51
|
Influence of scaffold properties on the inter-relationship between human bone marrow derived stromal cells and endothelial cells in pro-osteogenic conditions. Acta Biomater 2015; 25:16-23. [PMID: 26162586 DOI: 10.1016/j.actbio.2015.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/06/2015] [Accepted: 07/06/2015] [Indexed: 12/16/2022]
Abstract
One of the significant challenges in bone tissue engineering is the integration of biomaterials designed to facilitate and stimulate mineralization with a simultaneously rapid rate of angiogenesis and vascularization of the tissue construct, a challenge complicated by our lack of knowledge of the interactions among key cell types and scaffold properties. This study compared functional activity of human bone marrow-derived stromal cells (hMSC) seeded onto a porous salt-leached poly(D,L-lactic acid) (PDLLA) scaffolds, with and without the incorporation of silk fibroin fibers and then further investigated their co-culture with human umbilical vein endothelial cells (HUVECs). Cell viability, proliferation, and alkaline phosphatase activity were measured for a range of time points in culture, with osteogenic and angiogenic marker immunolocalization and gene expression at selected stages. Our findings suggest that, despite similar porosity and pore size distribution exhibited by the PDLLA and PDLLA plus silk fibroin scaffolds, there were marked differences in cell distribution and function. In the absence of fibers, a highly osteogenic response was observed in hMSCs in the scaffolds co-cultured with endothelial cells, greater than that observed with hMSCs alone or in either of the scaffolds with fibers added. However, fiber presence clearly better supported endothelial cell cultures, as determined by greater levels of endothelial marker expression at both the gene and protein level after 3 weeks of culture. The design of composite scaffolds integrating beneficial components of differing structures and materials to facilitate appropriate biological responses appears a promising yet challenging avenue of research. STATEMENT OF SIGNIFICANCE A significant challenge in bone tissue engineering is to promote a rapid vascularization of the tissue construct in parallel to the extracellular matrix mineralization. The design of composite scaffolds integrating beneficial components of differing structures and materials to facilitate appropriate biological responses appears a promising yet challenging avenue of research. Here we investigated cultures of hMSCs and HUVECs on a silk fibroin enhanced PDLLA scaffold, showing that the final output of this in vitro system is not the linear sum of the effects of the single variables. These results are of interest as they demonstrate how the addition of endothelial cells can affect hMSC phenotype and that the output can be further modulated by the introduction of silk fibroin fibers.
Collapse
|
52
|
Endothelial Progenitor Cell Fraction Contained in Bone Marrow-Derived Mesenchymal Stem Cell Populations Impairs Osteogenic Differentiation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:659542. [PMID: 26491682 PMCID: PMC4600555 DOI: 10.1155/2015/659542] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/28/2015] [Indexed: 12/11/2022]
Abstract
In bone tissue engineering (TE) endothelial cell-osteoblast cocultures are known to induce synergies of cell differentiation and activity. Bone marrow mononucleated cells (BMCs) are a rich source of mesenchymal stem cells (MSCs) able to develop an osteogenic phenotype. Endothelial progenitor cells (EPCs) are also present within BMC. In this study we investigate the effect of EPCs present in the BMC population on MSCs osteogenic differentiation. Human BMCs were isolated and separated into two populations. The MSC population was selected through plastic adhesion capacity. EPCs (CD34+ and CD133+) were removed from the BMC population and the resulting population was named depleted MSCs. Both populations were cultured over 28 days in osteogenic medium (Dex+) or medium containing platelet lysate (PL). MSC population grew faster than depleted MSCs in both media, and PL containing medium accelerated the proliferation for both populations. Cell differentiation was much higher in Dex+ medium in both cases. Real-time RT-PCR revealed upregulation of osteogenic marker genes in depleted MSCs. Higher values of ALP activity and matrix mineralization analyses confirmed these results. Our study advocates that absence of EPCs in the MSC population enables higher osteogenic gene expression and matrix mineralization and therefore may lead to advanced bone neoformation necessary for TE constructs.
Collapse
|
53
|
Schmidt Y, Simunovic F, Strassburg S, Pfeifer D, Stark GB, Finkenzeller G. miR-126 regulates platelet-derived growth factor receptor-α expression and migration of primary human osteoblasts. Biol Chem 2015; 396:61-70. [PMID: 25205714 DOI: 10.1515/hsz-2014-0168] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/26/2014] [Indexed: 12/11/2022]
Abstract
Adequate vascularization is an essential requirement for bone development, fracture healing and bone tissue engineering. We have previously described the coculture of primary human osteoblasts (hOBs) and human endothelial cells (HUVECs), designed to investigate the interactions between these cells. In this system, we showed that cocultivation of these two cell types leads to a downregulation of platelet-derived growth factor receptor-α (PDGFR-α) in hOBs, which was a consequence of reduced mRNA stability. In the current study we investigated the possible involvement of microRNAs in this process. Firstly, we performed a microarray analysis of osteoblastic miRNAs following cocultivation with HUVECs, revealing an upregulation of miR-126. This result was confirmed by RT-qPCR, and we observed that the increase is dependent on direct cell-to-cell contacts. Gain-of-function and loss-of-function experiments showed that miR-126 is a negative regulator of PDGFR-α mRNA. Additionally, migration of hOBs was inhibited by miR-126 overexpression and stimulated by miR-126 inhibition. Addition of PDGFR-α blocking antibody to hOB culture also inhibited hOB migration. There was no effect of miR-126 modulation on osteoblast proliferation, apoptosis rate or differentiation. In conclusion, we report that the miR-126/PDGFR-α system regulates the migratory behavior of human osteoblasts, without exerting effects on cell survival and differentiation.
Collapse
|
54
|
Jaatinen L, Vörös J, Hyttinen J. Controlling cell migration and adhesion into a scaffold by external electric currents. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2015:3549-3552. [PMID: 26737059 DOI: 10.1109/embc.2015.7319159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Fabrication of more complex tissue-engineered structures, resembling the tissues and organs in vivo requires combining more than one cell type within the same construct. This can be achieved by designing and fabricating complex scaffolds with asymmetric properties but controlled arrangement of cells within the scaffold could also be realized by using electric current. External electric currents are able to modify cell adhesion, orientation and migration and this can be used for influencing cell location within a scaffold. In this paper we studied the effect of an electric current on cell migration and adhesion into a three-dimensional scaffold through a conductive mesh.
Collapse
|
55
|
Leijten J, Chai Y, Papantoniou I, Geris L, Schrooten J, Luyten F. Cell based advanced therapeutic medicinal products for bone repair: Keep it simple? Adv Drug Deliv Rev 2015; 84:30-44. [PMID: 25451134 DOI: 10.1016/j.addr.2014.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 09/18/2014] [Accepted: 10/20/2014] [Indexed: 02/08/2023]
Abstract
The development of cell based advanced therapeutic medicinal products (ATMPs) for bone repair has been expected to revolutionize the health care system for the clinical treatment of bone defects. Despite this great promise, the clinical outcomes of the few cell based ATMPs that have been translated into clinical treatments have been far from impressive. In part, the clinical outcomes have been hampered because of the simplicity of the first wave of products. In response the field has set-out and amassed a plethora of complexities to alleviate the simplicity induced limitations. Many of these potential second wave products have remained "stuck" in the development pipeline. This is due to a number of reasons including the lack of a regulatory framework that has been evolving in the last years and the shortage of enabling technologies for industrial manufacturing to deal with these novel complexities. In this review, we reflect on the current ATMPs and give special attention to novel approaches that are able to provide complexity to ATMPs in a straightforward manner. Moreover, we discuss the potential tools able to produce or predict 'goldilocks' ATMPs, which are neither too simple nor too complex.
