1
|
Iqbal MZ, Riaz M, Biedermann T, Klar AS. Breathing new life into tissue engineering: exploring cutting-edge vascularization strategies for skin substitutes. Angiogenesis 2024; 27:587-621. [PMID: 38842751 PMCID: PMC11564345 DOI: 10.1007/s10456-024-09928-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering.
Collapse
Affiliation(s)
- M Zohaib Iqbal
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mahrukh Riaz
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Dousti M, Parsa S, Sani F, Bagherzadeh E, Zamanzadeh Z, Dara M, Sani M, Azarpira N. Enhancing bone regeneration: Unleashing the potential of magnetic nanoparticles in a microtissue model. J Cell Mol Med 2024; 28:e70040. [PMID: 39219020 PMCID: PMC11366680 DOI: 10.1111/jcmm.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Bone tissue engineering addresses the limitations of autologous resources and the risk of allograft disease transmission in bone diseases. In this regard, engineered three-dimensional (3D) models emerge as biomimetic alternatives to natural tissues, replicating intracellular communication. Moreover, the unique properties of super-paramagnetic iron oxide nanoparticles (SPIONs) were shown to promote bone regeneration via enhanced osteogenesis and angiogenesis in bone models. This study aimed to investigate the effects of SPION on both osteogenesis and angiogenesis and characterized a co-culture of Human umbilical vein endothelial cells (HUVEC) and MG-63 cells as a model of bone microtissue. HUVECs: MG-63s with a ratio of 4:1 demonstrated the best results among other cell ratios, and 50 μg/mL of SPION was the optimum concentration for maximum survival, cell migration and mineralization. In addition, the data from gene expression illustrated that the expression of osteogenesis-related genes, including osteopontin, osteocalcin, alkaline phosphatase, and collagen-I, as well as the expression of the angiogenesis-related marker, CD-31, and the tube formation, is significantly elevated when the 50 μg/mL concentration of SPION is applied to the microtissue samples. SPION application in a designed 3D bone microtissue model involving a co-culture of osteoblast and endothelial cells resulted in increased expression of specific markers related to angiogenesis and osteogenesis. This includes the design of a novel biomimetic model to boost blood compatibility and biocompatibility of primary materials while promoting osteogenic activity in microtissue bone models. Moreover, this can improve interaction with surrounding tissues and broaden the knowledge to promote superior-performance implants, preventing device failure.
Collapse
Affiliation(s)
- Maryam Dousti
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
- Department of Genetics, Faculty of Biological Sciences and TechnologyShahid Ashrafi Esfahani UniversityIsfahanIran
| | - Shima Parsa
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
| | - Farnaz Sani
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
| | | | - Zahra Zamanzadeh
- Department of Genetics, Faculty of Biological Sciences and TechnologyShahid Ashrafi Esfahani UniversityIsfahanIran
| | - Mahintaj Dara
- Stem Cells Technology Research CenterShiraz University of Medical SciencesShirazIran
| | - Mahsa Sani
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
- Tissue Engineering Department, School of Advanced Medical Science and TechnologyShiraz University of Medical ScienceShirazIran
| | - Negar Azarpira
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
- Transplant Research CenterShiraz University of Medical ScienceShirazIran
| |
Collapse
|
3
|
Pal D, Das P, Mukherjee P, Roy S, Chaudhuri S, Kesh SS, Ghosh D, Nandi SK. Biomaterials-Based Strategies to Enhance Angiogenesis in Diabetic Wound Healing. ACS Biomater Sci Eng 2024; 10:2725-2741. [PMID: 38630965 DOI: 10.1021/acsbiomaterials.4c00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Amidst the present healthcare issues, diabetes is unique as an emerging class of affliction with chronicity in a majority of the population. To check and control its effects, there have been huge turnover and constant development of management strategies, and though a bigger part of the health care area is involved in achieving its control and the related issues such as the effect of diabetes on wound healing and care and many of the works have reached certain successful outcomes, still there is a huge lack in managing it, with maximum effect yet to be attained. Studying pathophysiology and involvement of various treatment options, such as tissue engineering, application of hydrogels, drug delivery methods, and enhancing angiogenesis, are at constantly developing stages either direct or indirect. In this review, we have gathered a wide field of information and different new therapeutic methods and targets for the scientific community, paving the way toward more settled ideas and research advances to cure diabetic wounds and manage their outcomes.
Collapse
Affiliation(s)
- Debajyoti Pal
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Pratik Das
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Prasenjit Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Subhasis Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shubhamitra Chaudhuri
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shyam Sundar Kesh
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Debaki Ghosh
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| |
Collapse
|
4
|
Xiong J, Kaleja P, Ückert L, Nezaratizadeh N, Krantz S, Krause MF, Fitschen-Oestern S, Seekamp A, Cassidy L, Tholey A, Fuchs S. Alveolar-Capillary Barrier Protection In Vitro: Lung Cell Type-Specific Effects and Molecular Mechanisms Induced by 1α, 25-Dihydroxyvitamin D3. Int J Mol Sci 2023; 24:ijms24087298. [PMID: 37108455 PMCID: PMC10138495 DOI: 10.3390/ijms24087298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Low serum levels of 1α, 25-dihydroxyvitamin D3 (VD3) are associated with a higher mortality in trauma patients with sepsis or ARDS. However, the molecular mechanisms behind this observation are not yet understood. VD3 is known to stimulate lung maturity, alveolar type II cell differentiation, or pulmonary surfactant synthesis and guides epithelial defense during infection. In this study, we investigated the impact of VD3 on the alveolar-capillary barrier in a co-culture model of alveolar epithelial cells and microvascular endothelial cells respectively in the individual cell types. After stimulation with bacterial LPS (lipopolysaccharide), gene expression of inflammatory cytokines, surfactant proteins, transport proteins, antimicrobial peptide, and doublecortin-like kinase 1 (DCLK1) were analyzed by real-time PCR, while corresponding proteins were evaluated by ELISA, immune-fluorescence, or Western blot. The effect of VD3 on the intracellular protein composition in H441 cells was analyzed by quantitative liquid chromatography-mass spectrometry-based proteomics. VD3 effectively protected the alveolar-capillary barrier against LPS treatment, as indicated by TEER measurement and morphological assessment. VD3 did not inhibit the IL-6 secretion by H441 and OEC but restricted the diffusion of IL-6 to the epithelial compartment. Further, VD3 could significantly suppress the surfactant protein A expression induced in the co-culture system by LPS treatment. VD3 induced high levels of the antimicrobial peptide LL-37, which counteracted effects by LPS and strengthened the barrier. Quantitative proteomics identified VD3-dependent protein abundance changes ranging from constitutional extracellular matrix components and surfactant-associated proteins to immune-regulatory molecules. DCLK1, as a newly described target molecule for VD3, was prominently stimulated by VD3 (10 nM) and seems to influence the alveolar-epithelial cell barrier and regeneration.
Collapse
Affiliation(s)
- Junyu Xiong
- Experimental Trauma Surgery, Department of Trauma Surgery and Orthopedics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Patrick Kaleja
- Systematic Proteomics & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, 24015 Kiel, Germany
| | - Larissa Ückert
- Experimental Trauma Surgery, Department of Trauma Surgery and Orthopedics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Niloufar Nezaratizadeh
- Experimental Trauma Surgery, Department of Trauma Surgery and Orthopedics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Stefanie Krantz
- Experimental Trauma Surgery, Department of Trauma Surgery and Orthopedics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Martin Friedrich Krause
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Stefanie Fitschen-Oestern
- Experimental Trauma Surgery, Department of Trauma Surgery and Orthopedics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Andreas Seekamp
- Experimental Trauma Surgery, Department of Trauma Surgery and Orthopedics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Liam Cassidy
- Systematic Proteomics & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, 24015 Kiel, Germany
| | - Andreas Tholey
- Systematic Proteomics & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, 24015 Kiel, Germany
| | - Sabine Fuchs
- Experimental Trauma Surgery, Department of Trauma Surgery and Orthopedics, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| |
Collapse
|
5
|
Mierke CT. Physical and biological advances in endothelial cell-based engineered co-culture model systems. Semin Cell Dev Biol 2023; 147:58-69. [PMID: 36732105 DOI: 10.1016/j.semcdb.2023.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023]
Abstract
Scientific knowledge in the field of cell biology and mechanobiology heavily leans on cell-based in vitro experiments and models that favor the examination and comprehension of certain biological processes and occurrences across a variety of environments. Cell culture assays are an invaluable instrument for a vast spectrum of biomedical and biophysical investigations. The quality of experimental models in terms of simplicity, reproducibility, and combinability with other methods, and in particular the scale at which they depict cell fate in native tissues, is critical to advancing the knowledge of the comprehension of cell-cell and cell-matrix interactions in tissues and organs. Typically, in vitro models are centered on the experimental tinkering of mammalian cells, most often cultured as monolayers on planar, two-dimensional (2D) materials. Notwithstanding the significant advances and numerous findings that have been accomplished with flat biology models, their usefulness for generating further new biological understanding is constrained because the simple 2D setting does not reproduce the physiological response of cells in natural living tissues. In addition, the co-culture systems in a 2D stetting weakly mirror their natural environment of tissues and organs. Significant advances in 3D cell biology and matrix engineering have resulted in the creation and establishment of a new type of cell culture shapes that more accurately represents the in vivo microenvironment and allows cells and their interactions to be analyzed in a biomimetic approach. Contemporary biomedical and biophysical science has novel advances in technology that permit the design of more challenging and resilient in vitro models for tissue engineering, with a particular focus on scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips, which cover the purposes of co-cultures. Even these complex systems must be kept as simplified as possible in order to grasp a particular section of physiology too very precisely. In particular, it is highly appreciated that they bridge the space between conventional animal research and human (patho)physiology. In this review, the recent progress in 3D biomimetic culturation is presented with a special focus on co-cultures, with an emphasis on the technological building blocks and endothelium-based co-culture models in cancer research that are available for the development of more physiologically relevant in vitro models of human tissues under normal and diseased conditions. Through applications and samples of various physiological and disease models, it is possible to identify the frontiers and future engagement issues that will have to be tackled to integrate synthetic biomimetic culture systems far more successfully into biomedical and biophysical investigations.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, Leipzig, Germany.
| |
Collapse
|
6
|
Biomimetic nanofiber-enabled rapid creation of skin grafts. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00009-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
7
|
Mason J, Öhlund D. Key aspects for conception and construction of co-culture models of tumor-stroma interactions. Front Bioeng Biotechnol 2023; 11:1150764. [PMID: 37091337 PMCID: PMC10119418 DOI: 10.3389/fbioe.2023.1150764] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/31/2023] [Indexed: 04/25/2023] Open
Abstract
The tumor microenvironment is crucial in the initiation and progression of cancers. The interplay between cancer cells and the surrounding stroma shapes the tumor biology and dictates the response to cancer therapies. Consequently, a better understanding of the interactions between cancer cells and different components of the tumor microenvironment will drive progress in developing novel, effective, treatment strategies. Co-cultures can be used to study various aspects of these interactions in detail. This includes studies of paracrine relationships between cancer cells and stromal cells such as fibroblasts, endothelial cells, and immune cells, as well as the influence of physical and mechanical interactions with the extracellular matrix of the tumor microenvironment. The development of novel co-culture models to study the tumor microenvironment has progressed rapidly over recent years. Many of these models have already been shown to be powerful tools for further understanding of the pathophysiological role of the stroma and provide mechanistic insights into tumor-stromal interactions. Here we give a structured overview of different co-culture models that have been established to study tumor-stromal interactions and what we have learnt from these models. We also introduce a set of guidelines for generating and reporting co-culture experiments to facilitate experimental robustness and reproducibility.
