacial and craniofacial reconstruction.
Collapse
Affiliation(s)
- Fabien Kawecki
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Division of Regenerative Medicine CHU de Québec Research Center‐Université Laval Québec QC G1J 1Z4 Canada
- Department of Surgery Faculty of Medicine Université Laval Québec QC G1V 0A6 Canada
| | - William P. Clafshenkel
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Division of Regenerative Medicine CHU de Québec Research Center‐Université Laval Québec QC G1J 1Z4 Canada
- Department of Surgery Faculty of Medicine Université Laval Québec QC G1V 0A6 Canada
| | - Michel Fortin
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Division of Regenerative Medicine CHU de Québec Research Center‐Université Laval Québec QC G1J 1Z4 Canada
- Department of Oral and Maxillofacial Surgery Faculty of Dentistry Université Laval Québec QC G1V 0A6 Canada
| | - François A. Auger
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Division of Regenerative Medicine CHU de Québec Research Center‐Université Laval Québec QC G1J 1Z4 Canada
- Department of Surgery Faculty of Medicine Université Laval Québec QC G1V 0A6 Canada
| | - Julie Fradette
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Division of Regenerative Medicine CHU de Québec Research Center‐Université Laval Québec QC G1J 1Z4 Canada
- Department of Surgery Faculty of Medicine Université Laval Québec QC G1V 0A6 Canada
| |
Collapse
|
25
|
Bauman E, Feijão T, Carvalho DTO, Granja PL, Barrias CC. Xeno-free pre-vascularized spheroids for therapeutic applications. Sci Rep 2018; 8:230. [PMID: 29321569 PMCID: PMC5762877 DOI: 10.1038/s41598-017-18431-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/12/2017] [Indexed: 12/16/2022] Open
Abstract
Spheroid culture has gained increasing popularity, arising as a promising tool for regenerative medicine applications. Importantly, spheroids may present advantages over single-cell suspensions in cell-based therapies (CT). Unfortunately, most growth media used for spheroid culture contain animal origin-components, such as fetal bovine serum (FBS). The presence of FBS compromises the safety of CT and presents economic and ethical constraints. SCC (supplement for cell culture) is a novel xeno-free (XF) industrial cell culture supplement, derived from well-controlled pooled human plasma and processed under good manufacturing practice rules. Here, we developed a XF SCC-based formulation for 2D-culture of outgrowth endothelial cells (OEC), and then used it for generating co-culture spheroids of OEC and mesenchymal stem cells (MSC). XF MSC-OEC spheroids were characterized in detail and compared to spheroids cultured in FBS-supplemented medium. XF spheroids presented comparable integrity, size and morphology as the reference culture. The use of both media resulted in spheroids with similar structure, abundant extracellular matrix deposition and specific patterns of OEC distribution and organization. Notably, XF spheroids presented significantly enhanced angiogenic potential, both in vitro (fibrin sprouting assay) and in vivo (CAM assay). These findings are particularly promising in the context of potential therapeutic applications.
Collapse
Affiliation(s)
- E Bauman
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal
| | - T Feijão
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - D T O Carvalho
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - P L Granja
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - C C Barrias
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal. .,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal. .,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.
| |
Collapse
|
26
|
Lam J, Lee EJ, Clark EC, Mikos AG. Honing Cell and Tissue Culture Conditions for Bone and Cartilage Tissue Engineering. Cold Spring Harb Perspect Med 2017; 7:a025734. [PMID: 28348176 PMCID: PMC5710100 DOI: 10.1101/cshperspect.a025734] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
An avenue of tremendous interest and need in health care encompasses the regeneration of bone and cartilage. Over the years, numerous tissue engineering strategies have contributed substantial progress toward the realization of clinically relevant therapies. Cell and tissue culture protocols, however, show many variations that make experimental results among different publications challenging to compare. This collection surveys prevalent cell sources, soluble factors, culture medium formulations, environmental factors, and genetic modification approaches in the literature. The intent of consolidating this information is to provide a starting resource for scientists considering how to optimize the parameters for cell differentiation and tissue culture procedures within the context of bone and cartilage tissue engineering.
Collapse
Affiliation(s)
- Johnny Lam
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Esther J Lee
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Elisa C Clark
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Antonios G Mikos
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77251
| |
Collapse
|
27
|
αvβ3 and α5β1 integrin-specific ligands: From tumor angiogenesis inhibitors to vascularization promoters in regenerative medicine? Biotechnol Adv 2017; 36:208-227. [PMID: 29155160 DOI: 10.1016/j.biotechadv.2017.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 12/30/2022]
Abstract
Integrins are cell adhesion receptors predominantly important during normal and tumor angiogenesis. A sequence present on several extracellular matrix proteins composed of Arg-Gly-Asp (RGD) has attracted attention due to its role in cell adhesion mediated by integrins. The development of ligands that can bind to integrins involved in tumor angiogenesis and brake disease progression has resulted in new investigational drug entities reaching the clinical trial phase in humans. The use of integrin-specific ligands can be useful for the vascularization of regenerative medicine constructs, which remains a major limitation for translation into clinical practice. In order to enhance vascularization, immobilization of integrin-specific RGD peptidomimetics within constructs is a recommended approach, due to their high specificity and selectivity towards certain desired integrins. This review endeavours to address the potential of peptidomimetic-coated biomaterials as vascular network promoters for regenerative medicine purposes. Clinical studies involving molecules tracking active integrins in cancer angiogenesis and reasons for their failure are also addressed.