Collapse
|
56
|
Freeman FE, Haugh MG, McNamara LM. An in vitro bone tissue regeneration strategy combining chondrogenic and vascular priming enhances the mineralization potential of mesenchymal stem cells in vitro while also allowing for vessel formation. Tissue Eng Part A 2015; 21:1320-32. [PMID: 25588588 DOI: 10.1089/ten.tea.2014.0249] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chondrogenic priming (CP) of mesenchymal stem cells (MSCs) and coculture of MSCs with human umbilical vein endothelial stem cells (HUVECs) both have been shown to significantly increase the potential for MSCs to undergo osteogenic differentiation and mineralization in vitro and in vivo. Such strategies mimic cartilage template formation or vascularization that occur during endochondral ossification during early fetal development. However, although both chondrogenesis and vascularization are crucial precursors for bone formation by endochondral ossification, no in vitro bone tissue regeneration strategy has sought to incorporate both events simultaneously. The objective of this study is to develop an in vitro bone regeneration strategy that mimics critical aspects of the endochondral ossification process, specifically (1) the formation of a cartilage template and (2) subsequent vascularization of this template. We initially prime the MSCs with chondrogenic growth factors, to ensure the production of a cartilage template, and subsequently implement a coculture strategy involving MSC and HUVECs. Three experimental groups were compared; (1) CP for 21 days with no addition of cells; (2) CP for 21 days followed by coculture of HUVECs (250,000 cells); (3) CP for 21 days followed by coculture of HUVECs and MSCs (250,000 cells) at a ratio of 1:1. Each group was cultured for a further 21 days in osteogenic media after the initial CP period. Biochemical (DNA, Alkaline Phosphatase Activity, Calcium, and Vessel Endothelial Growth Factor) and histological analyses (Alcian blue, alizarin red, CD31(+), and collagen type X) were performed 1, 2, and 3 weeks after the media switch. The results of this study show that CP provides a cartilage-like template that provides a suitable platform for HUVEC and MSC cells to attach, proliferate, and infiltrate for up to 3 weeks. More importantly we show that the use of the coculture methodology, rudimentary vessels are formed within this cartilage template and enhanced the mineralization potential of MSCs. Taken together these results indicate for the first time that the application of both chondrogenic and vascular priming of MSCs enhances the mineralization potential of MSCs in vitro while also allowing the formation of immature vessels.
Collapse
Affiliation(s)
- Fiona E Freeman
- 1 Centre for Biomechanics Research (BMEC), Biomedical Engineering , NUI Galway, Galway, Ireland
| | | | | |
Collapse
|
57
|
Grayson WL, Bunnell BA, Martin E, Frazier T, Hung BP, Gimble JM. Stromal cells and stem cells in clinical bone regeneration. Nat Rev Endocrinol 2015; 11:140-50. [PMID: 25560703 PMCID: PMC4338988 DOI: 10.1038/nrendo.2014.234] [Citation(s) in RCA: 312] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stem-cell-mediated bone repair has been used in clinical trials for the regeneration of large craniomaxillofacial defects, to slow the process of bone degeneration in patients with osteonecrosis of the femoral head and for prophylactic treatment of distal tibial fractures. Successful regenerative outcomes in these investigations have provided a solid foundation for wider use of stromal cells in skeletal repair therapy. However, employing stromal cells to facilitate or enhance bone repair is far from being adopted into clinical practice. Scientific, technical, practical and regulatory obstacles prevent the widespread therapeutic use of stromal cells. Ironically, one of the major challenges lies in the limited understanding of the mechanisms via which transplanted cells mediate regeneration. Animal models have been used to provide insight, but these models largely fail to reproduce the nuances of human diseases and bone defects. Consequently, the development of targeted approaches to optimize cell-mediated outcomes is difficult. In this Review, we highlight the successes and challenges reported in several clinical trials that involved the use of bone-marrow-derived mesenchymal or adipose-tissue-derived stromal cells. We identify several obstacles blocking the mainstream use of stromal cells to enhance skeletal repair and highlight technological innovations or areas in which novel techniques might be particularly fruitful in continuing to advance the field of skeletal regenerative medicine.
Collapse
Affiliation(s)
- Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Bruce A Bunnell
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Elizabeth Martin
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Trivia Frazier
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Ben P Hung
- Department of Biomedical Engineering, Johns Hopkins University, 400 North Broadway, Baltimore, MD 21205, USA
| | - Jeffrey M Gimble
- Centre for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| |
Collapse
|
58
|
Fu WL, Xiang Z, Huang FG, Gu ZP, Yu XX, Cen SQ, Zhong G, Duan X, Liu M. Coculture of Peripheral Blood-Derived Mesenchymal Stem Cells and Endothelial Progenitor Cells on Strontium-Doped Calcium Polyphosphate Scaffolds to Generate Vascularized Engineered Bone. Tissue Eng Part A 2015; 21:948-59. [PMID: 25298026 DOI: 10.1089/ten.tea.2014.0267] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Wei-Li Fu
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Zhou Xiang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Fu-Guo Huang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Zhi-Peng Gu
- College of Polymer Science and Engineering, Sichuan University, Chengdu, P.R. China
| | - Xi-Xun Yu
- College of Polymer Science and Engineering, Sichuan University, Chengdu, P.R. China
| | - Shi-Qiang Cen
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Gang Zhong
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xin Duan
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Ming Liu
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
59
|
Osteoblastic alkaline phosphatase mRNA is stabilized by binding to vimentin intermediary filaments. Biol Chem 2015; 396:253-60. [DOI: 10.1515/hsz-2014-0274] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 12/17/2014] [Indexed: 01/04/2023]
Abstract
Abstract
Vascularization is essential in bone tissue engineering and recent research has focused on interactions between osteoblasts (hOBs) and endothelial cells (ECs). It was shown that cocultivation increases the stability of osteoblastic alkaline phosphatase (ALP) mRNA. We investigated the mechanisms behind this observation, focusing on mRNA binding proteins. Using a luciferase reporter assay, we found that the 3′-untranslated region (UTR) of ALP mRNA is necessary for human umbilical vein endothelial cells (HUVEC)-mediated stabilization of osteoblastic ALP mRNA. Using pulldown experiments and nanoflow-HPLC mass spectrometry, vimentin was identified to bind to the 3′-UTR of ALP mRNA. Validation was performed by Western blotting. Functional experiments inhibiting intermediate filaments with iminodipropionitrile and specific inhibition of vimentin by siRNA transfection showed reduced levels of ALP mRNA and protein. Therefore, ALP mRNA binds to and is stabilized by vimentin. This data add to the understanding of intracellular trafficking of ALP mRNA, its function, and have possible implications in tissue engineering applications.
Collapse
|
60
|
In vitro co-culture strategies to prevascularization for bone regeneration: A brief update. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-014-0095-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
61
|
Li J, Ma Y, Teng R, Guan Q, Lang J, Fang J, Long H, Tian G, Wu Q. Transcriptional profiling reveals crosstalk between mesenchymal stem cells and endothelial cells promoting prevascularization by reciprocal mechanisms. Stem Cells Dev 2014; 24:610-23. [PMID: 25299975 DOI: 10.1089/scd.2014.0330] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) show great promise in blood vessel restoration and vascularization enhancement in many therapeutic situations. Typically, the co-implantation of MSCs with vascular endothelial cells (ECs) is effective for the induction of functional vascularization in vivo, indicating its potential applications in regenerative medicine. The effects of MSCs-ECs-induced vascularization can be modeled in vitro, providing simplified models for understanding their underlying communication. In this article, a contact coculture model in vitro and an RNA-seq approach were employed to reveal the active crosstalk between MSCs and ECs within a short time period at both morphological and transcriptional levels. The RNA-seq results suggested that angiogenic genes were significantly induced upon coculture, and this prevascularization commitment might require the NF-κB signaling. NF-κB blocking and interleukin (IL) neutralization experiments demonstrated that MSCs potentially secreted IL factors including IL1β and IL6 to modulate NF-κB signaling and downstream chemokines during coculture. Conversely, RNA-seq results indicated that the MSCs were regulated by the coculture environment to a smooth muscle commitment within this short period, which largely induced myocardin, the myogenic co-transcriptional factor. These findings demonstrate the mutual molecular mechanism of MSCs-ECs-induced prevascularization commitment in a quick response.