Collapse
Affiliation(s)
- James Mason
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Daniel Öhlund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- *Correspondence: Daniel Öhlund,
| |
Collapse
|
8
|
Mahapatra C, Kumar P, Paul MK, Kumar A. Angiogenic stimulation strategies in bone tissue regeneration. Tissue Cell 2022; 79:101908. [DOI: 10.1016/j.tice.2022.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
|
9
|
Moss SM, Schilp J, Yaakov M, Cook M, Schuschke E, Hanke B, Strobel HA, Hoying JB. Point-of-use, automated fabrication of a 3D human liver model supplemented with human adipose microvessels. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:358-368. [PMID: 35772696 DOI: 10.1016/j.slasd.2022.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
Advanced in vitro tissue models better reflect healthy and disease tissue conditions in the body. However, complex tissue models are often manufactured using custom solutions and can be challenging to manufacture to scale. Here, we describe the automated fabrication of a cell-dense, thick (≤ 1 cm), human vascularized liver tissue model using a robotic biomanufacturing platform and off-the-shelf components to build, culture, and sample liver tissues hands-free without compromising tissue health or function. Fabrication of the tissue involved 3D bioprinting and incorporation of primary human hepatocytes, primary human non-parenchymal cells, and isolated fragments of intact human microvessels as vascular precursors. No differences were observed in select assessments of the liver tissues fabricated by hand or via automation. Furthermore, constant media exchange, via perfusion, improved urea output and elevated tissue metabolism. Interestingly, inclusion of adipose-derived human microvessels enhanced functional gene expression, including an enhanced response to a drug challenge. Our results describe the fabrication of a thick liver tissue environment useful for a variety of applications including liver disease modeling, infectious agent studies, and cancer investigations. We expect the automated fabrication of the vascularized liver tissue, at the point of use and using off-the-shelf platforms, eases fabrication of the complex model and increases its utility.
Collapse
Affiliation(s)
- Sarah M Moss
- Advanced Solutions Life Sciences, Manchester, NH 03101, United States
| | - Jillian Schilp
- Advanced Solutions Life Sciences, Manchester, NH 03101, United States
| | - Maya Yaakov
- Advanced Solutions Life Sciences, Manchester, NH 03101, United States
| | - Madison Cook
- Advanced Solutions Life Sciences, Manchester, NH 03101, United States
| | - Erik Schuschke
- Advanced Solutions Life Sciences, Louisville, KY 40223, United States
| | - Brandon Hanke
- Advanced Solutions Life Sciences, Louisville, KY 40223, United States
| | - Hannah A Strobel
- Advanced Solutions Life Sciences, Manchester, NH 03101, United States
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, NH 03101, United States.
| |
Collapse
|
10
|
Guerrero J, Dasen B, Frismantiene A, Pigeot S, Ismail T, Schaefer DJ, Philippova M, Resink TJ, Martin I, Scherberich A. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:213-229. [PMID: 35259280 PMCID: PMC8929526 DOI: 10.1093/stcltm/szab021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022] Open
Abstract
Cells of the stromal vascular fraction (SVF) of human adipose tissue have the capacity to generate osteogenic grafts with intrinsic vasculogenic properties. However, cultured adipose-derived stromal cells (ASCs), even after minimal monolayer expansion, lose osteogenic capacity in vivo. Communication between endothelial and stromal/mesenchymal cell lineages has been suggested to improve bone formation and vascularization by engineered tissues. Here, we investigated the specific role of a subpopulation of SVF cells positive for T-cadherin (T-cad), a putative endothelial marker. We found that maintenance during monolayer expansion of a T-cad-positive cell population, composed of endothelial lineage cells (ECs), is mandatory to preserve the osteogenic capacity of SVF cells in vivo and strongly supports their vasculogenic properties. Depletion of T-cad-positive cells from the SVF totally impaired bone formation in vivo and strongly reduced vascularization by SVF cells in association with decreased VEGF and Adiponectin expression. The osteogenic potential of T-cad-depleted SVF cells was fully rescued by co-culture with ECs from a human umbilical vein (HUVECs), constitutively expressing T-cad. Ectopic expression of T-cad in ASCs stimulated mineralization in vitro but failed to rescue osteogenic potential in vivo, indicating that the endothelial nature of the T-cad-positive cells is the key factor for induction of osteogenesis in engineered grafts based on SVF cells. This study demonstrates that crosstalk between stromal and T-cad expressing endothelial cells within adipose tissue critically regulates osteogenesis, with VEGF and adiponectin as associated molecular mediators.
Collapse
Affiliation(s)
- Julien Guerrero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Boris Dasen
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Agne Frismantiene
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastien Pigeot
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tarek Ismail
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Corresponding author: Arnaud Scherberich, Department of Biomedicine, Hebelstrasse 20, University Hospital Basel, 4031 Basel, Switzerland. Tel: +41 061 328 73 75;
| |
Collapse
|
11
|
Shafiee S, Shariatzadeh S, Zafari A, Majd A, Niknejad H. Recent Advances on Cell-Based Co-Culture Strategies for Prevascularization in Tissue Engineering. Front Bioeng Biotechnol 2021; 9:745314. [PMID: 34900955 PMCID: PMC8655789 DOI: 10.3389/fbioe.2021.745314] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, the fabrication of a functional vascular network to maintain the viability of engineered tissues is a major bottleneck in the way of developing a more advanced engineered construct. Inspired by vasculogenesis during the embryonic period, the in vitro prevascularization strategies have focused on optimizing communications and interactions of cells, biomaterial and culture conditions to develop a capillary-like network to tackle the aforementioned issue. Many of these studies employ a combination of endothelial lineage cells and supporting cells such as mesenchymal stem cells, fibroblasts, and perivascular cells to create a lumenized endothelial network. These supporting cells are necessary for the stabilization of the newly developed endothelial network. Moreover, to optimize endothelial network development without impairing biomechanical properties of scaffolds or differentiation of target tissue cells, several other factors, including target tissue, endothelial cell origins, the choice of supporting cell, culture condition, incorporated pro-angiogenic factors, and choice of biomaterial must be taken into account. The prevascularization method can also influence the endothelial lineage cell/supporting cell co-culture system to vascularize the bioengineered constructs. This review aims to investigate the recent advances on standard cells used in in vitro prevascularization methods, their co-culture systems, and conditions in which they form an organized and functional vascular network.
Collapse
Affiliation(s)
- Sepehr Shafiee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Zafari
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Majd
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
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
|
13
|
Ellero AA, van den Bout I, Vlok M, Cromarty AD, Hurrell T. Continual proteomic divergence of HepG2 cells as a consequence of long-term spheroid culture. Sci Rep 2021; 11:10917. [PMID: 34035320 PMCID: PMC8149451 DOI: 10.1038/s41598-021-89907-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/28/2021] [Indexed: 02/04/2023] Open
Abstract
Three-dimensional models are considered a powerful tool for improving the concordance between in vitro and in vivo phenotypes. However, the duration of spheroid culture may influence the degree of correlation between these counterparts. When using immortalised cell lines as model systems, the assumption for consistency and reproducibility is often made without adequate characterization or validation. It is therefore essential to define the biology of each spheroid model by investigating proteomic dynamics, which may be altered relative to culture duration. As an example, we assessed the influence of culture duration on the relative proteome abundance of HepG2 cells cultured as spheroids, which are routinely used to model aspects of the liver. Quantitative proteomic profiling of whole cell lysates labelled with tandem-mass tags was conducted using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In excess of 4800 proteins were confidently identified, which were shared across three consecutive time points over 28 days. The HepG2 spheroid proteome was divergent from the monolayer proteome after 14 days in culture and continued to change over the successive culture time points. Proteins representing the recognised core hepatic proteome, cell junction, extracellular matrix, and cell adhesion proteins were found to be continually modulated.