Collapse
|
28
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
29
|
The Transcription Factor Nrf2 Protects Angiogenic Capacity of Endothelial Colony-Forming Cells in High-Oxygen Radical Stress Conditions. Stem Cells Int 2017; 2017:4680612. [PMID: 28607561 PMCID: PMC5451769 DOI: 10.1155/2017/4680612] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 03/02/2017] [Accepted: 04/16/2017] [Indexed: 02/07/2023] Open
Abstract
Background Endothelial colony forming cells (ECFCs) have shown a promise in tissue engineering of vascular constructs, where they act as endothelial progenitor cells. After implantation, ECFCs are likely to be subjected to elevated reactive oxygen species (ROS). The transcription factor Nrf2 regulates the expression of antioxidant enzymes in response to ROS. Methods Stable knockdown of Nrf2 and Keap1 was achieved by transduction with lentiviral shRNAs; activation of Nrf2 was induced by incubation with sulforaphane (SFN). Expression of Nrf2 target genes was assessed by qPCR, oxidative stress was assessed using CM-DCFDA, and angiogenesis was quantified by scratch-wound and tubule-formation assays Results. Nrf2 knockdown led to a reduction of antioxidant gene expression and increased ROS. Angiogenesis was disturbed after Nrf2 knockdown even in the absence of ROS. Conversely, angiogenesis was preserved in high ROS conditions after knockdown of Keap1. Preincubation of ECFCs with SFN reduced intracellular ROS in the presence of H2O2 and preserved scratch-wound closure and tubule-formation. Results Nrf2 knockdown led to a reduction of antioxidant gene expression and increased ROS. Angiogenesis was disturbed after Nrf2 knockdown even in the absence of ROS. Conversely, angiogenesis was preserved in high ROS conditions after knockdown of Keap1. Preincubation of ECFCs with SFN reduced intracellular ROS in the presence of H2O2 and preserved scratch-wound closure and tubule-formation. Conclusion The results of this study indicate that Nrf2 plays an important role in the angiogenic capacity of ECFCs, particularly under conditions of increased oxidative stress. Pretreatment of ECFCs with SFN prior to implantation may be a protective strategy for tissue-engineered constructs or cell therapies.
Collapse
|
30
|
Ng J, Spiller K, Bernhard J, Vunjak-Novakovic G. Biomimetic Approaches for Bone Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:480-493. [PMID: 27912680 DOI: 10.1089/ten.teb.2016.0289] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Although autologous bone grafts are considered a gold standard for the treatment of bone defects, they are limited by donor site morbidities and geometric requirements. We propose that tissue engineering technology can overcome such limitations by recreating fully viable and biological bone grafts. Specifically, we will discuss the use of bone scaffolds and autologous cells with bioreactor culture systems as a tissue engineering paradigm to grow bone in vitro. We will also discuss emergent vascularization strategies to promote graft survival in vivo, as well as the role of inflammation during bone repair. Finally, we will highlight some recent advances and discuss new solutions to bone repair inspired by endochondral ossification.
Collapse
Affiliation(s)
- Johnathan Ng
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Kara Spiller
- 2 School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania
| | - Jonathan Bernhard
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Gordana Vunjak-Novakovic
- 1 Department of Biomedical Engineering, Columbia University , New York, New York.,3 Department of Medicine, Columbia University , New York, New York
| |
Collapse
|
31
|
Shanbhag S, Pandis N, Mustafa K, Nyengaard JR, Stavropoulos A. Cell Cotransplantation Strategies for Vascularized Craniofacial Bone Tissue Engineering: A Systematic Review and Meta-Analysis of Preclinical In Vivo Studies. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:101-117. [PMID: 27733094 DOI: 10.1089/ten.teb.2016.0283] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The regenerative potential of tissue-engineered bone constructs may be enhanced by in vitro coculture and in vivo cotransplantation of vasculogenic and osteogenic (progenitor) cells. The objective of this study was to systematically review the literature to answer the focused question: In animal models, does cotransplantation of osteogenic and vasculogenic cells enhance bone regeneration in craniofacial defects, compared with solely osteogenic cell-seeded constructs? Following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, electronic databases were searched for controlled animal studies reporting cotransplantation of endothelial cells (ECs) with mesenchymal stem cells (MSCs) or osteoblasts in craniofacial critical size defect (CSD) models. Twenty-two studies were included comparing outcomes of MSC/scaffold versus MSC+EC/scaffold (co)transplantation in calvarial (n = 15) or alveolar (n = 7) CSDs of small (rodents, rabbits) and large animal (minipigs, dogs) models. On average, studies presented with an unclear to high risk of bias. MSCs were derived from autologous, allogeneic, xenogeneic, or human (bone marrow, adipose tissue, periosteum) sources; in six studies, ECs were derived from MSCs by endothelial differentiation. In most studies, MSCs and ECs were cocultured in vitro (2-17 days) before implantation. Coculture enhanced MSC osteogenic differentiation and an optimal MSC:EC seeding ratio of 1:1 was identified. Alloplastic copolymer or composite scaffolds were most often used for in vivo implantation. Random effects meta-analyses were performed for histomorphometric and radiographic new bone formation (%NBF) and vessel formation in rodents' calvarial CSDs. A statistically significant benefit in favor of cotransplantation versus MSC-only transplantation for radiographic %NBF was observed in rat calvarial CSDs (weighted mean difference 7.80% [95% confidence interval: 1.39-14.21]); results for histomorphometric %NBF and vessel formation were inconclusive. Overall, heterogeneity in the meta-analyses was high (I2 > 80%). In summary, craniofacial bone regeneration is enhanced by cotransplantation of vasculogenic and osteogenic cells. Although the direction of treatment outcome is in favor of cotransplantation strategies, the magnitude of treatment effect does not seem to be of relevance, unless proven otherwise in clinical studies.