Collapse
Affiliation(s)
- Junxiang Li
- 1 School of Life Sciences, Tsinghua University , Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Im GI. Coculture in Musculoskeletal Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:545-54. [DOI: 10.1089/ten.teb.2013.0731] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Gun-Il Im
- Department of Orthopaedics, Dongguk University Ilsan Hospital, Goyang, Korea
| |
Collapse
|
63
|
Rohringer S, Hofbauer P, Schneider KH, Husa AM, Feichtinger G, Peterbauer-Scherb A, Redl H, Holnthoner W. Mechanisms of vasculogenesis in 3D fibrin matrices mediated by the interaction of adipose-derived stem cells and endothelial cells. Angiogenesis 2014; 17:921-33. [PMID: 25086616 DOI: 10.1007/s10456-014-9439-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/19/2014] [Indexed: 12/22/2022]
Abstract
Vascularization of tissue-engineered constructs is essential to provide sufficient nutrient supply and hemostasis after implantation into target sites. Co-cultures of adipose-derived stem cells (ASC) with outgrowth endothelial cells (OEC) in fibrin gels were shown to provide an effective possibility to induce vasculogenesis in vitro. However, the mechanisms of the interaction between these two cell types remain unclear so far. The aim of this study was to evaluate differences of direct and indirect stimulation of ASC-induced vasculogenesis, the influence of ASC on network stabilization and molecular mechanisms involved in vascular structure formation. Endothelial cells (EC) were embedded in fibrin gels either containing non-coated or ASC-coated microcarrier beads as well as ASC alone. Moreover, EC-seeded constructs incubated with ASC-conditioned medium were used in addition to constructs with ASC seeded on top. Vascular network formation was visualized by green fluorescent protein expressing cells or immunostaining for CD31 and quantified. RT-qPCR of cells derived from co-cultures in fibrin was performed to evaluate changes in the expression of EC marker genes during the first week of culture. Moreover, angiogenesis-related protein levels were measured by performing angiogenesis proteome profiler arrays. The results demonstrate that proximity of endothelial cells and ASC is required for network formation and ASC stabilize EC networks by developing pericyte characteristics. We further showed that ASC induce controlled vessel growth by secreting pro-angiogenic and regulatory proteins. This study reveals angiogenic protein profiles involved in EC/ASC interactions in fibrin matrices and confirms the usability of OEC/ASC co-cultures for autologous vascular tissue engineering.
Collapse
Affiliation(s)
- Sabrina Rohringer
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Guerreiro SG, Oliveira MJ, Barbosa MA, Soares R, Granja PL. Neonatal Human Dermal Fibroblasts Immobilized in RGD–Alginate Induce Angiogenesis. Cell Transplant 2014; 23:945-57. [PMID: 23866308 DOI: 10.3727/096368913x670183] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Promoting angiogenesis in a damaged tissue is a major challenge for tissue regeneration. Recent findings in tissue engineering suggest that fibroblasts (FBs) play an important role in orchestrating the angiogenic process. Fibroblasts maintain the structural integrity of connective tissue by continuously secreting growth factors and extracellular matrix precursors, which are essential for endothelial cell (EC) adhesion and spreading, thus playing a crucial role in angiogenesis. We hypothesized that FBs immobilized in alginate gels grafted with the RGD peptidic sequence could influence the recruitment of ECs to improve vascularization. In this work, the modulation of immobilized human FBs within the 3D synthetic extracellular matrix was assessed. Experiments using cocultures of ECs and FBs in indirect contact as well as angiogenic assays were performed to assess the influence of FBs immobilized in RGD–alginate in ECs' viability, stabilization, sprouting, and assembly into capillary-like structures. This study demonstrates the ability of FBs immobilized within RGD–alginate microspheres to modulate and support capillary-like structures' assembly. These findings indicate that the microenvironment created by these stromal cells in the scaffold modulates capillary morphogenesis, thus stimulating angiogenesis in situ and can potentially be used in regenerative medicine in clinical scenarios where vascularization is essential.
Collapse
Affiliation(s)
- Susana G. Guerreiro
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Medicina (FMUP), Departamento de Bioquímica, Porto, Portugal
- Universidade do Porto, Faculdade de Engenharia (FEUP), Porto, Portugal
| | - Maria J. Oliveira
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Medicina (FMUP), Departamento de Anatomia Patológica, Porto, Portugal
| | - Mário A. Barbosa
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Engenharia (FEUP), Porto, Portugal
- Universidade do Porto, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Porto, Portugal
| | - Raquel Soares
- Universidade do Porto, Faculdade de Medicina (FMUP), Departamento de Bioquímica, Porto, Portugal
| | - Pedro L. Granja
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Engenharia (FEUP), Porto, Portugal
| |
Collapse
|
65
|
Koç A, Finkenzeller G, Elçin AE, Stark GB, Elçin YM. Evaluation of adenoviral vascular endothelial growth factor-activated chitosan/hydroxyapatite scaffold for engineering vascularized bone tissue using human osteoblasts: In vitro and in vivo studies. J Biomater Appl 2014; 29:748-60. [PMID: 25062670 DOI: 10.1177/0885328214544769] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bone tissue is dependent on an efficient blood supply to ensure delivery of nutrients and oxygen. One method to acquire a vascular-engineered bone tissue could be the use of an angiogenic gene-activated scaffold. In the current study, porous chitosan/hydroxyapatite (C/HA) scaffolds were fabricated via freeze-drying with desired pore size, and then combined with the adenoviral vector encoding vascular endothelial growth factor and green fluorescence protein (Ad-VEGF). Human osteoblasts were cultured and seeded on characterized scaffolds. The attachment, proliferation, and differentiation of cells on gene-activated and unactivated C/HA scaffolds were evaluated in vitro and in vivo by histo- and immunohistochemistry. Findings confirmed that human osteoblasts cultured on gene-activated C/HA scaffold secreted vascular endothelial growth factor, besides maintaining its characteristic phenotype with specific extracellular matrix production. In vivo experiments indicated that scaffolds were tissue biocompatible, and that gene-activated scaffold provided a suitable environment for neovessel formation by recruiting host endothelial cells into the newly forming ectopic bone-like tissue. This study revealed that the Ad-VEGF-activated C/HA composite scaffold has potential for vascular bone regeneration applications.