Collapse
Affiliation(s)
- Andrea Antonio Ellero
- Department of Pharmacology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
- Centre for Neuroendocrinology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Iman van den Bout
- Centre for Neuroendocrinology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Maré Vlok
- Proteomics Unit, Central Analytical Facility, Stellenbosch University, Stellenbosch, South Africa
| | - Allan Duncan Cromarty
- Department of Pharmacology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Tracey Hurrell
- Bioengineering and Integrated Genomics Group, Next Generation Health Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
| |
Collapse
|
14
|
Wang F, Xiao Y, Neupane S, Ptak SH, Römer R, Xiong J, Ohmes J, Seekamp A, Fretté X, Alban S, Fuchs S. Influence of Fucoidan Extracts from Different Fucus Species on Adult Stem Cells and Molecular Mediators in In Vitro Models for Bone Formation and Vascularization. Mar Drugs 2021; 19:194. [PMID: 33805470 PMCID: PMC8066524 DOI: 10.3390/md19040194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 01/03/2023] Open
Abstract
Fucoidans, sulfated polysaccharides extracted from brown algae, are marine products with the potential to modulate bone formation and vascularization processes. The bioactivity and safety of fucoidans are highly associated with their chemical structure, which may vary with algae species and extraction method. Thus, in depth evaluation of fucoidan extracts in terms of endotoxin content, cytotoxicity, and their detailed molecular biological impact on the individual cell types in bone is needed. In this study, we characterized fucoidan extracts from three different Fucus species including Fucus vesiculosus (Fv), Fucus serratus (Fs), and Fucus distichus subsp. evanescens (Fe) for their chemical features, endotoxin content, cytotoxicity, and bioactive effects on human outgrowth endothelial cells (OEC) and human mesenchymal stem cells (MSC) as in vitro models for bone function and vascularization. Extracts contained mainly high molecular weight (HMW) fucoidans and were free of endotoxins that may cause inflammation or influence vascularization. OEC tolerated fucoidan concentrations up to 200 µg/mL, and no indication of cytotoxicity was observed. The inflammatory response, however, investigated by real-time PCR (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) and endothelial barrier assessed by impedance measurement differed for the individual extracts. MSC in comparison with endothelial cells were more sensitive to fucoidans and showed partly reduced metabolic activity and proliferation at higher doses of fucoidans. Further results for MSC indicated impaired osteogenic functions in alkaline phosphatase and calcification assays. All tested extracts consistently lowered important molecular mediators involved in angiogenesis, such a VEGF (vascular endothelial growth factor), ANG-1 (angiopoietin 1), and ANG-2 (angiopoietin 2), as indicated by RT-PCR and ELISA. This was associated with antiangiogenic effects at the functional level using selected extracts in co-culture models to mimic bone vascularization processes during bone regeneration or osteosarcoma.
Collapse
Affiliation(s)
- Fanlu Wang
- Experimental Trauma Surgery, Department of Orthopedics and Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany; (F.W.); (Y.X.); (R.R.); (J.X.); (J.O.); (A.S.)
| | - Yuejun Xiao
- Experimental Trauma Surgery, Department of Orthopedics and Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany; (F.W.); (Y.X.); (R.R.); (J.X.); (J.O.); (A.S.)
| | - Sandesh Neupane
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Kiel University, 24148 Kiel, Germany; (S.N.); (S.A.)
| | - Signe Helle Ptak
- SDU Chemical Engineering, University of Southern Denmark, 5230 Odense, Denmark; (S.H.P.); (X.F.)
| | - Ramona Römer
- Experimental Trauma Surgery, Department of Orthopedics and Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany; (F.W.); (Y.X.); (R.R.); (J.X.); (J.O.); (A.S.)
| | - Junyu Xiong
- Experimental Trauma Surgery, Department of Orthopedics and Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany; (F.W.); (Y.X.); (R.R.); (J.X.); (J.O.); (A.S.)
| | - Julia Ohmes
- Experimental Trauma Surgery, Department of Orthopedics and Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany; (F.W.); (Y.X.); (R.R.); (J.X.); (J.O.); (A.S.)
| | - Andreas Seekamp
- Experimental Trauma Surgery, Department of Orthopedics and Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany; (F.W.); (Y.X.); (R.R.); (J.X.); (J.O.); (A.S.)
| | - Xavier Fretté
- SDU Chemical Engineering, University of Southern Denmark, 5230 Odense, Denmark; (S.H.P.); (X.F.)
| | - Susanne Alban
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Kiel University, 24148 Kiel, Germany; (S.N.); (S.A.)
| | - Sabine Fuchs
- Experimental Trauma Surgery, Department of Orthopedics and Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany; (F.W.); (Y.X.); (R.R.); (J.X.); (J.O.); (A.S.)
| |
Collapse
|
15
|
Human amniotic membrane as a delivery vehicle for stem cell-based therapies. Life Sci 2021; 272:119157. [PMID: 33524418 DOI: 10.1016/j.lfs.2021.119157] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
Stem cell-based therapy is known as a regenerative approach for a variety of diseases and tissue injuries. These cells exert their therapeutic effects through paracrine secretions namely extracellular vesicles. To achieve higher therapeutic potential, a variety of delivery routes have been tested in clinical and preclinical studies. Direct cell injection, intra-venous administration, and intra-arterial infusion are widely used methods of stem cells delivery but these methods are associated with several complications. As one of the most popular biological delivery systems, amniotic membrane has been widely utilized to support cell proliferation and differentiation therefore facilitating tissue regeneration without endangering the stem cells' viability. It is composed of several extracellular matrix components and growth factors. Due to these characteristics, amniotic membrane can mimic the stem cell's niche and can be an ideal carrier for stem cell transplantation. Here, we provide an overview of the recent progress, challenges, and future perspectives in the use of amniotic membrane as a delivery platform for stem cells.
Collapse
|
16
|
Bellani C, Yue K, Flaig F, Hébraud A, Ray P, Annabi N, Selistre de Araújo HS, Branciforti MC, Minarelli Gaspar AM, Shin SR, Khademhosseini A, Schlatter G. Suturable elastomeric tubular grafts with patterned porosity for rapid vascularization of 3D constructs. Biofabrication 2021; 13. [PMID: 33482658 DOI: 10.1088/1758-5090/abdf1d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022]
Abstract
Vascularization is considered to be one of the key challenges in engineering functional 3D tissues. Engineering suturable vascular grafts containing pores with diameter of several tens of microns in tissue engineered constructs may provide an instantaneous blood perfusion through the grafts improving cell infiltration and thus, allowing rapid vascularization and vascular branching. The aim of this work was to develop suturable tubular scaffolds to be integrated in biofabricated constructs, enabling the direct connection of the biofabricated construct with the host blood stream, providing an immediate blood flow inside the construct. Here, tubular grafts with customizable shapes (tubes, Y-shape capillaries) and controlled diameter ranging from several hundreds of microns to few mm are fabricated based on poly(glycerol sebacate) (PGS) / poly(vinyl alcohol) (PVA) electrospun scaffolds. Furthermore, a network of pore channels of diameter in the order of 100 µm was machined by laser femtosecond ablation in the tube wall. Both non-machined and laser machined tubular scaffolds elongated more than 100% of their original size have shown suture retention, being 5.85 and 3.96 N/mm2 respectively. To demonstrate the potential of application, the laser machined porous grafts were embedded in gelatin methacryloyl (GelMA) hydrogels, resulting in elastomeric porous tubular graft/GelMA 3D constructs. These constructs were then co-seeded with osteoblast-like cells (MG-63) at the external side of the graft and endothelial cells (HUVEC) inside, forming a bone osteon model. The laser machined pore network allowed an immediate endothelial cell flow towards the osteoblasts enabling the osteoblasts and endothelial cells to interact and form 3D structures. This rapid vascularization approach could be applied, not only for bone tissue regeneration, but also for a variety of tissues and organs.
Collapse
Affiliation(s)
- Caroline Bellani
- University of Sao Paulo, AVENIDA TRABALHADOR SÃO-CARLENSE, 400, Sao Carlos, São Paulo, 13566-590, BRAZIL
| | - Kan Yue
- South China Advanced Institute for Soft Matter Science and Technology, South China University of Technology, 381 Wushan Rd, Guangzhou, Guangdong, 510641, CHINA
| | - Florence Flaig
- ICPEES, University of Strasbourg, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| | - Anne Hébraud
- ICPEES, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| | - Pengfei Ray
- Division of Health Sciences and Technology, MIT, 45 Carleton Street, Cambridge, Massachusetts, 02142, UNITED STATES
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, UCLA, 5531 Boelter Hall, Los Angeles, California, CA 90095, UNITED STATES
| | | | - Marcia Cristina Branciforti
- Depatament of Materials Engineering, University of Sao Paulo, AVENIDA TRABALHADOR SÃO-CARLENSE, 400, ARNOLD SCHMITED, SAO CARLOS, Sao Paulo, SAO PAULO, 13566-590, BRAZIL
| | - Ana Maria Minarelli Gaspar
- Department of Morphology, School of Dentistry at Araraquara, Sao Paulo State University Julio de Mesquita Filho, R. Humaitá, 1680, Araraquara, SP, 14801-385, BRAZIL
| | - Su Ryon Shin
- Medicine, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, MA 02115, UNITED STATES
| | - Ali Khademhosseini
- Department of Chemical and Biomolecular Engineering, UCLA, 5531 Boelter Hall, Los Angeles, California, CA 90095, UNITED STATES
| | - Guy Schlatter
- ICPEES, University of Strasbourg, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| |
Collapse
|
17
|
Xu C, Liu H, He Y, Li Y, He X. Endothelial progenitor cells promote osteogenic differentiation in co-cultured with mesenchymal stem cells via the MAPK-dependent pathway. Stem Cell Res Ther 2020; 11:537. [PMID: 33308309 PMCID: PMC7731475 DOI: 10.1186/s13287-020-02056-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The role of bone tissue engineering is to regenerate tissue using biomaterials and stem cell-based approaches. Combination of two or more cell types is one of the strategies to promote bone formation. Endothelial progenitor cells (EPCs) may enhance the osteogenic properties of mesenchymal stem cells (MSCs) and promote bone healing; this study aimed to investigate the possible mechanisms of EPCs on promoting osteogenic differentiation of MSCs. METHODS MSCs and EPCs were isolated and co-cultured in Transwell chambers, the effects of EPCs on the regulation of MSC biological properties were investigated. Real-time PCR array, and western blotting were performed to explore possible signaling pathways involved in osteogenesis. The expression of osteogenesis markers and calcium nodule formation was quantified by qRT-PCR, western blotting, and Alizarin Red staining. RESULTS Results showed that MSCs exhibited greater alkaline phosphatase (ALP) activity and increased calcium mineral deposition significantly when co-cultured with EPCs. The mitogen-activated protein kinase (MAPK) signaling pathway was involved in this process. p38 gene expression and p38 protein phosphorylation levels showed significant upregulation in co-cultured MSCs. Silencing expression of p38 in co-cultured MSCs reduced osteogenic gene expression, protein synthesis, ALP activity, and calcium nodule formation. CONCLUSIONS These data suggest paracrine signaling from EPCs influences the biological function and promotes MSCs osteogenic differentiation. Activation of the p38MAPK pathway may be the key to enhancing MSCs osteogenic differentiation via indirect interactions with EPCs.