Collapse
Affiliation(s)
- Siddharth Shanbhag
- 1 Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen , Bergen, Norway .,2 Department of Periodontology, Faculty of Odontology, Malmö University , Malmö, Sweden
| | - Nikolaos Pandis
- 3 Department of Orthodontics and Dentofacial Orthopedics, School of Dental Medicine, University of Bern , Bern, Switzerland
| | - Kamal Mustafa
- 1 Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen , Bergen, Norway
| | - Jens R Nyengaard
- 4 Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, Aarhus University , Aarhus, Denmark
| | - Andreas Stavropoulos
- 2 Department of Periodontology, Faculty of Odontology, Malmö University , Malmö, Sweden
| |
Collapse
|
32
|
Patterning of Endothelial Cells and Mesenchymal Stem Cells by Laser-Assisted Bioprinting to Study Cell Migration. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3569843. [PMID: 27833916 PMCID: PMC5090075 DOI: 10.1155/2016/3569843] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/14/2016] [Indexed: 12/23/2022]
Abstract
Tissue engineering of large organs is currently limited by the lack of potent vascularization in vitro. Tissue-engineered bone grafts can be prevascularized in vitro using endothelial cells (ECs). The microvascular network architecture could be controlled by printing ECs following a specific pattern. Using laser-assisted bioprinting, we investigated the effect of distance between printed cell islets and the influence of coprinted mesenchymal cells on migration. When printed alone, ECs spread out evenly on the collagen hydrogel, regardless of the distance between cell islets. However, when printed in coculture with mesenchymal cells by laser-assisted bioprinting, they remained in the printed area. Therefore, the presence of mesenchymal cell is mandatory in order to create a pattern that will be conserved over time. This work describes an interesting approach to study cell migration that could be reproduced to study the effect of trophic factors.
Collapse
|
33
|
Jiang Y, Wang Y, Tang G. Cyclic tensile strain promotes the osteogenic differentiation of a bone marrow stromal cell and vascular endothelial cell co-culture system. Arch Biochem Biophys 2016; 607:37-43. [DOI: 10.1016/j.abb.2016.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/11/2016] [Accepted: 08/19/2016] [Indexed: 01/09/2023]
|
34
|
Rouwkema J, Khademhosseini A. Vascularization and Angiogenesis in Tissue Engineering: Beyond Creating Static Networks. Trends Biotechnol 2016; 34:733-745. [DOI: 10.1016/j.tibtech.2016.03.002] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/10/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
|
35
|
Amini AR, Xu TO, Chidambaram RM, Nukavarapu SP. Oxygen Tension-Controlled Matrices with Osteogenic and Vasculogenic Cells for Vascularized Bone Regeneration In Vivo. Tissue Eng Part A 2016; 22:610-20. [PMID: 26914219 PMCID: PMC4841084 DOI: 10.1089/ten.tea.2015.0310] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 02/23/2016] [Indexed: 12/23/2022] Open
Abstract
Despite recent progress, segmental bone defect repair is still a significant challenge in orthopedic surgery. While bone tissue engineering approaches using biodegradable matrices along with bone/blood vessel forming cells offered improved possibilities, current regenerative strategies lack the ability to achieve vascularized bone regeneration in critical-sized/segmental bone defects. In this study, we introduced and evaluated a two-pronged approach for vascularized bone regeneration in vivo. The goal was to demonstrate vascularized bone formation using oxygen tension-controlled (OTC) matrices seeded with bone and blood vessel forming cells. OTC matrices were coimplanted with rabbit mesenchymal stem cells (MSCs) and peripheral blood-derived endothelial progenitor cells (PB-EPCs) to demonstrate the osteogenic and vasculogenic differentiation of these cells, postseeding on a matrix, especially deep inside the matrix pore structure. Matrices coimplanted with varied rabbit MSC and PB-EPC ratios (1:4, 1:1, and 4:1) were assessed in a nude mouse subcutaneous implantation model to determine a coimplantation ratio with superior osteogenic as well as vasculogenic properties. The implants were analyzed, at week 8, for endothelial (CD31 and Von Willebrand factor [vWF]) and osteogenic marker (RunX2 and Col I) staining qualitatively and collagen deposition and number of vessel formation quantitatively. Results from these experiments established MSC-to-PB-EPC ratio 1:1 as the best coimplantation ratio. OTC matrix with 1:1 coimplantation ratio was assessed for segmental bone defect repair in a rabbit critical-sized bone defect model. The group under investigation was OTC matrix, and the matrix was seeded with MSCs, EPCs, or MSCs:EPCs in a 1:1 ratio. Explants at week 12 were evaluated for bone defect repair via micro-CT and histology. Results from rabbit in vivo experiments show enhanced mineralization and vascularization for the 1:1 coimplantation group. Overall, the study establishes a two-pronged approach involving OTC matrix and effective progenitors for large-area and vascularized bone regeneration.