Collapse
Affiliation(s)
- Aysel Koç
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Günter Finkenzeller
- Department of Plastic and Hand Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | - A Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science and Ankara University Stem Cell Institute, Ankara, Turkey
| | - G Björn Stark
- Department of Plastic and Hand Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | - Y Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science and Ankara University Stem Cell Institute, Ankara, Turkey
| |
Collapse
|
66
|
Chong MSK, Lim J, Goh J, Sia MW, Chan JKY, Teoh SH. Cocultures of Mesenchymal Stem Cells and Endothelial Cells As Organotypic Models of Prostate Cancer Metastasis. Mol Pharm 2014; 11:2126-33. [DOI: 10.1021/mp500141b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mark S. K. Chong
- Division
of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Jing Lim
- Division
of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Junwei Goh
- Bioengineering Laboratory, Technology Centre
for Life Sciences, Singapore Polytechnic, Singapore 139651
| | - Ming W. Sia
- Division
of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Jerry K. Y. Chan
- Department of Obstetrics
and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077
- Department
of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore 229899
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore 169857
| | - Swee H. Teoh
- Division
of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| |
Collapse
|
67
|
Ritz U, Spies V, Mehling I, Gruszka D, Rommens PM, Hofmann A. Mobilization of CD34+-progenitor cells in patients with severe trauma. PLoS One 2014; 9:e97369. [PMID: 24826895 PMCID: PMC4020858 DOI: 10.1371/journal.pone.0097369] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/17/2014] [Indexed: 01/31/2023] Open
Abstract
Circulating CD34+ progenitor cells () gained importance in the field of regenerative medicine due to their potential to home in on injury sites and differentiate into cells of both endothelial and osteogenic lineages. In this study, we analyzed the mobilization kinetics and the numbers of CD34+, CD31+, CD45+, and CD133+ cells in twenty polytrauma patients (n = 13 male, n = 7 female, mean age 46.5±17.2 years, mean injury severity score (ISS) 35.8±12.5 points). In addition, the endothelial differentiation capacity of enriched CD34+cells was assessed by analyzing DiI-ac-LDL/lectin uptake, the expression of endothelial markers, and the morphological characteristics of these cells in Matrigel and spheroid cultures. We found that on days 1, 3, and 7 after a major trauma, the number of CD34+cells increased from 6- up to 12-fold (p<0.0001) over the number of CD34+cells from a control population of healthy, age-matched volunteers. The numbers of CD31+ cells were consistently higher on days 1 (1.4-fold, p<0.01) and 7 (1.3-fold, p<0.01), whereas the numbers of CD133+ cell did not change during the time course of investigation. Expression of endothelial marker molecules in CD34+cells was significantly induced in the polytrauma patients. In addition, we show that the CD34+ cell levels in severely injured patients were not correlated with clinical parameters, such as the ISS score, the acute physiology and chronic health evaluation II score (APACHE II), as well as the sequential organ failure assessment score (SOFA-2). Our results clearly indicate that pro-angiogenic cells are systemically mobilized after polytrauma and that their numbers are sufficient for the development of novel therapeutic models in regenerative medicine.
Collapse
Affiliation(s)
- Ulrike Ritz
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Volker Spies
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Isabella Mehling
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Dominik Gruszka
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Pol Maria Rommens
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
| | - Alexander Hofmann
- BiomaTiCS-Group, University Medical Centre of the Johannes Gutenberg University, Center of Orthopaedic and Trauma Surgery, Mainz, Germany
- * E-mail:
| |
Collapse
|
68
|
Kirkpatrick CJ. Developing Cellular Systems In Vitro to Simulate Regeneration. Tissue Eng Part A 2014; 20:1355-7. [DOI: 10.1089/ten.tea.2014.0002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Charles James Kirkpatrick
- REPAIR-Lab, Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Department of Biomaterials, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
69
|
Cell communication in a coculture system consisting of outgrowth endothelial cells and primary osteoblasts. BIOMED RESEARCH INTERNATIONAL 2014; 2014:320123. [PMID: 24967356 PMCID: PMC4016919 DOI: 10.1155/2014/320123] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/29/2014] [Accepted: 03/12/2014] [Indexed: 12/13/2022]
Abstract
Bone tissue is a highly vascularized and dynamic system with a complex construction. In order to develop a construct for implant purposes in bone tissue engineering, a proper understanding of the complex dependencies between different cells and cell types would provide further insight into the highly regulated processes during bone repair, namely, angiogenesis and osteogenesis, and might result in sufficiently equipped constructs to be beneficial to patients and thereby accomplish their task. This study is based on an in vitro coculture model consisting of outgrowth endothelial cells and primary osteoblasts and is currently being used in different studies of bone repair processes with special regard to angiogenesis and osteogenesis. Coculture systems of OECs and pOBs positively influence the angiogenic potential of endothelial cells by inducing the formation of angiogenic structures in long-term cultures. Although many studies have focused on cell communication, there are still numerous aspects which remain poorly understood. Therefore, the aim of this study is to investigate certain growth factors and cell communication molecules that are important during bone repair processes. Selected growth factors like VEGF, angiopoietins, BMPs, and IGFs were investigated during angiogenesis and osteogenesis and their expression in the cultures was observed and compared after one and four weeks of cultivation. In addition, to gain a better understanding on the origin of different growth factors, both direct and indirect coculture strategies were employed. Another important focus of this study was to investigate the role of “gap junctions,” small protein pores which connect adjacent cells. With these bridges cells are able to exchange signal molecules, growth factors, and other important mediators. It could be shown that connexins, the gap junction proteins, were located around cell nuclei, where they await their transport to the cell membrane. In addition, areas in which two cells formed gap junctions were found.
Collapse
|
70
|
Dithmer M, Fuchs S, Shi Y, Schmidt H, Richert E, Roider J, Klettner A. Fucoidan reduces secretion and expression of vascular endothelial growth factor in the retinal pigment epithelium and reduces angiogenesis in vitro. PLoS One 2014; 9:e89150. [PMID: 24558482 PMCID: PMC3928431 DOI: 10.1371/journal.pone.0089150] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 01/15/2014] [Indexed: 12/20/2022] Open
Abstract
Fucoidan is a polysaccharide isolated from brown algae which is of current interest for anti-tumor therapy. In this study, we investigated the effect of fucoidan on the retinal pigment epithelium (RPE), looking at physiology, vascular endothelial growth factor (VEGF) secretion, and angiogenesis, thus investigating a potential use of fucoidan for the treatment of exudative age-related macular degeneration. For this study, human RPE cell line ARPE-19 and primary porcine RPE cells were used, as well as RPE/choroid perfusion organ cultures. The effect of fucoidan on RPE cells was investigated with methyl thiazolyl tetrazolium--assay, trypan blue exclusion assay, phagocytosis assay and a wound healing assay. VEGF expression was evaluated in immunocytochemistry and Western blot, VEGF secretion was evaluated in ELISA. The effect of fucoidan on angiogenesis was tested in a Matrigel assay using calcein-AM vital staining, evaluated by confocal laser scanning microcopy and quantitative image analysis. Fucoidan displays no toxicity and does not diminish proliferation or phagocytosis, but reduces wound healing in RPE cells. Fucoidan decreases VEGF secretion in RPE/choroid explants and RPE cells. Furthermore, it diminishes VEGF expression in RPE cells even when co-applied with bevacizumab. Furthermore, fucoidan reduces RPE-supernatant- and VEGF-induced angiogenesis of peripheral endothelial cells. In conclusion, fucoidan is a non-toxic agent that reduces VEGF expression and angiogenesis in vitro and may be of interest for further studies as a potential therapy against exudative age-related macular degeneration.
Collapse
Affiliation(s)
- Michaela Dithmer
- University of Kiel, University Medical Center, Department of Ophthalmology, Kiel, Germany
| | - Sabine Fuchs
- University of Kiel, University Medical Center, Experimental Trauma Surgery, Kiel, Germany
| | - Yang Shi
- University of Kiel, University Medical Center, Experimental Trauma Surgery, Kiel, Germany
| | | | - Elisabeth Richert
- University of Kiel, University Medical Center, Department of Ophthalmology, Kiel, Germany
| | - Johann Roider
- University of Kiel, University Medical Center, Department of Ophthalmology, Kiel, Germany
| | - Alexa Klettner
- University of Kiel, University Medical Center, Department of Ophthalmology, Kiel, Germany
- * E-mail:
| |
Collapse
|
71
|
Paschos NK, Brown WE, Eswaramoorthy R, Hu JC, Athanasiou KA. Advances in tissue engineering through stem cell-based co-culture. J Tissue Eng Regen Med 2014; 9:488-503. [PMID: 24493315 DOI: 10.1002/term.1870] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/19/2013] [Accepted: 01/03/2014] [Indexed: 12/13/2022]
Abstract
Stem cells are the future in tissue engineering and regeneration. In a co-culture, stem cells not only provide a target cell source with multipotent differentiation capacity, but can also act as assisting cells that promote tissue homeostasis, metabolism, growth and repair. Their incorporation into co-culture systems seems to be important in the creation of complex tissues or organs. In this review, critical aspects of stem cell use in co-culture systems are discussed. Direct and indirect co-culture methodologies used in tissue engineering are described, along with various characteristics of cellular interactions in these systems. Direct cell-cell contact, cell-extracellular matrix interaction and signalling via soluble factors are presented. The advantages of stem cell co-culture strategies and their applications in tissue engineering and regenerative medicine are portrayed through specific examples for several tissues, including orthopaedic soft tissues, bone, heart, vasculature, lung, kidney, liver and nerve. A concise review of the progress and the lessons learned are provided, with a focus on recent developments and their implications. It is hoped that knowledge developed from one tissue can be translated to other tissues. Finally, we address challenges in tissue engineering and regenerative medicine that can potentially be overcome via employing strategies for stem cell co-culture use.