Collapse
Affiliation(s)
- Chu Xu
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China.,Department of General Dentistry, School of Stomatology, China Medical University, Shenyang, 110001, Liaoning, China
| | - Haijie Liu
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China
| | - Yuanjia He
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China
| | - Yuanqing Li
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China
| | - Xiaoning He
- Department of Stomatology, The 4th Affiliated Hospital of China Medical University, No.4 Chongshan Dong Road, Shenyang, 110032, Liaoning, China.
| |
Collapse
|
18
|
Ehnert S, Rinderknecht H, Aspera-Werz RH, Häussling V, Nussler AK. Use of in vitro bone models to screen for altered bone metabolism, osteopathies, and fracture healing: challenges of complex models. Arch Toxicol 2020; 94:3937-3958. [PMID: 32910238 PMCID: PMC7655582 DOI: 10.1007/s00204-020-02906-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Approx. every third hospitalized patient in Europe suffers from musculoskeletal injuries or diseases. Up to 20% of these patients need costly surgical revisions after delayed or impaired fracture healing. Reasons for this are the severity of the trauma, individual factors, e.g, the patients' age, individual lifestyle, chronic diseases, medication, and, over 70 diseases that negatively affect the bone quality. To investigate the various disease constellations and/or develop new treatment strategies, many in vivo, ex vivo, and in vitro models can be applied. Analyzing these various models more closely, it is obvious that many of them have limits and/or restrictions. Undoubtedly, in vivo models most completely represent the biological situation. Besides possible species-specific differences, ethical concerns may question the use of in vivo models especially for large screening approaches. Challenging whether ex vivo or in vitro bone models can be used as an adequate replacement for such screenings, we here summarize the advantages and challenges of frequently used ex vivo and in vitro bone models to study disturbed bone metabolism and fracture healing. Using own examples, we discuss the common challenge of cell-specific normalization of data obtained from more complex in vitro models as one example of the analytical limits which lower the full potential of these complex model systems.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany.
| | - Helen Rinderknecht
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| | - Romina H Aspera-Werz
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| | - Victor Häussling
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| | - Andreas K Nussler
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen, Germany
| |
Collapse
|
19
|
Muniswami DM, Reddy LVK, Amirtham SM, Babu S, Raj AN, Sen D, Manivasagam G. Endothelial progenitor/stem cells in engineered vessels for vascular transplantation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:119. [PMID: 33247781 DOI: 10.1007/s10856-020-06458-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/27/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Dysfunction of blood vessel leads to aneurysms, myocardial infarction and other thrombosis conditions. Current treatment strategies are transplantation of blood vessels from one part of the body to other dysfunction area, or allogenic, synthetic. Due to shortage of the donor, painful dissection, and lack of efficacy in synthetic, there is a need for alternative to native blood vessels for transplantation. METHODS Human umbilical-cord tissue obtained from the hospital with the informed consent. Umbilical-cord blood vessels were isolated for decellularization and to establish endothelial cell culture. Cultured cells were characterized by immunophenotype, gene expression and in vitro angiogenesis assay. Decellularized blood vessels were recellularized with the endothelial progenitors and Wharton jelly, CL MSCs (1:1), which was characterized by MTT, biomechanical testing, DNA content, SEM and histologically. Bioengineered vessels were transplanted into the animal models to evaluate their effect. RESULTS Cultured cells express CD31 and CD14 determining endothelial progenitor cells (EPCs). EPCs expresses various factors such as angiopoitin1, VWF, RANTES, VEGF, BDNF, FGF1, FGF2, HGF, IGF, GDNF, NGF, PLGF, NT3, but fail to express NT4, EGF, and CNTF. Pro and anti-inflammatory cytokine expressions were noticed. Functionally, these EPCs elicit in vitro tube formation. Negligible DNA content and intact ECM confirms the efficient decellularization of tissue. The increased MTT activity in recellularized tissue determines proliferating cells and biocompatibility of the scaffolds. Moreover, significant (P < 0.05) increase in maximum force and tensile of recellularized biomaterial as compared to the decellularized scaffolds. Integration of graft with host tissue, suggesting biocompatible therapeutic biomaterial with cells. CONCLUSION EPCs with stem cells in engineered blood vessels could be therapeutically applicable in vascular surgery.
Collapse
Affiliation(s)
- Durai Murugan Muniswami
- Centre for Biomaterials, Cellular & Molecular Theranostics (CBCMT), VIT, Vellore, India.
- Department of Microbiology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore, 641021, India.
| | - L Vinod Kumar Reddy
- Centre for Biomaterials, Cellular & Molecular Theranostics (CBCMT), VIT, Vellore, India
| | | | | | - Arunai Nambi Raj
- Centre for Biomaterials, Cellular & Molecular Theranostics (CBCMT), VIT, Vellore, India
| | - Dwaipayan Sen
- Centre for Biomaterials, Cellular & Molecular Theranostics (CBCMT), VIT, Vellore, India
| | - Geetha Manivasagam
- Centre for Biomaterials, Cellular & Molecular Theranostics (CBCMT), VIT, Vellore, India
| |
Collapse
|
20
|
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
|
21
|
Naudot M, Barre A, Caula A, Sevestre H, Dakpé S, Mueller AA, Devauchelle B, Testelin S, Marolleau JP, Le Ricousse S. Co-transplantation of Wharton's jelly mesenchymal stem cell-derived osteoblasts with differentiated endothelial cells does not stimulate blood vessel and osteoid formation in nude mice models. J Tissue Eng Regen Med 2020; 14:257-271. [PMID: 31713308 DOI: 10.1002/term.2989] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 09/23/2019] [Accepted: 10/11/2019] [Indexed: 12/30/2022]
Abstract
A major challenge in bone tissue engineering is the lack of post-implantation vascular growth into biomaterials. In the skeletal system, blood vessel growth appears to be coupled to osteogenesis-suggesting the existence of molecular crosstalk between endothelial cells (ECs) and osteoblastic cells. The present study (performed in two murine ectopic models) was designed to determine whether co-transplantation of human Wharton's jelly mesenchymal stem cell-derived osteoblasts (WJMSC-OBs) and human differentiated ECs enhances bone regeneration and stimulates angiogenesis, relative to the seeding of WJMSC-OBs alone. Human WJMSC-OBs and human ECs were loaded into a silicate-substituted calcium phosphate (SiCaP) scaffold and then ectopically implanted at subcutaneous or intramuscular sites in nude mice. At both subcutaneous and intramuscular implantation sites, we observed ectopic bone formation and osteoids composed of host cells when WJMSC-OBs were seeded into the scaffold. However, the addition of ECs was associated with a lower level of osteogenesis, and we did not observe stimulation of blood vessel ingrowth. in vitro studies demonstrated that WJMSC-OBs lost their ability to secrete vascular endothelial growth factor and stromal cell-derived factor 1-including when ECs were present. In these two murine ectopic models, our cell-matrix environment combination did not seem to be optimal for inducing vascularized bone reconstruction.
Collapse
Affiliation(s)
- Marie Naudot
- EA 7516, CHIMERE, University of Picardie Jules Verne, Amiens, France
| | - Anaïs Barre
- EA 7516, CHIMERE, University of Picardie Jules Verne, Amiens, France
| | - Alexandre Caula
- Service de chirurgie maxillo-faciale, Centre Hospitalier Universitaire Amiens Picardie, Amiens, France
| | - Henri Sevestre
- Service d'anatomie et de cytology pathologique, Centre Hospitalier Universitaire Amiens Picardie, Amiens, France
| | - Stéphanie Dakpé
- EA 7516, CHIMERE, University of Picardie Jules Verne, Amiens, France.,Service de chirurgie maxillo-faciale, Centre Hospitalier Universitaire Amiens Picardie, Amiens, France.,Institut Faire Faces, Amiens, France
| | - Andreas Albert Mueller
- Department of Cranio-Maxillofacial Surgery, University and University Hospital Basel, Basel, Switzerland.,Department of Biomedical Engineering, Regenerative Medicine and Oral Health Technologies, University of Basel, Allschwil, Switzerland
| | - Bernard Devauchelle
- EA 7516, CHIMERE, University of Picardie Jules Verne, Amiens, France.,Service de chirurgie maxillo-faciale, Centre Hospitalier Universitaire Amiens Picardie, Amiens, France.,Institut Faire Faces, Amiens, France
| | - Sylvie Testelin
- EA 7516, CHIMERE, University of Picardie Jules Verne, Amiens, France.,Service de chirurgie maxillo-faciale, Centre Hospitalier Universitaire Amiens Picardie, Amiens, France.,Institut Faire Faces, Amiens, France
| | - Jean Pierre Marolleau
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire Amiens Picardie, Amiens, France.,EA 4666, HEMATIM, University of Picardie Jules Verne, Amiens, France
| | - Sophie Le Ricousse
- EA 7516, CHIMERE, University of Picardie Jules Verne, Amiens, France.,Institut Faire Faces, Amiens, France
| |
Collapse
|
22
|
Zhou Y, Liu C, He J, Dong L, Zhu H, Zhang B, Feng X, Weng W, Cheng K, Yu M, Wang H. KLF2 + stemness maintains human mesenchymal stem cells in bone regeneration. Stem Cells 2019; 38:395-409. [PMID: 31721356 DOI: 10.1002/stem.3120] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 09/01/2019] [Indexed: 01/04/2023]
Abstract
Mesenchymal stem cells (MSCs), which are undifferentiated stem cells with the property of stemness and the potential to differentiate into multiple lineages, including osteoblasts, have attracted a great deal of attention in bone tissue engineering. Consistent with the heterogeneity of MSCs, various surface markers have been used. However, it is still unclear which markers of MSCs are best for cell amplification in vitro and later bone regeneration in vivo. Krüppel-like Factor 2 (KLF2) is an important indicator of the stemness of human MSCs (hMSCs) and as early vascularization is also critical for bone regeneration, we used KLF2 as a novel in vitro marker for MSCs and investigated the angiogenesis and osteogenesis between KLF2+ MSCs and endothelial cells (ECs). We found a synergistic interaction between hMSCs and human umbilical vein ECs (HUVECs) in that KLF2+ stemness-maintained hMSCs initially promoted the angiogenesis of HUVECs, which in turn more efficiently stimulated the osteogenesis of hMSCs. In fact, KLF2+ hMSCs secreted angiogenic factors initially, with some of the cells then differentiating into pericytes through the PDGF-BB/PDGFR-β signaling pathway, which improved blood vessel formation. The matured HUVECs in turn synergistically enhanced the osteogenesis of KLF2+ hMSCs through upregulated vascular endothelial growth factor. A three-dimensional coculture model using cell-laden gelatin methacrylate (GelMA) hydrogel further confirmed these results. This study provides insight into the stemness-directed synergistic interaction between hMSCs and HUVECs, and our results will have a profound impact on further strategies involving the application of KLF2+ hMSC/HUVEC-laden GelMA hydrogel in vascular network bioengineering and bone regeneration.