Collapse
Affiliation(s)
- Ami R. Amini
- Oral and Maxillofacial Surgery, Massachusetts General Hospital, Boston, Massachusetts
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut
| | - Thomas O. Xu
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Ramaswamy M. Chidambaram
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Syam P. Nukavarapu
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut
- Department of Materials Science & Engineering, University of Connecticut, Storrs, Connecticut
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
36
|
Kumar VV, Heller M, Götz H, Schiegnitz E, Al-Nawas B, Kämmerer PW. Comparison of growth & function of endothelial progenitor cells cultured on deproteinized bovine bone modified with covalently bound fibronectin and bound vascular endothelial growth factor. Clin Oral Implants Res 2016; 28:543-550. [PMID: 26992449 DOI: 10.1111/clr.12832] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVES The objective of this study was to assess and compare the growth and function of Endothelial Progenitor Cells (EPCs) cultured on covalently bonded Vascular Endothelial Growth Factor (VEGF) and covalently bonded Fibronectin (FN) coating on deproteinized bovine bone (DBB) (test samples), compared to non-modified DBB blocks (control sample). MATERIALS AND METHODS The test samples were prepared by plasma polymerization of allylamine onto DBB blocks. Group1 of test samples were prepared with VEGF coating (VEGF-DBB) where as the Group2 test samples were coated with FN (FN-DBB). Non-modified DBB blocks served as a Control. EPCs were isolated and cultivated from buffy coats of peripheral blood of healthy volunteers and cultivated in the different samples and examined at time intervals of 24 h, 3 days, and 7 days. Evaluation of growth by cell count and cell morphology was done using Confocal Laser Scanning Electron Microscopy; vitality and function of cells was assessed using MTT assay and RT-PCR and ELISA for eNOS and iNOS respectively. RESULTS The results of the study show that both VEGF and FN could be successfully immobilized by plasma polymerization onto a complex, porous, three-dimensional structure of DBB. When comparing vital cell coverage, proliferation and function of EPCs, FN-DBB provided more positive values followed by VEGF-DBB as compared to DBB samples. eNOS level were significant higher in VEGF-DBB and FN-DBB when compared to DBB (P = 0.019 and P = 0.002). The difference between VEGF-DBB and FN-DBB was not significant. CONCLUSIONS Biomimetic coatings of Fibronectin may clinically relate to faster angiogenesis and earlier healing potential.
Collapse
Affiliation(s)
- Vinay V Kumar
- Department of Oral and Maxillofacial Surgery, University Medical Center of Johannes Gutenberg University, Mainz, Germany.,Department of Oral and Maxillofacial Surgery, University Medical Center of Rostock University, Rostock, Germany.,Head and Neck Institute, Mazumdar Shaw Medical Center and Center for Translational Research, Narayana Health, Bangalore, India
| | - Martin Heller
- Max Plank Institute for Polymer Research, Mainz, Germany
| | - Hermann Götz
- Department of Applied Structure and Microanalysis, University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Eik Schiegnitz
- Department of Oral and Maxillofacial Surgery, University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery, University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Peer W Kämmerer
- Department of Oral and Maxillofacial Surgery, University Medical Center of Rostock University, Rostock, Germany
| |
Collapse
|
37
|
Cherubino M, Valdatta L, Balzaretti R, Pellegatta I, Rossi F, Protasoni M, Tedeschi A, Accolla RS, Bernardini G, Gornati R. Human adipose-derived stem cells promote vascularization of collagen-based scaffolds transplanted into nude mice. Regen Med 2016; 11:261-71. [PMID: 26965659 PMCID: PMC4976995 DOI: 10.2217/rme-2015-0010] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM After in vivo implantation of cell-loaded devices, only the cells close to the capillaries can obtain nutrients to maintain their functions. It is known that factors secreted by stem cells, rather than stem cells themselves, are fundamental to guarantee new vascularization in the area of implant. MATERIALS & METHODS To investigate this possibility, we have grafted mice with Bilayer and Flowable Integra(®) scaffolds, loaded or not with human adipose-derived stem cells. RESULTS Our results support the therapeutic potential of human adipose-derived stem cells to induce new vascular networks of engineered organs and tissues. CONCLUSION This finding suggests that our approach can help to form new vascular networks that allow sufficient vascularization of engineered organs and tissues in cases of difficult wound healing due to ischemic conditions.