Collapse
Affiliation(s)
- Nikolaos K Paschos
- Department of Biomedical Engineering and Orthopedic Surgery, University of California at Davis, CA, 95616, USA
| | | | | | | | | |
Collapse
|
72
|
Kirkpatrick J. Developing cellular systems in vitro to simulate regeneration. Tissue Eng Part A 2014. [DOI: 10.1089/ten.tea.2013.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
73
|
Brown A, Burke GA, Meenan BJ. Patterned cell culture substrates created by hot embossing of tissue culture treated polystyrene. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:2797-2807. [PMID: 23900705 DOI: 10.1007/s10856-013-5011-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/16/2013] [Indexed: 05/28/2023]
Abstract
Patterning materials such that they elicit a different cell response in different regions would have significant implications in fields such as implantable biomaterials, in vitro cell culture and tissue engineering and regenerative medicine. Moreover, the ability to pattern polymers using inexpensive, currently available processes, without the need for adding proteins or other biochemical agents could lead to new opportunities in biomaterials research. The research reported here demonstrates that by combining the plasma surface treatments used to create commercial grade tissue culture treated polystyrene, with controlled hot embossing processes, that distinct regions can be created on a substrate that result in spatial control of endothelial cell adhesion and proliferation. As well as the topographical changes that result from hot embossing, significant changes in surface chemistry and wettability have been observed and characterised and the resultant effects on endothelial cell responses evaluated. By spatially controlling endothelial cell adhesion, proliferation and subsequent angiogenesis, the processes outlined here have the potential to be used to create a range of different substrates, with applications in the development of assays for high throughput screening, the patterning of implantable biomaterials or the development of smart scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Alan Brown
- University of Ulster, Shore Road, Newtownabbey, BT37 0QB, UK,
| | | | | |
Collapse
|
74
|
Eldesoqi K, Seebach C, Nguyen Ngoc C, Meier S, Nau C, Schaible A, Marzi I, Henrich D. High calcium bioglass enhances differentiation and survival of endothelial progenitor cells, inducing early vascularization in critical size bone defects. PLoS One 2013; 8:e79058. [PMID: 24244419 PMCID: PMC3828289 DOI: 10.1371/journal.pone.0079058] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 09/21/2013] [Indexed: 01/21/2023] Open
Abstract
Early vascularization is a prerequisite for successful bone healing and endothelial progenitor cells (EPC), seeded on appropriate biomaterials, can improve vascularization. The type of biomaterial influences EPC function with bioglass evoking a vascularizing response. In this study the influence of a composite biomaterial based on polylactic acid (PLA) and either 20 or 40% bioglass, BG20 and BG40, respectively, on the differentiation and survival of EPCs in vitro was investigated. Subsequently, the effect of the composite material on early vascularization in a rat calvarial critical size defect model with or without EPCs was evaluated. Human EPCs were cultured with β-TCP, PLA, BG20 or BG40, and seeding efficacy, cell viability, cell morphology and apoptosis were analysed in vitro. BG40 released the most calcium, and improved endothelial differentiation and vitality best. This effect was mimicked by adding an equivalent amount of calcium to the medium and was diminished in the presence of the calcium chelator, EGTA. To analyze the effect of BG40 and EPCs in vivo, a 6-mm diameter critical size calvarial defect was created in rats (n = 12). Controls (n = 6) received BG40 and the treatment group (n = 6) received BG40 seeded with 5×105 rat EPCs. Vascularization after 1 week was significantly improved when EPCs were seeded onto BG40, compared to implanting BG40 alone. This indicates that Ca2+ release improves EPC differentiation and is useful for enhanced early vascularization in critical size bone defects.
Collapse
Affiliation(s)
- Karam Eldesoqi
- Department of Trauma-, Hand- and Reconstructive Surgery, Hospital of the Goethe- University, Frankfurt/Main, Germany
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Li M, Fuchs S, Böse T, Schmidt H, Hofmann A, Tonak M, Unger R, Kirkpatrick CJ. Mild heat stress enhances angiogenesis in a co-culture system consisting of primary human osteoblasts and outgrowth endothelial cells. Tissue Eng Part C Methods 2013; 20:328-39. [PMID: 23998634 DOI: 10.1089/ten.tec.2013.0087] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The repair and regeneration of large bone defects, including the formation of functional vasculature, represents a highly challenging task for tissue engineering and regenerative medicine. Recent studies have shown that vascularization and ossification can be stimulated by mild heat stress (MHS), which would offer the option to enhance the bone regeneration process by relatively simple means. However, the mechanisms of MHS-enhanced angiogenesis and osteogenesis, as well as potential risks for the treated cells are unclear. We have investigated the direct effect of MHS on angiogenesis and osteogenesis in a co-culture system of human outgrowth endothelial cells (OECs) and primary osteoblasts (pOBs), and assessed cytotoxic effects, as well as the levels of various heat shock proteins (HSPs) synthesized under these conditions. Enhanced formation of microvessel-like structures was observed in co-cultures exposed to MHS (41°C, 1 h), twice per week, over a time period of 7-14 days. As shown by real-time polymerase chain reaction (PCR), the expression of vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1), angiopoietin-2 (Ang-2), and tumor necrosis factor-alpha was up-regulated in MHS-treated co-cultures 24 h post-treatment. At the protein level, significantly elevated VEGF and Ang-1 concentrations were observed in MHS-treated co-cultures and pOB mono-cultures compared with controls, indicating paracrine effects associated with MHS-induced angiogenesis. MHS-stimulated co-cultures and OEC mono-cultures released higher levels of Ang-2 than untreated cultures. On the other hand MHS treatment of co-cultures did not result in a clear effect regarding osteogenesis. Nevertheless, real-time PCR demonstrated that MHS increased the expression of mitogen-activated protein kinase, interleukin-6, and bone morphogenetic protein 2, known as HSP-related molecules in angiogenic and osteogenic regulation pathways. In agreement with these observations, the expression of some selected HSPs also increased at both the mRNA and protein levels in MHS-treated co-cultures.
Collapse
Affiliation(s)
- Ming Li
- 1 REPAIR-Lab, Institute of Pathology, University Medical Centre of the Johannes Gutenberg University , Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Weyand B, Kasper C, Israelowitz M, Gille C, von Schroeder HP, Reimers K, Vogt PM. A differential pressure laminar flow reactor supports osteogenic differentiation and extracellular matrix formation from adipose mesenchymal stem cells in a macroporous ceramic scaffold. Biores Open Access 2013; 1:145-56. [PMID: 23515420 PMCID: PMC3559213 DOI: 10.1089/biores.2012.9901] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We present a laminar flow reactor for bone tissue engineering that was developed based on a computational fluid dynamics model. The bioreactor design permits a laminar flow field through its specific internal shape. An integrated bypass system that prevents pressure build-up through bypass openings for pressure release allows for a constant pressure environment during the changing of permeability values that are caused by cellular growth within a porous scaffold. A macroporous ceramic scaffold, composed of zirconium dioxide, was used as a test biomaterial that studies adipose stem cell behavior within a controlled three-dimensional (3D) flow and pressure environment. The topographic structure of the material provided a basis for stem cell proliferation and differentiation toward the osteogenic lineage. Dynamic culture conditions in the bioreactor supported cell viability during long-term culture and induced cell cluster formation and extra-cellular matrix deposition within the porous scaffold, though no complete closure of the pores with new-formed tissue was observed. We postulate that our system is suitable for studying fluid shear stress effects on stem cell proliferation and differentiation toward bone formation in tissue-engineered 3D constructs.