Collapse
Affiliation(s)
- Ying Zhou
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Chao Liu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jianxiang He
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Lingqing Dong
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- State Key Laboratory of Silicon Materials, Cyrus Tang Center for Sensor Materials and Applications, School of Materials Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Huiyong Zhu
- The First Affiliated Hospital of Medical College, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Bin Zhang
- The State Key Laboratory of Fluid Power Transmission and Control, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoxia Feng
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Wenjian Weng
- State Key Laboratory of Silicon Materials, Cyrus Tang Center for Sensor Materials and Applications, School of Materials Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Kui Cheng
- State Key Laboratory of Silicon Materials, Cyrus Tang Center for Sensor Materials and Applications, School of Materials Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- The First Affiliated Hospital of Medical College, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Huiming Wang
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- The First Affiliated Hospital of Medical College, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
23
|
Núñez-Toldrà R, Montori S, Bosch B, Hupa L, Atari M, Miettinen S. S53P4 Bioactive Glass Inorganic Ions for Vascularized Bone Tissue Engineering by Dental Pulp Pluripotent-Like Stem Cell Cocultures. Tissue Eng Part A 2019; 25:1213-1224. [DOI: 10.1089/ten.tea.2018.0256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Raquel Núñez-Toldrà
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Barcelona, Spain
| | - Sheyla Montori
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Begoña Bosch
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Leena Hupa
- Johan Gadolin Process Chemistry Centre, Åbo Akademi University, Turku, Finland
| | - Maher Atari
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
- Surgery and Oral Implantology Department, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Susanna Miettinen
- Adult Stem Cell Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
24
|
Deegan AJ, Hendrikson WJ, El Haj AJ, Rouwkema J, Yang Y. Regulation of endothelial cell arrangements within hMSC - HUVEC co-cultured aggregates. Biomed J 2019; 42:166-177. [PMID: 31466710 PMCID: PMC6717755 DOI: 10.1016/j.bj.2019.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/25/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background Micro-mass culturing or cellular aggregation is an effective method used to form mineralised bone tissue. Poor core cell viability, however, is often an impeding characteristic of large micro-mass cultures, and equally for large tissue-engineered bone grafts. Because of this, efforts are being made to enhance large graft perfusion, often through pre-vascularisation, which involves the co-culture of endothelial cells and bone cells or stem cells. Methods This study investigated the effects of different aggregation techniques and culture conditions on endothelial cell arrangements in mesenchymal stem cell and human umbilical vein endothelial cell co-cultured aggregates when endothelial cells constituted just 5%. Two different cellular aggregation techniques, i.e. suspension culture aggregation and pellet culture aggregation, were applied alongside two subsequent culturing techniques, i.e. hydrostatic loading and static culturing. Endothelial cell arrangements were assessed under such conditions to indicate potential pre-vascularisation. Results Our study found that the suspension culture aggregates cultured under hydrostatic loading offered the best environment for enhanced endothelial cell regional arrangements, closely followed by the pellet culture aggregates cultured under hydrostatic loading, the suspension culture aggregates cultured under static conditions, and the pellet culture aggregates cultured under static conditions. Conclusions The combination of particular aggregation techniques with dynamic culturing conditions appeared to have a synergistic effect on the cellular arrangements within the co-cultured aggregates.
Collapse
Affiliation(s)
- Anthony J Deegan
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Wim J Hendrikson
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, AE, the Netherlands
| | - Alicia J El Haj
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Ying Yang
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom.
| |
Collapse
|
25
|
Schneider J, Pultar M, Holnthoner W. Ex vivo engineering of blood and lymphatic microvascular networks. VASCULAR BIOLOGY 2019; 1:H17-H22. [PMID: 32923949 PMCID: PMC7439851 DOI: 10.1530/vb-19-0012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
Upon implantation, engineered tissues rely on the supply with oxygen and nutrients as well as the drainage of interstitial fluid. This prerequisite still represents one of the current challenges in the engineering and regeneration of tissues. Recently, different vascularization strategies have been developed. Besides technical approaches like 3D printing or laser processing and de-/recelluarization of natural scaffolds, mainly co-cultures of endothelial cells (ECs) with supporting cell types are being used. This mini-review provides a brief overview of different co-culture systems for the engineering of blood and lymphatic microvascular networks.
Collapse
Affiliation(s)
- Jaana Schneider
- Ludwig-Boltzmann-Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Marianne Pultar
- Ludwig-Boltzmann-Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Wolfgang Holnthoner
- Ludwig-Boltzmann-Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
26
|
Wang K, Lin RZ, Melero-Martin JM. Bioengineering human vascular networks: trends and directions in endothelial and perivascular cell sources. Cell Mol Life Sci 2019; 76:421-439. [PMID: 30315324 PMCID: PMC6349493 DOI: 10.1007/s00018-018-2939-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022]
Abstract
Tissue engineering holds great promise in regenerative medicine. However, the field of tissue engineering faces a myriad of difficulties. A major challenge is the necessity to integrate vascular networks into bioengineered constructs to enable physiological functions including adequate oxygenation, nutrient delivery, and removal of waste products. The last two decades have seen remarkable progress in our collective effort to bioengineer human-specific vascular networks. Studies have included both in vitro and in vivo investigations, and multiple methodologies have found varying degrees of success. What most approaches to bioengineer human vascular networks have in common, however, is the synergistic use of both (1) endothelial cells (ECs)-the cells used to line the lumen of the vascular structures and (2) perivascular cells-usually used to support EC function and provide perivascular stability to the networks. Here, we have highlighted trends in the use of various cellular sources over the last two decades of vascular network bioengineering research. To this end, we comprehensively reviewed all life science and biomedical publications available at the MEDLINE database up to 2018. Emphasis was put on selective studies that definitively used human ECs and were specifically related to bioengineering vascular networks. To facilitate this analysis, all papers were stratified by publication year and then analyzed according to their use of EC and perivascular cell types. This study provides an illustrating discussion on how each alternative source of cells has come to be used in the field. Our intention was to reveal trends and to provide new insights into the trajectory of vascular network bioengineering with regard to cellular sources.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - 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
|
27
|
Osteogenic and angiogenic characterization of mandible and femur osteoblasts. J Mol Histol 2019; 50:105-117. [PMID: 30635760 DOI: 10.1007/s10735-019-09810-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/06/2019] [Indexed: 12/13/2022]
|
28
|
Verboket R, Leiblein M, Seebach C, Nau C, Janko M, Bellen M, Bönig H, Henrich D, Marzi I. Autologous cell-based therapy for treatment of large bone defects: from bench to bedside. Eur J Trauma Emerg Surg 2018; 44:649-665. [PMID: 29352347 PMCID: PMC6182650 DOI: 10.1007/s00068-018-0906-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/08/2018] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Reconstruction of long segmental bone defects is demanding for patients and surgeons, and associated with long-term treatment periods and substantial complication rates in addition to high costs. While defects up to 4-5 cm length might be filled up with autologous bone graft, heterologous bone from cadavers, or artificial bone graft substitutes, current options to reconstruct bone defects greater than 5 cm consist of either vascularized free bone transfers, the Masquelet technique or the Ilizarov distraction osteogenesis. Alternatively, autologous cell transplantation is an encouraging treatment option for large bone defects as it eliminates problems such as limited autologous bone availability, allogenic bone immunogenicity, and donor-site morbidity, and might be used for stabilizing loose alloplastic implants. METHODS The authors show different cell therapies without expansion in culture, with ex vivo expansion and cell therapy in local bone defects, bone healing and osteonecrosis. Different kinds of cells and scaffolds investigated in our group as well as in vivo transfer studies and BMC used in clinical phase I and IIa clinical trials of our group are shown. RESULTS Our research history demonstrated the great potential of various stem cell species to support bone defect healing. It was clearly shown that the combination of different cell types is superior to approaches using single cell types. We further demonstrate that it is feasible to translate preclinically developed protocols from in vitro to in vivo experiments and follow positive convincing results into a clinical setting to use autologous stem cells to support bone healing.
Collapse
Affiliation(s)
- R. Verboket
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - M. Leiblein
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - C. Seebach
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - C. Nau
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - M. Janko
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - M. Bellen
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - H. Bönig
- Department of Transfusion Medicine and Immune Hematology, University Hospital Frankfurt and DRK Blood Donor Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - D. Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - I. Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
29
|
Hughes BR, Mirbagheri M, Waldman SD, Hwang DK. Direct cell-cell communication with three-dimensional cell morphology on wrinkled microposts. Acta Biomater 2018; 78:89-97. [PMID: 30092377 DOI: 10.1016/j.actbio.2018.07.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/23/2018] [Accepted: 07/29/2018] [Indexed: 01/09/2023]
Abstract
Cell-cell communication plays a critical role in a myriad of processes, such as homeostasis, angiogenesis, and carcinogenesis, in multi-cellular organisms. Monolayer cell models have notably improved our understanding of cellular interactions. However, the cultured cells on the planar surfaces adopt a two-dimensional morphology, which poorly imitates cellular organization in vivo, providing physiologically-irrelevant cell responses. Non-planar surfaces comprising various patterns have demonstrated great abilities in directing cellular growth and producing different cell morphologies. In recent years, a few topographical substrates have provided valuable information about cell-cell signalling, however, none of these studies have reported a three-dimensional (3D) cell morphology. Here, we introduce a structurally tunable topographical platform that can maintain cell coupling while inducing a true 3D cell morphology. Optical imaging and fluorescence recovery after photobleaching are used to illustrate these capabilities. Our analyses suggest that the intercellular signalling on the present platform, which we propose is mainly through gap junctions, is comparable to that in natural tissue. STATEMENT OF SIGNIFICANCE A better understanding of direct cellular communication can help treating neurological diseases and cancers, which may be caused by dysfunctional intercellular signaling. To investigate cell-cell contact, cells are conventionally plated onto planar surfaces, where they flatten and adopt a two-dimensional cell morphology. These unrealistic models are physiologically-irrelevant since cells exhibit a three-dimensional (3D) shape in the body. Therefore, porous scaffolds and topographical surfaces, capable of inducing various cell morphologies, have been introduced, in which the latter is more desirable for sample imaging and screening. However, the few non-planar substrates used to study cell coupling have not produced a 3D cell shape. Here, we present a tunable culture platform that can control direct cell-cell communication while maintaining true 3D cell morphologies.