Collapse
Affiliation(s)
- Mario Cherubino
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via Dunant 3, Varese, Italy
| | - Luigi Valdatta
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via Dunant 3, Varese, Italy
| | - Riccardo Balzaretti
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via Dunant 3, Varese, Italy
| | - Igor Pellegatta
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via Dunant 3, Varese, Italy
| | - Federica Rossi
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via Dunant 3, Varese, Italy
| | - Marina Protasoni
- Dipartimento di Scienze Chirurgiche e Morfologiche, Università degli Studi dell'Insubria, Via Guicciardini 9, 21100 Varese, Italy
| | - Alessandra Tedeschi
- Dipartimento di Scienze Chirurgiche e Morfologiche, Università degli Studi dell'Insubria, Via Guicciardini 9, 21100 Varese, Italy
| | - Roberto S Accolla
- Dipartimento di Scienze Chirurgiche e Morfologiche, Università degli Studi dell'Insubria, Via Guicciardini 9, 21100 Varese, Italy
| | - Giovanni Bernardini
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via Dunant 3, Varese, Italy.,"The Protein Factory" Research Center, Politecnico di Milano, ICRM-CNR Milano & Università dell'Insubria, Via Mancinelli 7, Milano, Italy
| | - Rosalba Gornati
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via Dunant 3, Varese, Italy.,"The Protein Factory" Research Center, Politecnico di Milano, ICRM-CNR Milano & Università dell'Insubria, Via Mancinelli 7, Milano, Italy
| |
Collapse
|
38
|
Eman RM, Meijer HA, Öner FC, Dhert WJ, Alblas J. Establishment of an Early Vascular Network Promotes the Formation of Ectopic Bone. Tissue Eng Part A 2016; 22:253-62. [DOI: 10.1089/ten.tea.2015.0227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Rhandy M. Eman
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Henriette A.W. Meijer
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F. Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wouter J.A. Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
39
|
Wei H, Zhao X, Yuan R, Dai X, Li Y, Liu L. Effects of PB-EPCs on Homing Ability of Rabbit BMSCs via Endogenous SDF-1 and MCP-1. PLoS One 2015; 10:e0145044. [PMID: 26660527 PMCID: PMC4682485 DOI: 10.1371/journal.pone.0145044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 11/29/2015] [Indexed: 01/07/2023] Open
Abstract
Traumas, infections, tumors, and some congenital malformations can lead to bone defects or even bone loss. The goal of the present study was to investigate whether inclusion of endothelial progenitor cells derived from peripheral blood (PB–EPCs) in cell-seeded partially deproteinized bone (PDPB) implants would stimulate recruitment of systemically injected bone marrow stromal cells (BMSCs) to the implant. Methods: BMSCs were injected intravenously with lentiviral expression vector expressing enhanced green fluorescent protein (eGFP) for tracing. Recruitment of eGFP-positive BMSCs was tested for the following implant configurations: 1) seeded with both BMSC and PB-EPC, 2) BMSC alone, 3) PB-EPC alone, and 4) unseeded PDPB. Protein and mRNA levels of endogenous stromal-derived factor-1 (SDF-1) and its receptor CXCR4, as well as monocyte chemotactic protein-1 (MCP-1) and its receptor CCR2, were evaluated on the 8th week. Immunohistochemical staining was performed to determine eGFP-positive areas at the defective sites. Masson’s trichrome staining was conducted to observe the distribution of collagen deposition and evaluate the extent of osteogenesis. Results: The mRNA and protein levels of SDF-1 and CXCR4 in the co-culture group were higher than those in other groups (p < 0.05) 8 weeks after the surgery. MCP-1 mRNA level in the co-culture group was also higher than that in the other groups (p < 0.05). Immunohistochemical assays revealed that the area covered by eGFP-positive cells was larger in the co-culture group than in the other groups (p < 0.05) after 4 weeks. Masson’s trichrome staining revealed better osteogenic potential of the co-culture group compared to the other groups (p < 0.05). Conclusion: These experiments demonstrate an association between PB-EPC and BMSC recruitment mediated by the SDF-1/CXCR4 axis that can enhance repair of bone defects.