Collapse
Affiliation(s)
- Birgit Weyand
- Laboratory of Experimental Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Hannover Medical School , Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
77
|
Stoppato M, Stevens HY, Carletti E, Migliaresi C, Motta A, Guldberg RE. Effects of silk fibroin fiber incorporation on mechanical properties, endothelial cell colonization and vascularization of PDLLA scaffolds. Biomaterials 2013; 34:4573-81. [PMID: 23522374 DOI: 10.1016/j.biomaterials.2013.02.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 02/02/2013] [Indexed: 10/27/2022]
Abstract
Attainment of functional vascularization of engineered constructs is one of the fundamental challenges of tissue engineering. However, the development of an extracellular matrix in most tissues, including bone, is dependent upon the establishment of a well developed vascular supply. In this study a poly(d,l-lactic acid) (PDLLA) salt-leached sponge was modified by incorporation of silk fibroin fibers to create a multicomponent scaffold, in an effort to better support endothelial cell colonization and to promote in vivo vascularization. Scaffolds with and without silk fibroin fibers were compared for microstructure, mechanical properties, ability to maintain cell populations in vitro as well as to permit vascular ingrowth into acellular constructs in vivo. We demonstrated that adding silk fibroin fibers to a PDLLA salt-leached sponge enhanced scaffold properties and heightened its capacity to support endothelial cells in vitro and to promote vascularization in vivo. Therefore refinement of scaffold properties by inclusion of materials with beneficial attributes may promote and shape cellular responses.
Collapse
Affiliation(s)
- Matteo Stoppato
- Department of Industrial Engineering and Biotech Research Center, University of Trento, Italy
| | | | | | | | | | | |
Collapse
|
78
|
Denecke B, Horsch LD, Radtke S, Fischer JC, Horn PA, Giebel B. Human endothelial colony-forming cells expanded with an improved protocol are a useful endothelial cell source for scaffold-based tissue engineering. J Tissue Eng Regen Med 2013; 9:E84-97. [PMID: 23436759 DOI: 10.1002/term.1673] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 09/21/2012] [Accepted: 11/05/2012] [Indexed: 01/02/2023]
Abstract
One of the major challenges in tissue engineering is to supply larger three-dimensional (3D) bioengineered tissue transplants with sufficient amounts of nutrients and oxygen and to allow metabolite removal. Consequently, artificial vascularization strategies of such transplants are desired. One strategy focuses on endothelial cells capable of initiating new vessel formation, which are settled on scaffolds commonly used in tissue engineering. A bottleneck in this strategy is to obtain sufficient amounts of endothelial cells, as they can be harvested only in small quantities directly from human tissues. Thus, protocols are required to expand appropriate cells in sufficient amounts without interfering with their capability to settle on scaffold materials and to initiate vessel formation. Here, we analysed whether umbilical cord blood (CB)-derived endothelial colony-forming cells (ECFCs) fulfil these requirements. In a first set of experiments, we showed that marginally expanded ECFCs settle and survive on different scaffold biomaterials. Next, we improved ECFC culture conditions and developed a protocol for ECFC expansion compatible with 'Good Manufacturing Practice' (GMP) standards. We replaced animal sera with human platelet lysates and used a novel type of tissue-culture ware. ECFCs cultured under the new conditions revealed significantly lower apoptosis and increased proliferation rates. Simultaneously, their viability was increased. Since extensively expanded ECFCs could still settle on scaffold biomaterials and were able to form tubular structures in Matrigel assays, we conclude that these ex vivo-expanded ECFCs are a novel, very potent cell source for scaffold-based tissue engineering.
Collapse
Affiliation(s)
- Bernd Denecke
- Interdisciplinary Centre for Clinical Research Aachen (IZKF Aachen), RWTH Aachen, Germany
| | - Liska D Horsch
- Institute for Transfusion Medicine, University Hospital Essen, Germany
| | - Stefan Radtke
- Institute for Transfusion Medicine, University Hospital Essen, Germany
| | - Johannes C Fischer
- Institute for Transplantation Diagnostics and Cellular Therapeutics, University Hospital Düsseldorf, Germany
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, Germany
| |
Collapse
|
79
|
Liu Y, Teoh SH, Chong MSK, Lee ESM, Mattar CNZ, Randhawa NK, Zhang ZY, Medina RJ, Kamm RD, Fisk NM, Choolani M, Chan JKY. Vasculogenic and osteogenesis-enhancing potential of human umbilical cord blood endothelial colony-forming cells. Stem Cells 2013; 30:1911-24. [PMID: 22761003 DOI: 10.1002/stem.1164] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Umbilical cord blood-derived endothelial colony-forming cells (UCB-ECFC) show utility in neovascularization, but their contribution to osteogenesis has not been defined. Cocultures of UCB-ECFC with human fetal-mesenchymal stem cells (hfMSC) resulted in earlier induction of alkaline phosphatase (ALP) (Day 7 vs. 10) and increased mineralization (1.9×; p < .001) compared to hfMSC monocultures. This effect was mediated through soluble factors in ECFC-conditioned media, leading to 1.8-2.2× higher ALP levels and a 1.4-1.5× increase in calcium deposition (p < .01) in a dose-dependent manner. Transcriptomic and protein array studies demonstrated high basal levels of osteogenic (BMPs and TGF-βs) and angiogenic (VEGF and angiopoietins) regulators. Comparison of defined UCB and adult peripheral blood ECFC showed higher osteogenic and angiogenic gene expression in UCB-ECFC. Subcutaneous implantation of UCB-ECFC with hfMSC in immunodeficient mice resulted in the formation of chimeric human vessels, with a 2.2-fold increase in host neovascularization compared to hfMSC-only implants (p = .001). We conclude that this study shows that UCB-ECFC have potential in therapeutic angiogenesis and osteogenic applications in conjunction with MSC. We speculate that UCB-ECFC play an important role in skeletal and vascular development during perinatal development but less so in later life when expression of key osteogenesis and angiogenesis genes in ECFC is lower.
Collapse
Affiliation(s)
- Yuchun Liu
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Li H, Daculsi R, Bareille R, Bourget C, Amedee J. uPA and MMP-2 were involved in self-assembled network formation in a two dimensional co-culture model of bone marrow stromal cells and endothelial cells. J Cell Biochem 2013; 114:650-7. [DOI: 10.1002/jcb.24407] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/21/2012] [Indexed: 11/11/2022]
|
81
|
Williams C, Rauch MF, Michaud M, Robinson R, Xu H, Madri J, Lavik E. Short term interactions with long term consequences: modulation of chimeric vessels by neural progenitors. PLoS One 2012; 7:e53208. [PMID: 23300890 PMCID: PMC3531360 DOI: 10.1371/journal.pone.0053208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/27/2012] [Indexed: 12/26/2022] Open
Abstract
Vessels are a critical and necessary component of most tissues, and there has been substantial research investigating vessel formation and stabilization. Several groups have investigated coculturing endothelial cells with a second cell type to promote formation and stabilization of vessels. Some have noted that long-term vessels derived from implanted cocultures are often chimeric consisting of both host and donor cells. The questions arise as to whether the coculture cell might impact the chimeric nature of the microvessels and can modulate the density of donor cells over time. If long-term engineered microvessels are primarily of host origin, any impairment of the host's angiogenic ability has significant implications for the long-term success of the implant. If one can modulate the host versus donor response, one may be able to overcome a host's angiogenic impairment. Furthermore, if one can modulate the donor contribution, one may be able to engineer microvascular networks to deliver molecules a patient lacks systemically for long times. To investigate the impact of the cocultured cell on the host versus donor contributions of endothelial cells in engineered microvascular networks, we varied the ratio of the neural progenitors to endothelial cells in subcutaneously implanted poly(ethylene glycol)/poly-L-lysine hydrogels. We found that the coculture of neural progenitors with endothelial cells led to the formation of chimeric host-donor vessels, and the ratio of neural progenitors has a significant impact on the long term residence of donor endothelial cells in engineered microvascular networks in vivo even though the neural progenitors are only present transiently in the system. We attribute this to the short term paracrine signaling between the two cell types. This suggests that one can modulate the host versus donor contributions using short-term paracrine signaling which has broad implications for the application of engineered microvascular networks and cellular therapy more broadly.