Collapse
|
30
|
Simunovic F, Winninger O, Strassburg S, Koch HG, Finkenzeller G, Stark GB, Lampert FM. Increased differentiation and production of extracellular matrix components of primary human osteoblasts after cocultivation with endothelial cells: A quantitative proteomics approach. J Cell Biochem 2018; 120:396-404. [PMID: 30126049 DOI: 10.1002/jcb.27394] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022]
Abstract
Coculturing of bone-forming and blood vessel-forming cells is a strategy aimed at increasing vascularity of implanted bone constructs in tissue-engineering applications. We previously described that the coculture of primary human osteoblasts (hOBs) and human umbilical vein endothelial cells (HUVECs) improves the differentiation of both cell types, leading to the formation of functional blood vessels and enhanced bone regeneration. The objective of this study was to further delineate the multifaceted interactions between both cell types. To investigate the proteome of hOBs after cocultivation with HUVECs we used stable isotope labeling by amino acids in cell culture, revealing 49 significantly upregulated, and 54 significantly downregulated proteins. Amongst the highest regulated proteins, we found the proteins important for osteoblast differentiation, cellular adhesion, and extracellular matrix function, notably: connective tissue growth factor, desmoplakin, galectin-3, and cyclin-dependent kinase 6. The findings were confirmed by enzyme-linked immunosorbent assays. We also investigated whether the mRNA transcripts correlate with the changes in protein levels by quantitative real-time reverse transcription polymerase chain reaction. In addition, the data was compared to our previous microarray analysis of hOB transcriptome. Taken together, this in-depth analysis delivers reliable data suggesting the importance of coculturing of hOBs and HUVECs in tissue engineering.
Collapse
Affiliation(s)
- F Simunovic
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - O Winninger
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - S Strassburg
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - H G Koch
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, Albert-Ludwigs University Freiburg, Freiburg, Germany
| | - G Finkenzeller
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - G B Stark
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - F M Lampert
- Department of Plastic and Hand Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
31
|
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
|
32
|
Laurent J, Blin G, Chatelain F, Vanneaux V, Fuchs A, Larghero J, Théry M. Convergence of microengineering and cellular self-organization towards functional tissue manufacturing. Nat Biomed Eng 2017; 1:939-956. [DOI: 10.1038/s41551-017-0166-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/07/2017] [Indexed: 12/18/2022]
|
33
|
Herath TDK, Larbi A, Teoh SH, Kirkpatrick CJ, Goh BT. Neutrophil-mediated enhancement of angiogenesis and osteogenesis in a novel triple cell co-culture model with endothelial cells and osteoblasts. J Tissue Eng Regen Med 2017; 12:e1221-e1236. [PMID: 28715156 DOI: 10.1002/term.2521] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 06/02/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023]
Abstract
Repair and regeneration of critical-sized bone defects remain a major challenge in orthopaedic and craniomaxillofacial surgery. Until now, attempts to bioengineer bone tissue have been hindered by the inability to establish proper angiogenesis and osteogenesis in the tissue construct. In the present study, we established a novel triple cell co-culture model consisting of osteoblasts, endothelial cells, and neutrophils and conducted a systematic investigation of the effects of neutrophils on angiogenesis and osteogenesis. Neutrophils significantly increased angiogenesis in the tissue construct, evidenced by the formation of microvessel-like structures with an extensive lattice-like, stable tubular network in the co-culture model. Moreover, neutrophils significantly induced the expression of pro-angiogenic markers, such as VEGF-A, CD34, EGF, and FGF-2 in a dose- and time-dependent manner. Subsequently, PCR arrays corroborated that neutrophils upregulate the important angiogenic markers and MMPs. Moreover, neutrophils also enhanced osteogenic markers, such as ALP, OCN, OPN, and COL-1 compared with the controls. As shown by the osteogenic gene arrays, neutrophils significantly regulated major osteogenic markers such as BMP2, BMP3, BMP4, BMP5, TGF-β2, RUNX2, and ECM proteins. Significantly higher mineralization was observed in triple cell co-culture compared with controls. Foregoing data indicate that the triple cell co-culture model can be used to stimulate the growth of microvasculature within a bone bioengineering construct to improve cell viability. Neutrophil-mediated enhancement of angiogenesis and osteogenesis could be a viable, clinically relevant tissue engineering strategy to obtain optimal bone growth in defect sites, in the field of oral and maxillofacial surgery.
Collapse
Affiliation(s)
| | - Anis Larbi
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Swee Hin Teoh
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - C James Kirkpatrick
- REPAIR-Lab, Institute of Pathology, University Medical Centre, Johannes Gutenberg University, Mainz, Germany
| | | |
Collapse
|
34
|
Manipulation of Human Primary Endothelial Cell and Osteoblast Coculture Ratios to Augment Vasculogenesis and Mineralization. Ann Plast Surg 2017; 77:122-8. [PMID: 25144419 DOI: 10.1097/sap.0000000000000318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tissue-engineering scaffolds are often seeded with a single type of cell, but there has been more focus on cocultures to improve angiogenesis and bone formation for craniofacial applications. Investigation of bone-derived osteoblasts (OBs) is important because of the use of bone grafts and migration of OBs from native bone into constructs in vivo and therefore, their contribution to bone formation in vivo. The limitation of primary OBs has been their inability to mineralize without osteogenic factors in vitro. Through coculture of OBs and endothelial cells (ECs) and manipulation of the coculture ratio, mineralization can be achieved without osteogenic media or additional growth factors, thus enhancing their utility for tissue-engineering applications. An optimal ratio of EC/OB for vasculogenesis and mineralization has not been determined for human primary cells. Human umbilical vein ECs were cultured with normal human primary OBs in different EC/OB ratios, namely, 10:1, 5:1, 1:1, 1:5, and 1:10 with EC and OB monocultures as controls. The number of vasculogenic networks in a collagen matrix was highest in ratios of 5:1 and 1:1. ECs lined up and formed capillary-like networks by day 10, which was not seen in the other groups. On polystyrene, cells were cocultured with ECs and OBs in direct contact (direct coculture) or separated by a transwell membrane (indirect coculture). At day 21, Alizarin Red staining showed mineralization on the 1:5 and 1:10 direct coculture ratios, with 1:5 having more mineralization nodules present than 1:10. No mineralization was seen in other direct coculture ratios or in any of the indirect coculture ratios. Alkaline phosphatase secretion was highest in the 1:5 direct coculture group. Vascular endothelial growth factor secretion from OBs was present in the 1:5 and 1:10 direct coculture ratios at all time points and inhibited after day 1 in other coculture groups. To improve vasculogenesis, cocultures of primary human ECs and OBs in ratios of 5:1 should be used, but to improve bone formation, the 1:5 direct coculture ratio results in most mineralization.
Collapse
|
35
|
Dohle E, El Bagdadi K, Sader R, Choukroun J, James Kirkpatrick C, Ghanaati S. Platelet-rich fibrin-based matrices to improve angiogenesis in an in vitro co-culture model for bone tissue engineering. J Tissue Eng Regen Med 2017; 12:598-610. [PMID: 28509340 PMCID: PMC5888144 DOI: 10.1002/term.2475] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022]
Abstract
In the context of prevascularization strategies for tissue‐engineering purposes, co‐culture systems consisting of outgrowth endothelial cells (OECs) and primary osteoblasts (pOBs) have been established as a promising in vitro tool to study regeneration mechanisms and to identify factors that might positively influence repair processes such as wound healing or angiogenesis. The development of autologous injectable platelet‐rich fibrin (PRF), which can be generated from peripheral blood in a minimal invasive procedure, fulfils several requirements for clinically applicable cell‐based tissue‐engineering strategies. During this study, the established co‐culture system of OECs and pOBs was mixed with injectable PRF and was cultivated in vitro for 24 h or 7 days. The aim of this study was to analyse whether PRF might have a positive effect on wound healing processes and angiogenic activation of OECs in the co‐culture with regard to proinflammatory factors, adhesion molecules and proangiogenic growth factor expression. Histological cell detection revealed the formation of lumina and microvessel‐like structures in the PRF/co‐culture complexes after 7 days of complex cultivation. Interestingly, the angiogenic activation of OECs was accompanied by an upregulation of wound healing‐associated factors, as well as by a higher expression of the proangiogenic factor vascular endothelial growth factor, which was evaluated both on the mRNA level as well as on the protein level. Thus, PRF might positively influence wound healing processes, in particular angiogenesis, in the in vitro co‐culture, making autologous PRF‐based matrices a beneficial therapeutic tool for tissue‐engineering purposes by simply profiting from the PRF, which contains blood plasma, platelets and leukocytes.