Collapse
Affiliation(s)
- Hanxiao Wei
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Xian Zhao
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Ruihong Yuan
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Xiaoming Dai
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Yisong Li
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
| | - Liu Liu
- Department of Plastic Surgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR of China
- * E-mail:
| |
Collapse
|
40
|
Mihaila SM, Resende MF, Reis RL, Gomes ME, Marques AP. Interactive endothelial phenotype maintenance and osteogenic differentiation of adipose tissue stromal vascular fraction SSEA-4+-derived cells. J Tissue Eng Regen Med 2015; 11:1998-2013. [DOI: 10.1002/term.2096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 07/29/2015] [Accepted: 09/15/2015] [Indexed: 01/07/2023]
Affiliation(s)
- Silvia M. Mihaila
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics; University of Minho; Barco GMR Portugal
- ICVS/3Bs; PT Government Associated Laboratory; Braga Guimarães Portugal
| | - Margarida F. Resende
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics; University of Minho; Barco GMR Portugal
- ICVS/3Bs; PT Government Associated Laboratory; Braga Guimarães Portugal
| | - Rui L. Reis
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics; University of Minho; Barco GMR Portugal
- ICVS/3Bs; PT Government Associated Laboratory; Braga Guimarães Portugal
| | - Manuela E. Gomes
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics; University of Minho; Barco GMR Portugal
- ICVS/3Bs; PT Government Associated Laboratory; Braga Guimarães Portugal
| | - Alexandra P. Marques
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics; University of Minho; Barco GMR Portugal
- ICVS/3Bs; PT Government Associated Laboratory; Braga Guimarães Portugal
| |
Collapse
|
41
|
Labeling and qualification of endothelial progenitor cells for tracking in tissue engineering: An in vitro study. Int J Artif Organs 2015; 38:224-32. [PMID: 25952997 DOI: 10.5301/ijao.5000405] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2015] [Indexed: 01/24/2023]
Abstract
PURPOSE In order to track location and distribution of endothelial cells (ECs) within scaffolds in vitro, we chose lentiPGK-TdTomato transduction of human endothelial progenitor cells (EPCs) isolated and differentiated from cord blood. Because transduction could have a functional impact on cell behavior, we checked different parameters for qualification of labeled- EPCs as well as their use for potential applications in the context of vascular and bone tissue engineering. METHODS After isolation and expansion, EPCs were classically characterized then transduced with the lentiviral vector containing the TdTomato protein gene under the control of the phosphoglycerate kinase (PGK) promoter. Conventional karyotyping, differentiation capacity, viability, proliferation assays were performed with labeled and unlabeled EPCs. Scaffolds and co-cultures were explored with labeled EPCs, in static or shear stress conditions. RESULTS Our results show that cell labeling did not affect cell adhesion nor induce cell death. Cell labeling did not induce more chromosomal aberrations. Phenotypical characterization was not affected. In the context of tissue engineering applications, labeled EPCs maintained their ability to line 2D or 3D scaffolds, withstand physiological arterial shear stress, and form tubular networks in co-cultures with human osteoblast progenitor cells. CONCLUSIONS It is possible to label human EPCs with TdTomato without affecting their behavior by the transduction procedure. This creates an important tool for numerous applications. Our results provide a qualification of labeled EPCs in comparison with unlabeled ones for vascular and bone tissue engineering.
Collapse
|
42
|
Samorezov JE, Alsberg E. Spatial regulation of controlled bioactive factor delivery for bone tissue engineering. Adv Drug Deliv Rev 2015; 84:45-67. [PMID: 25445719 PMCID: PMC4428953 DOI: 10.1016/j.addr.2014.11.018] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 12/29/2022]
Abstract
Limitations of current treatment options for critical size bone defects create a significant clinical need for tissue engineered bone strategies. This review describes how control over the spatiotemporal delivery of growth factors, nucleic acids, and drugs and small molecules may aid in recapitulating signals present in bone development and healing, regenerating interfaces of bone with other connective tissues, and enhancing vascularization of tissue engineered bone. State-of-the-art technologies used to create spatially controlled patterns of bioactive factors on the surfaces of materials, to build up 3D materials with patterns of signal presentation within their bulk, and to pattern bioactive factor delivery after scaffold fabrication are presented, highlighting their applications in bone tissue engineering. As these techniques improve in areas such as spatial resolution and speed of patterning, they will continue to grow in value as model systems for understanding cell responses to spatially regulated bioactive factor signal presentation in vitro, and as strategies to investigate the capacity of the defined spatial arrangement of these signals to drive bone regeneration in vivo.
Collapse
Affiliation(s)
- Julia E Samorezov
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH, USA; National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
43
|
Lloyd-Griffith C, Duffy GP, O'Brien FJ. Investigating the effect of hypoxic culture on the endothelial differentiation of human amniotic fluid-derived stem cells. J Anat 2015; 227:767-80. [PMID: 25833670 DOI: 10.1111/joa.12283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2015] [Indexed: 12/14/2022] Open
Abstract
Amniotic fluid-derived stem cells (AFSCs) are a unique stem cell source that may have great potential for use in tissue engineering (TE) due to their pluripotentiality. AFSCs have previously shown angiogenic potential and may present an alternative cell source for endothelial-like cells that could be used in range of applications, including the pre-vascularisation of TE constructs and the treatment of ischaemic diseases. This study investigated the ability of these cells to differentiate down an endothelial lineage with the aim of producing an endothelial-like cell suitable for use in pre-vascularisation. As hypoxia and the associated HIF-1 pathway have been implicated in the induction of angiogenesis in a number of biological processes, it was hypothesised that culture in hypoxic conditions could enhance the endothelial differentiation of AFSCs. The cells were cultured in endothelial cell media supplemented with 50 ng mL(-1) of VEGF, maintained in normoxia, intermittent hypoxia or continuous hypoxia and assessed for markers of endothelial differentiation at day 7 and 14. The results demonstrated that AFSCs subjected to these culture conditions display an endothelial gene expression profile and adopted functional endothelial cell characteristics indicative of early endothelial differentiation. Culture in continuous hypoxia enhanced endothelial gene expression but did not enhance functional endothelial cell characteristics. Overall, AFSCs subjected to endothelial stimuli demonstrated a less mature endothelial gene expression profile and phenotype when compared with HUVECs, the endothelial cell control. However, this study is the first time that the positive effect of an extended period of continuous hypoxic culture on endothelial differentiation in AFSCs has been demonstrated.