Collapse
Affiliation(s)
- Cicely Williams
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, United States of America
| | - Millicent Ford Rauch
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, United States of America
| | - Michael Michaud
- Department of Pathology, Yale University, New Haven, Connecticut, United States of America
| | - Rebecca Robinson
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, United States of America
| | - Hao Xu
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States of America
| | - Joseph Madri
- Department of Pathology, Yale University, New Haven, Connecticut, United States of America
| | - Erin Lavik
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
82
|
Liu Y, Chan JKY, Teoh SH. Review of vascularised bone tissue-engineering strategies with a focus on co-culture systems. J Tissue Eng Regen Med 2012; 9:85-105. [DOI: 10.1002/term.1617] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/13/2012] [Accepted: 08/25/2012] [Indexed: 12/16/2022]
Affiliation(s)
- Yuchun Liu
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore 637459
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine; National University of Singapore; Singapore 119228
| | - Jerry K Y Chan
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine; National University of Singapore; Singapore 119228
- Department of Reproductive Medicine, KK Women's and Children's Hospital; Singapore 229899
- Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School; Singapore
| | - Swee-Hin Teoh
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore 637459
| |
Collapse
|
83
|
Dong Z, Li B, Zhao J, Ma Q, Bai S, Yang W, Li G, Ma G, Liu Y. Prefabrication of vascularized bone grafts using a combination of bone marrow mesenchymal stem cells and vascular bundles with β-tricalcium phosphate ceramics. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 114:S153-9. [DOI: 10.1016/j.oooo.2011.09.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/18/2011] [Accepted: 09/11/2011] [Indexed: 01/18/2023]
|
84
|
Leszczynska J, Zyzynska-Granica B, Koziak K, Ruminski S, Lewandowska-Szumiel M. Contribution of endothelial cells to human bone-derived cells expansion in coculture. Tissue Eng Part A 2012; 19:393-402. [PMID: 22924666 DOI: 10.1089/ten.tea.2011.0710] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Creating a functional vascularized bone tissue remains one of the main goals of bone tissue engineering. Recently, a growing interest in the crosstalk between endothelial cells (EC) and osteoblasts (OB), the two main players in a new bone formation, has been observed. However, only a few reports have addressed a mutual influence of OB and EC on cell proliferation. Our study focuses on this issue by investigating cocultures of human bone-derived cells (HBDC) and human umbilical vein endothelial cells (HUVEC). Three various proportions of cells have been used that is, HBDC:HUVEC 1:1, 1:4, and 4:1 and the cocultures were investigated on day 1, 4, and 7, while HUVEC and HBDC monocultures served as reference. We have detected enhanced alkaline phosphatase (ALP) activity in a direct HBDC-HUVEC coculture. This effect was not observed when cells were separated by an insert, which is consistent with other reports on various OB-EC lineages. The appearance of gap-junctions in coculture was confirmed by a positive staining for connexin 43. The number of cells of both phenotypes has been determined by flow cytometry: CD-31-positive cells have been considered EC, while CD-31-negative have been counted as OB. We have observed an over 14-fold increase in OB number after a week in the 1:4 HBDC:HUVEC coculture as compared with less than fourfold in monoculture. The increase in HBDC number in 1:1 coculture has been less pronounced and has reached the value of about sevenfold. These results correspond well with the cell proliferation rate, which has been measured by 5-bromo-2'-deoxyuridine incorporation. Moreover, at day 7 EC have been still present in the coculture, which is inconsistent with some other reports. Real-time polymerase chain reaction analysis has revealed the upregulation of ALP and collagen type I genes, but not osteocalcin gene, in all the cocultures grown without pro-osteogenic additives. Our study indicates that HUVEC significantly promote HBDC expansion and upregulate collagen I gene expression in these cells. We believe that these findings have application potency in bone tissue engineering.
Collapse
Affiliation(s)
- Joanna Leszczynska
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | | | | | | | | |
Collapse
|
85
|
Baiguera S, Ribatti D. Endothelialization approaches for viable engineered tissues. Angiogenesis 2012; 16:1-14. [PMID: 23010872 DOI: 10.1007/s10456-012-9307-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 09/15/2012] [Indexed: 12/21/2022]
Abstract
One of the main limitation in obtaining thick, 3-dimensional viable engineered constructs is the inability to provide a sufficient and functional blood vessel system essential for the in vitro survival and the in vivo integration of the construct. Different strategies have been proposed to simulate the ingrowth of new blood vessels into engineered tissue, such as the use of growth factors, fabrication scaffold technologies, in vivo prevascularization and cell-based strategies, and it has been demonstrated that endothelial cells play a central role in the neovascularization process and in the control of blood vessel function. In particular, different "environmental" settings (origin, presence of supporting cells, biomaterial surface, presence of hemodynamic forces) strongly influence endothelial cell function, angiogenic potential and the in vivo formation of durable vessels. This review provides an overview of the different techniques developed so far for the vascularization of tissue-engineered constructs (with their advantages and pitfalls), focusing the attention on the recent development in the cell-based vascularization strategy and the in vivo applications.
Collapse
Affiliation(s)
- Silvia Baiguera
- BIOAIRLab, European Center for Thoracic Surgery, University Hospital Careggi, Florence, Italy.
| | | |
Collapse
|
86
|
Kuttenberger J, Polska E, Schaefer BM. A novel three-dimensional bone chip organ culture. Clin Oral Investig 2012; 17:1547-55. [DOI: 10.1007/s00784-012-0833-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 08/27/2012] [Indexed: 10/27/2022]
|
87
|
Vo TN, Kasper FK, Mikos AG. Strategies for controlled delivery of growth factors and cells for bone regeneration. Adv Drug Deliv Rev 2012; 64:1292-309. [PMID: 22342771 PMCID: PMC3358582 DOI: 10.1016/j.addr.2012.01.016] [Citation(s) in RCA: 436] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 01/23/2012] [Accepted: 01/30/2012] [Indexed: 12/15/2022]
Abstract
The controlled delivery of growth factors and cells within biomaterial carriers can enhance and accelerate functional bone formation. The carrier system can be designed with pre-programmed release kinetics to deliver bioactive molecules in a localized, spatiotemporal manner most similar to the natural wound healing process. The carrier can also act as an extracellular matrix-mimicking substrate for promoting osteoprogenitor cellular infiltration and proliferation for integrative tissue repair. This review discusses the role of various regenerative factors involved in bone healing and their appropriate combinations with different delivery systems for augmenting bone regeneration. The general requirements of protein, cell and gene therapy are described, with elaboration on how the selection of materials, configurations and processing affects growth factor and cell delivery and regenerative efficacy in both in vitro and in vivo applications for bone tissue engineering.