Collapse
Affiliation(s)
- Eva Dohle
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Karima El Bagdadi
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Robert Sader
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Joseph Choukroun
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Johann Wolfgang Goethe University, Frankfurt am Main, Germany.,Pain Therapy Center, Nice, France
| | - C James Kirkpatrick
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Johann Wolfgang Goethe University, Frankfurt am Main, Germany.,Department of Biomaterials, Gothenburg, Sweden
| | - Shahram Ghanaati
- FORM, Frankfurt Oral Regenerative Medicine, Clinic for Maxillofacial and Plastic Surgery, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
36
|
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
|
37
|
Abstract
Background. The polysaccharide fucoidan is widely investigated as an anti-cancer agent. Here, we tested the effect of fucoidan on uveal melanoma cell lines. Methods. The effect of 100 µM fucoidan was investigated on five cell lines (92.1, Mel270 OMM1, OMM2.3, OMM2.5) and of 1 µg/mL–1 mg/mL fucoidan in two cell lines (OMM1, OMM2.3). Cell proliferation and viability were investigated with a WST-1 assay, migration in a wound healing (scratch) assay. Vascular Endothelial Growth Factor (VEGF) was measured in ELISA. Angiogenesis was evaluated in co-cultures with endothelial cells. Cell toxicity was induced by hydrogen-peroxide. Protein expression (Akt, ERK1/2, Bcl-2, Bax) was investigated in Western blot. Results. Fucoidan increased proliferation in two and reduced it in one cell line. Migration was reduced in three cell lines. The effect of fucoidan on VEGF was cell type and concentration dependent. In endothelial co-culture with 92.1, fucoidan significantly increased tubular structures. Moreover, fucoidan significantly protected all tested uveal melanoma cell lines from hydrogen-peroxide induced cell death. Under oxidative stress, fucoidan did not alter the expression of Bcl-2, Bax or ERK1/2, while inducing Akt expression in 92.1 cells but not in any other cell line. Conclusion. Fucoidan did not show anti-tumorigenic effects but displayed protective and pro-angiogenic properties, rendering fucoidan unsuitable as a potential new drug for the treatment of uveal melanoma.
Collapse
|
38
|
Wu Z, Yang T, Bao C, Li M, Xu HHK, Liao X, Li L, Huang R, Pan J, Liu X. Effect of Electrospun Fibrous Scaffolds with Different Fiber Orientations on the Alignment of Microvessel-Like Structures. J Med Biol Eng 2017. [DOI: 10.1007/s40846-017-0284-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
Chen W, Liu X, Chen Q, Bao C, Zhao L, Zhu Z, Xu HHK. Angiogenic and osteogenic regeneration in rats via calcium phosphate scaffold and endothelial cell co-culture with human bone marrow mesenchymal stem cells (MSCs), human umbilical cord MSCs, human induced pluripotent stem cell-derived MSCs and human embryonic stem cell-derived MSCs. J Tissue Eng Regen Med 2017; 12:191-203. [PMID: 28098961 DOI: 10.1002/term.2395] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 10/03/2016] [Accepted: 01/09/2017] [Indexed: 02/05/2023]
Abstract
Angiogenesis is a limiting factor in regenerating large bone defects. The objective of this study was to investigate angiogenic and osteogenic effects of co-culture on calcium phosphate cement (CPC) scaffold using human umbilical vein endothelial cells (hUVECs) and mesenchymal stem cells (MSCs) from different origins for the first time. hUVECs were co-cultured with four types of cell: human umbilical cord MSCs (hUCMSCs), human bone marrow MSCs (hBMSCs) and MSCs from induced pluripotent stem cells (hiPSC-MSCs) and embryonic stem cells (hESC-MSCs). Constructs were implanted in 8 mm cranial defects of rats for 12 weeks. CPC without cells served as control 1. CPC with hBMSCs served as control 2. Microcapillary-like structures were successfully formed on CPC in vitro in all four co-cultured groups. Microcapillary lengths increased with time (p < 0.05). Osteogenic and angiogenic gene expressions were highly elevated and mineralization by co-cultured cells increased with time (p < 0.05). New bone amount and blood vessel density of co-cultured groups were much greater than controls (p < 0.05) in an animal study. hUVECs co-cultured with hUCMSCs, hiPSC-MSCs and hESC-MSCs achieved new bone and vessel density similar to hUVECs co-cultured with hBMSCs (p > 0.1). Therefore, hUCMSCs, hiPSC-MSCs and hESC-MSCs could serve as alternative cell sources to hBMSCs, which require an invasive procedure to harvest. In conclusion, this study showed for the first time that co-cultures of hUVECs with hUCMSCs, hiPSC-MSCs, hESC-MSCs and hBMSCs delivered via CPC scaffold achieved excellent osteogenic and angiogenic capabilities in vivo. The novel co-culture constructs are promising for bone reconstruction with improved angiogenesis for craniofacial/orthopaedic applications. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Wenchuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Xian Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Qianmin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Liang Zhao
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA.,Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhimin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, University of Maryland at Baltimore County, Baltimore County, MD, USA
| |
Collapse
|
40
|
Combined chemical and structural signals of biomaterials synergistically activate cell-cell communications for improving tissue regeneration. Acta Biomater 2017; 55:249-261. [PMID: 28377306 DOI: 10.1016/j.actbio.2017.03.056] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/24/2017] [Accepted: 03/31/2017] [Indexed: 01/20/2023]
Abstract
Biomaterials are only used as carriers of cells in the conventional tissue engineering. Considering the multi-cell environment and active cell-biomaterial interactions in tissue regeneration process, in this study, structural signals of aligned electrospun nanofibers and chemical signals of bioglass (BG) ionic products in cell culture medium are simultaneously applied to activate fibroblast-endothelial co-cultured cells in order to obtain an improved skin tissue engineering construct. Results demonstrate that the combined biomaterial signals synergistically activate fibroblast-endothelial co-culture skin tissue engineering constructs through promotion of paracrine effects and stimulation of gap junctional communication between cells, which results in enhanced vascularization and extracellular matrix protein synthesis in the constructs. Structural signals of aligned electrospun nanofibers play an important role in stimulating both of paracrine and gap junctional communication while chemical signals of BG ionic products mainly enhance paracrine effects. In vivo experiments reveal that the activated skin tissue engineering constructs significantly enhance wound healing as compared to control. This study indicates the advantages of synergistic effects between different bioactive signals of biomaterials can be taken to activate communication between different types of cells for obtaining tissue engineering constructs with improved functions. STATEMENT OF SIGNIFICANCE Tissue engineering can regenerate or replace tissue or organs through combining cells, biomaterials and growth factors. Normally, for repairing a specific tissue, only one type of cells, one kind of biomaterials, and specific growth factors are used to support cell growth. In this study, we proposed a novel tissue engineering approach by simply using co-cultured cells and combined biomaterial signals. Using a skin tissue engineering model, we successfully proved that the combined biomaterial signals such as surface nanostructures and bioactive ions could synergistically stimulate the cell-cell communication in co-culture system through paracrine effects and gap junction activation, and regulated expression of growth factors and extracellular matrix proteins, resulting in an activated tissue engineering constructs that significantly enhanced skin regeneration.
Collapse
|
41
|
Wei D, Sun J, Bolderson J, Zhong M, Dalby MJ, Cusack M, Yin H, Fan H, Zhang X. Continuous Fabrication and Assembly of Spatial Cell-Laden Fibers for a Tissue-Like Construct via a Photolithographic-Based Microfluidic Chip. ACS APPLIED MATERIALS & INTERFACES 2017; 9:14606-14617. [PMID: 28157291 DOI: 10.1021/acsami.7b00078] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Engineering three-dimensional (3D) scaffolds with in vivo like architecture and function has shown great potential for tissue regeneration. Here we developed a facile microfluidic-based strategy for the continuous fabrication of cell-laden microfibers with hierarchically organized architecture. We show that photolithographically fabricated microfluidic devices offer a simple and reliable way to create anatomically inspired complex structures. Furthermore, the use of photo-cross-linkable methacrylated alginate allows modulation of both the mechanical properties and biological activity of the hydrogels for targeted applications. Via this approach, multilayered hollow microfibers were continuously fabricated, which can be easily assembled in situ, using 3D printing, into a larger, tissue-like construct. Importantly, this biomimetic approach promoted the development of phenotypical functions of the target tissue. As a model to engineer a complex tissue construct, osteon-like fiber was biomimetically engineered, and enhanced vasculogenic and osteogenic expression were observed in the encapsulated human umbilical cord vein endothelial cells and osteoblast-like MG63 cells respectively within the osteon fibers. The capability of this approach to create functional building blocks will be advantageous for bottom-up regeneration of complex, large tissue defects and, more broadly, will benefit a variety of applications in tissue engineering and biomedical research.
Collapse
Affiliation(s)
- Dan Wei
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064, Sichuan, China
| | - Jing Sun
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064, Sichuan, China
| | - Jason Bolderson
- Division of Biomedical Engineering, School of Engineering, University of Glasgow , Glasgow G12 8LT, U.K
| | - Meiling Zhong
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064, Sichuan, China
| | | | | | - Huabing Yin
- Division of Biomedical Engineering, School of Engineering, University of Glasgow , Glasgow G12 8LT, U.K
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064, Sichuan, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University , Chengdu 610064, Sichuan, China
| |
Collapse
|
42
|
Xu Y, Wu Z, Dong X, Li H. Combined biomaterial signals stimulate communications between bone marrow stromal cell and endothelial cell. RSC Adv 2017. [DOI: 10.1039/c6ra28101j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Combined chemical and structural signals of biomaterials stimulate communications between bone marrow stromal cell and endothelial cell.
Collapse
Affiliation(s)
- Yachen Xu
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Zhi Wu
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Xin Dong
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Haiyan Li
- Med-X Research Institute
- School of Biomedical Engineering
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| |
Collapse
|
43
|
Bardsley K, Deegan AJ, El Haj A, Yang Y. Current State-of-the-Art 3D Tissue Models and Their Compatibility with Live Cell Imaging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1035:3-18. [PMID: 29080127 DOI: 10.1007/978-3-319-67358-5_1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mammalian cells grow within a complex three-dimensional (3D) microenvironment where multiple cells are organized and surrounded by extracellular matrix (ECM). The quantity and types of ECM components, alongside cell-to-cell and cell-to-matrix interactions dictate cellular differentiation, proliferation and function in vivo. To mimic natural cellular activities, various 3D tissue culture models have been established to replace conventional two dimensional (2D) culture environments. Allowing for both characterization and visualization of cellular activities within possibly bulky 3D tissue models presents considerable challenges due to the increased thickness and subsequent light scattering features of such 3D models. In this chapter, state-of-the-art methodologies used to establish 3D tissue models are discussed, first with a focus on both scaffold-free and scaffold-based 3D tissue model formation. Following on, multiple 3D live cell imaging systems, mainly optical imaging modalities, are introduced. Their advantages and disadvantages are discussed, with the aim of stimulating more research in this highly demanding research area.