Collapse
Affiliation(s)
- Cai Lloyd-Griffith
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Dublin 2, Ireland
| | - Garry P Duffy
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Dublin 2, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Dublin 2, Ireland
| |
Collapse
|
44
|
Mercado-Pagán ÁE, Stahl AM, Shanjani Y, Yang Y. Vascularization in bone tissue engineering constructs. Ann Biomed Eng 2015; 43:718-29. [PMID: 25616591 PMCID: PMC4979539 DOI: 10.1007/s10439-015-1253-3] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 01/13/2015] [Indexed: 01/04/2023]
Abstract
Vascularization of large bone grafts is one of the main challenges of bone tissue engineering (BTE), and has held back the clinical translation of engineered bone constructs for two decades so far. The ultimate goal of vascularized BTE constructs is to provide a bone environment rich in functional vascular networks to achieve efficient osseointegration and accelerate restoration of function after implantation. To attain both structural and vascular integration of the grafts, a large number of biomaterials, cells, and biological cues have been evaluated. This review will present biological considerations for bone function restoration, contemporary approaches for clinical salvage of large bone defects and their limitations, state-of-the-art research on the development of vascularized bone constructs, and perspectives on evaluating and implementing novel BTE grafts in clinical practice. Success will depend on achieving full graft integration at multiple hierarchical levels, both between the individual graft components as well as between the implanted constructs and their surrounding host tissues. The paradigm of vascularized tissue constructs could not only revolutionize the progress of BTE, but could also be readily applied to other fields in regenerative medicine for the development of new innovative vascularized tissue designs.
Collapse
Affiliation(s)
| | - Alexander M. Stahl
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Yaser Shanjani
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
| | - Yunzhi Yang
- Department of Orthopedic Surgery, Stanford University, Stanford, CA, USA
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
45
|
In vitro co-culture strategies to prevascularization for bone regeneration: A brief update. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-014-0095-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
46
|
Guerrero J, Oliveira H, Catros S, Siadous R, Derkaoui SM, Bareille R, Letourneur D, Amédée J. The use of total human bone marrow fraction in a direct three-dimensional expansion approach for bone tissue engineering applications: focus on angiogenesis and osteogenesis. Tissue Eng Part A 2014; 21:861-74. [PMID: 25333855 DOI: 10.1089/ten.tea.2014.0367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current approaches in bone tissue engineering have shown limited success, mostly owing to insufficient vascularization of the construct. A common approach consists of co-culture of endothelial cells and osteoblastic cells. This strategy uses cells from different sources and differentiation states, thus increasing the complexity upstream of a clinical application. The source of reparative cells is paramount for the success of bone tissue engineering applications. In this context, stem cells obtained from human bone marrow hold much promise. Here, we analyzed the potential of human whole bone marrow cells directly expanded in a three-dimensional (3D) polymer matrix and focused on the further characterization of this heterogeneous population and on their ability to promote angiogenesis and osteogenesis, both in vitro and in vivo, in a subcutaneous model. Cellular aggregates were formed within 24 h and over the 12-day culture period expressed endothelial and bone-specific markers and a specific junctional protein. Ectopic implantation of the tissue-engineered constructs revealed osteoid tissue and vessel formation both at the periphery and within the implant. This work sheds light on the potential clinical use of human whole bone marrow for bone regeneration strategies, focusing on a simplified approach to develop a direct 3D culture without two-dimensional isolation or expansion.
Collapse
Affiliation(s)
- Julien Guerrero
- 1 Inserm, U1026, Tissue Bioengineering, University of Bordeaux , Bordeaux, France
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Dissanayaka WL, Hargreaves KM, Jin L, Samaranayake LP, Zhang C. The interplay of dental pulp stem cells and endothelial cells in an injectable peptide hydrogel on angiogenesis and pulp regeneration in vivo. Tissue Eng Part A 2014; 21:550-63. [PMID: 25203774 DOI: 10.1089/ten.tea.2014.0154] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Securing an adequate blood supply for the survival of cell transplants is critical for a successful outcome in tissue engineering. Interactions between endothelial and progenitor/stem cells are important for vascularization of regenerating tissue. Recently, self-assembling peptide nanofibers were described as a promising environment for pulp regeneration due to their synthetic nature and controlled physicochemical properties. In this study, the peptide hydrogel PuraMatrix™ was used as a scaffold system to investigate the role of dental pulp stem cells (DPSCs) in triggering angiogenesis and the potential for regenerating vascularized pulp in vivo. Human umbilical vein endothelial cells (HUVECs), DPSCs, or cocultures of both cell types were encapsulated in three-dimensional PuraMatrix. The peptide nanofiber microenvironment supported cell survival, cell migration, and capillary network formation in the absence of exogenous growth factors. DPSCs increased early vascular network formation by facilitating the migration of HUVECs and by increasing vascular endothelial growth factor (VEGF) expression. Both the DPSC-monoculture and coculture groups exhibited vascularized pulp-like tissue with patches of osteodentin after transplantation in mice. The cocultured groups exhibited more extracellular matrix, vascularization, and mineralization than the DPSC-monocultures in vivo. The DPSCs play a critical role in initial angiogenesis, whereas coordinated efforts by the HUVECs and DPSCs are required to achieve a balance between extracellular matrix deposition and mineralization. The findings of this study also highlighted the importance of a microenvironment that supports cell-cell interactions and cell migration, which contribute to successful dental pulp regeneration.