Collapse
Affiliation(s)
- Katie Bardsley
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Anthony J Deegan
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Alicia El Haj
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Ying Yang
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK.
| |
Collapse
|
44
|
Freeman FE, McNamara LM. Endochondral Priming: A Developmental Engineering Strategy for Bone Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:128-141. [PMID: 27758156 DOI: 10.1089/ten.teb.2016.0197] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue engineering and regenerative medicine have significant potential to treat bone pathologies by exploiting the capacity for bone progenitors to grow and produce tissue constituents under specific biochemical and physical conditions. However, conventional tissue engineering approaches, which combine stem cells with biomaterial scaffolds, are limited as the constructs often degrade, due to a lack of vascularization, and lack the mechanical integrity to fulfill load bearing functions, and as such are not yet widely used for clinical treatment of large bone defects. Recent studies have proposed that in vitro tissue engineering approaches should strive to simulate in vivo bone developmental processes and, thereby, imitate natural factors governing cell differentiation and matrix production, following the paradigm recently defined as "developmental engineering." Although developmental engineering strategies have been recently developed that mimic specific aspects of the endochondral ossification bone formation process, these findings are not widely understood. Moreover, a critical comparison of these approaches to standard biomaterial-based bone tissue engineering has not yet been undertaken. For that reason, this article presents noteworthy experimental findings from researchers focusing on developing an endochondral-based developmental engineering strategy for bone tissue regeneration. These studies have established that in vitro approaches, which mimic certain aspects of the endochondral ossification process, namely the formation of the cartilage template and the vascularization of the cartilage template, can promote mineralization and vascularization to a certain extent both in vitro and in vivo. Finally, this article outlines specific experimental challenges that must be overcome to further exploit the biology of endochondral ossification and provide a tissue engineering construct for clinical treatment of large bone/nonunion defects and obviate the need for bone tissue graft.
Collapse
Affiliation(s)
- Fiona E Freeman
- Centre for Biomechanics Research (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway , Galway, Ireland
| | - Laoise M McNamara
- Centre for Biomechanics Research (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway , Galway, Ireland
| |
Collapse
|
45
|
Freeman FE, Stevens HY, Owens P, Guldberg RE, McNamara LM. Osteogenic Differentiation of Mesenchymal Stem Cells by Mimicking the Cellular Niche of the Endochondral Template. Tissue Eng Part A 2016; 22:1176-1190. [PMID: 27604384 DOI: 10.1089/ten.tea.2015.0339] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In vitro bone regeneration strategies that prime mesenchymal stem cells (MSCs) with chondrogenic factors, to mimic aspects of the endochondral ossification process, have been shown to promote mineralization and vascularization by MSCs both in vitro and when implanted in vivo. However, these approaches required the use of osteogenic supplements, namely dexamethasone, ascorbic acid, and β-glycerophosphate, none of which are endogenous mediators of bone formation in vivo. Rather MSCs, endothelial progenitor cells, and chondrocytes all reside in proximity within the cartilage template and might paracrineally regulate osteogenic differentiation. Thus, this study tests the hypothesis that an in vitro bone regeneration approach that mimics the cellular niche existing during endochondral ossification, through coculture of MSCs, endothelial cells, and chondrocytes, will obviate the need for extraneous osteogenic supplements and provide an alternative strategy to elicit osteogenic differentiation of MSCs and mineral production. The specific objectives of this study were to (1) mimic the cellular niche existing during endochondral ossification and (2) investigate whether osteogenic differentiation could be induced without the use of any external growth factors. To test the hypothesis, we evaluated the mineralization and vessel formation potential of (a) a novel methodology involving both chondrogenic priming and the coculture of human umbilical vein endothelial cells (HUVECs) and MSCs compared with (b) chondrogenic priming of MSCs alone, (c) addition of HUVECs to chondrogenically primed MSC aggregates, (d-f) the same experimental groups cultured in the presence of osteogenic supplements and (g) a noncoculture group cultured in the presence of osteogenic growth factors alone. Biochemical (DNA, alkaline phosphatase [ALP], calcium, CD31+, vascular endothelial growth factor [VEGF]), histological (alcian blue, alizarin red), and immunohistological (CD31+) analyses were conducted to investigate osteogenic differentiation and vascularization at various time points (1, 2, and 3 weeks). The coculture methodology enhanced both osteogenesis and vasculogenesis compared with osteogenic differentiation alone, whereas osteogenic supplements inhibited the osteogenesis and vascularization (ALP, calcium, and VEGF) induced through coculture alone. Taken together, these results suggest that chondrogenic and vascular priming can obviate the need for osteogenic supplements to induce osteogenesis of human MSCs in vitro, while allowing for the formation of rudimentary vessels.
Collapse
Affiliation(s)
- Fiona E Freeman
- 1 Biomedical Engineering, Centre for Biomechanics Research (BMEC), National University of Ireland Galway , Galway, Ireland
| | - Hazel Y Stevens
- 2 George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | - Peter Owens
- 3 Centre for Microscopy and Imaging, National University of Ireland , Galway, Galway, Ireland
| | - Robert E Guldberg
- 2 George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | - Laoise M McNamara
- 1 Biomedical Engineering, Centre for Biomechanics Research (BMEC), National University of Ireland Galway , Galway, Ireland
| |
Collapse
|
46
|
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]
|
47
|
Jeong SY, Lee JH, Shin Y, Chung S, Kuh HJ. Co-Culture of Tumor Spheroids and Fibroblasts in a Collagen Matrix-Incorporated Microfluidic Chip Mimics Reciprocal Activation in Solid Tumor Microenvironment. PLoS One 2016; 11:e0159013. [PMID: 27391808 PMCID: PMC4938568 DOI: 10.1371/journal.pone.0159013] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/31/2016] [Indexed: 11/27/2022] Open
Abstract
Multicellular 3D culture and interaction with stromal components are considered essential elements in establishing a ‘more clinically relevant’ tumor model. Matrix-embedded 3D cultures using a microfluidic chip platform can recapitulate the microscale interaction within tumor microenvironments. As a major component of tumor microenvironment, cancer-associated fibroblasts (CAFs) play a role in cancer progression and drug resistance. Here, we present a microfluidic chip-based tumor tissue culture model that integrates 3D tumor spheroids (TSs) with CAF in proximity within a hydrogel scaffold. HT-29 human colorectal carcinoma cells grew into 3D TSs and the growth was stimulated when co-cultured with fibroblasts as shown by 1.5-folds increase of % changes in diameter over 5 days. TS cultured for 6 days showed a reduced expression of Ki-67 along with increased expression of fibronectin when co-cultured with fibroblasts compared to mono-cultured TSs. Fibroblasts were activated under co-culture conditions, as demonstrated by increases in α-SMA expression and migratory activity. When exposed to paclitaxel, a survival advantage was observed in TSs co-cultured with activated fibroblasts. Overall, we demonstrated the reciprocal interaction between TSs and fibroblasts in our 7-channel microfluidic chip. The co-culture of 3D TS-CAF in a collagen matrix-incorporated microfluidic chip may be useful to study the tumor microenvironment and for evaluation of drug screening and evaluation.
Collapse
Affiliation(s)
- Su-Yeong Jeong
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji-Hyun Lee
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoojin Shin
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
48
|
Johari B, Ahmadzadehzarajabad M, Azami M, Kazemi M, Soleimani M, Kargozar S, Hajighasemlou S, Farajollahi MM, Samadikuchaksaraei A. Repair of rat critical size calvarial defect using osteoblast-like and umbilical vein endothelial cells seeded in gelatin/hydroxyapatite scaffolds. J Biomed Mater Res A 2016; 104:1770-8. [DOI: 10.1002/jbm.a.35710] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 03/01/2016] [Accepted: 03/04/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Behrooz Johari
- Department of Medical Biotechnology Faculty of Allied Medicine; Tehran University of Medical Sciences; Tehran Iran
- Department of Biotechnology; Pasteur Institute of Iran, Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Maryam Ahmadzadehzarajabad
- Department of Pharmaceutical Biotechnology School of Pharmacy; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mahmoud Azami
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Mansure Kazemi
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Mansooreh Soleimani
- Department of Anatomy Faculty of Medicine; Iran University of Medical Sciences; Tehran Iran
- Cellular and Molecular Research Center; Iran University of Medical Sciences; Tehran Iran
| | - Saied Kargozar
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Saieh Hajighasemlou
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Mohammad M Farajollahi
- Cellular and Molecular Research Center; Iran University of Medical Sciences; Tehran Iran
- Department of Medical Biotechnology Faculty of Allied Medicine; Iran University of Medical Sciences; Tehran Iran
| | - Ali Samadikuchaksaraei
- Cellular and Molecular Research Center; Iran University of Medical Sciences; Tehran Iran
- Department of Medical Biotechnology Faculty of Allied Medicine; Iran University of Medical Sciences; Tehran Iran
- Department of Tissue Engineering and Regenerative Medicine Faculty of Advanced Technologies in Medicine; Iran University of Medical Sciences; Tehran Iran
| |
Collapse
|
49
|
Hoppe A, Brandl A, Bleiziffer O, Arkudas A, Horch RE, Jokic B, Janackovic D, Boccaccini AR. In vitro cell response to Co-containing 1393 bioactive glass. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 57:157-63. [DOI: 10.1016/j.msec.2015.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 06/21/2015] [Accepted: 07/09/2015] [Indexed: 02/07/2023]
|
50
|
Improving vascularization of engineered bone through the generation of pro-angiogenic effects in co-culture systems. Adv Drug Deliv Rev 2015; 94:116-25. [PMID: 25817732 DOI: 10.1016/j.addr.2015.03.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/27/2015] [Accepted: 03/20/2015] [Indexed: 01/31/2023]
Abstract
One of the major problems with bone tissue engineering is the development of a rapid vascularization after implantation to supply the growing osteoblast cells with the nutrients to grow and survive as well as to remove waste products. It has been demonstrated that capillary-like structures produced in vitro will anastomose rapidly after implantation and become functioning blood vessels. For this reason, in recent years many studies have examined a variety of human osteoblast and endothelial cell co-culture systems in order to distribute osteoblasts on all parts of the bone scaffold and at the same time provide conditions for the endothelial cells to migrate to form a network of capillary-like structures throughout the osteoblast-colonized scaffold. The movement and proliferation of endothelial cells to form capillary-like structures is known as angiogenesis and is dependent on a variety of pro-angiogenic factors. This review summarizes human 2- and 3-D co-culture models to date, the types and origins of cells used in the co-cultures and the proangiogenic factors that have been identified in the co-culture models.
Collapse
|