Collapse
Affiliation(s)
- Waruna Lakmal Dissanayaka
- 1 Department of Endodontics, Comprehensive Dental Care, Faculty of Dentistry, The University of Hong Kong , Hong Kong, China
| | | | | | | | | |
Collapse
|
48
|
King A, Balaji S, Keswani SG, Crombleholme TM. The Role of Stem Cells in Wound Angiogenesis. Adv Wound Care (New Rochelle) 2014; 3:614-625. [PMID: 25300298 DOI: 10.1089/wound.2013.0497] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 03/19/2014] [Indexed: 12/18/2022] Open
Abstract
Significance: Revascularization plays a critical role in wound healing and is regulated by a complex milieu of growth factors and cytokines. Deficiencies in revascularization contribute to the development of chronic nonhealing wounds. Recent Advances: Stem-cell-based therapy provides a novel strategy to enhance angiogenesis and improve wound healing. With bioethical concerns associated with embryonic stem cells, focus has shifted to different populations of vascular precursors, isolated from adult somatic tissue. Three main populations have been identified: endothelial progenitor cells, mesenchymal stem cells, and induced-pluripotent stem cells. These populations demonstrate great promise to positively influence neovascularization and wound repair. Critical Issues: Further studies to more definitively define each population are necessary to efficiently translate stem-cell-based therapeutic angiogenesis to the bedside. Better understanding of the physiologic pathways of how stem cells contribute to angiogenesis in normal tissue repair will help identify targets for successful therapeutic angiogenesis. Future Directions: Active studies in both animal models and clinical trials are being conducted to develop effective delivery routes, including dosing, route, and timing. Stem-cell-based therapy holds significant potential as a strategy for therapeutic angiogenesis in the care of patients with chronic nonhealing wounds.
Collapse
Affiliation(s)
- Alice King
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Swathi Balaji
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Sundeep G. Keswani
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Timothy M. Crombleholme
- Center for Children's Surgery, Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital Colorado, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
49
|
Dissanayaka WL, Zhu L, Hargreaves KM, Jin L, Zhang C. Scaffold-free Prevascularized Microtissue Spheroids for Pulp Regeneration. J Dent Res 2014; 93:1296-303. [PMID: 25201919 DOI: 10.1177/0022034514550040] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Creating an optimal microenvironment that mimics the extracellular matrix (ECM) of natural pulp and securing an adequate blood supply for the survival of cell transplants are major hurdles that need to be overcome in dental pulp regeneration. However, many currently available scaffolds fail to mimic essential functions of natural ECM. The present study investigated a novel approach involving the use of scaffold-free microtissue spheroids of dental pulp stem cells (DPSCs) prevascularized by human umbilical vein endothelial cells (HUVECs) in pulp regeneration. In vitro-fabricated microtissue spheroids were inserted into the canal space of tooth-root slices and were implanted subcutaneously into immunodeficient mice. Histological examination revealed that, after four-week implantation, tooth-root slices containing microtissue spheroids resulted in well-vascularized and cellular pulp-like tissues, compared with empty tooth-root slices, which were filled with only subcutaneous fat tissue. Immunohistochemical staining indicated that the tissue found in the tooth-root slices was of human origin, as characterized by the expression of human mitochondria, and contained odontoblast-like cells organized along the dentin, as assessed by immunostaining for nestin and dentin sialoprotein (DSP). Vascular structures formed by HUVECs in vitro were successfully anastomosed with the host vasculature upon transplantation in vivo, as shown by immunostaining for human CD31. Collectively, these findings demonstrate that prevascularized, scaffold-free, microtissue spheroids can successfully regenerate vascular dental pulp-like tissue and also highlight the significance of the microtissue microenvironment as an optimal environment for successful pulp-regeneration strategies.
Collapse
Affiliation(s)
- W L Dissanayaka
- Endodontics, Comprehensive Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - L Zhu
- Endodontics, Comprehensive Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - K M Hargreaves
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - L Jin
- Periodontology and Public Health, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - C Zhang
- Endodontics, Comprehensive Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
50
|
Bertanha M, Moroz A, Jaldin RG, Silva RA, Rinaldi JC, Golim MA, Felisbino SL, Domingues MA, Sobreira ML, Reis PP, Deffune E. Morphofunctional characterization of decellularized vena cava as tissue engineering scaffolds. Exp Cell Res 2014; 326:103-11. [DOI: 10.1016/j.yexcr.2014.05.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/24/2014] [Accepted: 05/26/2014] [Indexed: 11/28/2022]
|