Collapse
Affiliation(s)
- Tiffany N. Vo
- Department of Bioengineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
| | - F. Kurtis Kasper
- Department of Bioengineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
| | - Antonios G. Mikos
- Department of Bioengineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
- Department of Chemical and Biomolecular Engineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
| |
Collapse
|
88
|
Mendes LF, Pirraco RP, Szymczyk W, Frias AM, Santos TC, Reis RL, Marques AP. Perivascular-like cells contribute to the stability of the vascular network of osteogenic tissue formed from cell sheet-based constructs. PLoS One 2012; 7:e41051. [PMID: 22829909 PMCID: PMC3400580 DOI: 10.1371/journal.pone.0041051] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/16/2012] [Indexed: 12/11/2022] Open
Abstract
In recent years several studies have been supporting the existence of a close relationship in terms of function and progeny between Mesenchymal Stem Cells (MSCs) and Pericytes. This concept has opened new perspectives for the application of MSCs in Tissue Engineering (TE), with special interest for the pre-vascularization of cell dense constructs. In this work, cell sheet technology was used to create a scaffold-free construct composed of osteogenic, endothelial and perivascular-like (CD146+) cells for improved in vivo vessel formation, maturation and stability. The CD146 pericyte-associated phenotype was induced from human bone marrow mesenchymal stem cells (hBMSCs) by the supplementation of standard culture medium with TGF-β1. Co-cultured cell sheets were obtained by culturing perivascular-like (CD146+) cells and human umbilical vein endothelial cells (HUVECs) on an hBMSCs monolayer maintained in osteogenic medium for 7 days. The perivascular-like (CD146+) cells and the HUVECs migrated and organized over the collagen-rich osteogenic cell sheet, suggesting the existence of cross-talk involving the co-cultured cell types. Furthermore the presence of that particular ECM produced by the osteoblastic cells was shown to be the key regulator for the singular observed organization. The osteogenic and angiogenic character of the proposed constructs was assessed in vivo. Immunohistochemistry analysis of the explants revealed the integration of HUVECs with the host vasculature as well as the osteogenic potential of the created construct, by the expression of osteocalcin. Additionally, the analysis of the diameter of human CD146 positive blood vessels showed a higher mean vessel diameter for the co-cultured cell sheet condition, reinforcing the advantage of the proposed model regarding blood vessels maturation and stability and for the in vitro pre-vascularization of TE constructs.
Collapse
Affiliation(s)
- Luís F. Mendes
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Wojciech Szymczyk
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Ana M. Frias
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Tírcia C. Santos
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Alexandra P. Marques
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
- * E-mail:
| |
Collapse
|
89
|
Traphagen SB, Titushkin I, Sun S, Wary KK, Cho M. Endothelial invasive response in a co-culture model with physically-induced osteodifferentiation. J Tissue Eng Regen Med 2012; 7:621-30. [PMID: 22696416 DOI: 10.1002/term.554] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 08/26/2011] [Accepted: 11/15/2011] [Indexed: 02/06/2023]
Abstract
Manipulation of stem cells using physicochemical stimuli has emerged as an important tool in regenerative medicine. While 2D substrates with tunable elasticity have been studied for control of stem cell differentiation, we recently developed a stratified co-culture model of angiogenesis of human mesenchymal stem cells (hMSCs) that differentiate on a tunable polydimethylsiloxane (PDMS) substrate, thereby creating a physiologic context for elasticity-induced differentiation. Endothelial cells (EC) were cultured on top of the hMSC construct on a collagen gel to monitor network formation. Media composition influenced EC invasion due to the conditioning media, the reduction of serum and supplemental growth factors, and the addition of recombinant growth factors. Conditioned media, recombinant growth factors and direct co-culture were compared for endothelial cell invasive response using quantitative image analysis. As anticipated, use of recombinant vascular endothelial growth factor (VEGF) induced the deepest EC invasions while direct co-culture caused shallow invasions compared to other conditions. However, endothelial cells displayed lumen-like morphology, suggesting that cell-cell interaction in the co-culture model could mimic sprouting behaviour. In summary, an engineered suitable biochemical and physical environment facilitated endothelial cells to form 3D vessel structures onto hMSCs. These structures were plated on a stiff surface known to induce osteodifferentiation of stem cells. This low cost co-culture system, with its minimal chemical supplementation and physically controllable matrix, could potentially model in vivo potential in engineered and pre-vascularized bone grafts.
Collapse
|
90
|
Cornejo A, Sahar DE, Stephenson SM, Chang S, Nguyen S, Guda T, Wenke JC, Vasquez A, Michalek JE, Sharma R, Krishnegowda NK, Wang HT. Effect of adipose tissue-derived osteogenic and endothelial cells on bone allograft osteogenesis and vascularization in critical-sized calvarial defects. Tissue Eng Part A 2012; 18:1552-61. [PMID: 22440012 DOI: 10.1089/ten.tea.2011.0515] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The use of processed bone allograft to repair large osseous defects of the skull has been limited, given that it lacks the osteogenic cellularity and intrinsic vascular supply which are essential elements for successful graft healing and, at the same time, the areas to be targeted through tissue-engineering applications. In this study, we investigated the effect of predifferentiated rat adipose tissue-derived osteoblastic cells (OBs) and endothelial cells (ECs) on calvarial bone allograft healing and vascularization using an orthotopic critical-sized calvarial defect model. For this purpose, thirty-seven 8 mm critical calvarial defects in Lewis rats were treated with bone allografts seeded with no cells, undifferentiated adipose tissue-derived stem cells (ASC), OBs, ECs, and OBs and ECs simultaneously. After 8 weeks, the bone volume and mineral density were calculated using microcomputed tomography and the microvessel formation using immunohistochemical staining and imaging software. The amount of bone within the 8 mm defect was significantly higher for the allografts treated with ECs compared with the allografts treated with OBs (p=0.05) and simultaneously with the two cell lineages (p=0.02). There were no significant differences in bone formation between the latter two groups and the control groups (allografts treated with no cells and undifferentiated ASC). There were no significant differences in bone mineral density among the groups. The amount of microvessels was significantly higher in the group treated with ECs relative to all groups (p=< 0.05). Our results show that the implantation of ASC-derived ECs improves the vascularization of calvarial bone allografts at 8 weeks after treatment. This cell-based vascularization strategy can be used to improve the paucity of perfusion in allogenic bone implants. However, in this study, the treatment of allografts with OBs alone or in combination with ECs did not support bone formation or vascularization.
Collapse
Affiliation(s)
- Agustin Cornejo
- Division of Plastic and Reconstructive Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Effect of Endothelial Differentiated Adipose-Derived Stem Cells on Vascularity and Osteogenesis in Poly(D,L-Lactide) Scaffolds In Vivo. J Craniofac Surg 2012; 23:913-8. [DOI: 10.1097/scs.0b013e31824e5cd8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
92
|
Nahles S, Nack C, Gratecap K, Lage H, Nelson JJ, Nelson K. Bone physiology in human grafted and non-grafted extraction sockets - an immunohistochemical study. Clin Oral Implants Res 2012; 24:812-9. [DOI: 10.1111/j.1600-0501.2012.02462.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2012] [Indexed: 01/08/2023]
Affiliation(s)
- Susanne Nahles
- Oral- and Maxillofacial Surgery; Charité Campus Virchow; Berlin; Germany
| | - Claudia Nack
- Oral- and Maxillofacial Surgery; Charité Campus Virchow; Berlin; Germany
| | - Kerrin Gratecap
- Oral- and Maxillofacial Surgery; Charité Campus Virchow; Berlin; Germany
| | - Hermann Lage
- Department of Pathology; Campus Mitte, Charite; Berlin; Germany
| | - John J. Nelson
- Department of Pathology; University of South Alabama; Mobile; Alabama; USA
| | - Katja Nelson
- Department of CMF-Surgery; University of Freiburg; Berlin; Germany
| |
Collapse
|
93
|
Fuchs S, Dohle E, Kirkpatrick CJ. Sonic Hedgehog-mediated synergistic effects guiding angiogenesis and osteogenesis. VITAMINS AND HORMONES 2012; 88:491-506. [PMID: 22391318 DOI: 10.1016/b978-0-12-394622-5.00022-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sonic hedgehog (Shh) is a morphogen controlling the skeletal and vascular development in the embryo but is also reactivated during adult repair processes. Thus, this molecule holds great therapeutic potential for biotechnological and biomedical approaches aiming to enhance tissue regeneration or to replace damaged tissues. According to present knowledge, Shh signaling controls the expression of several families of growth factors involved in neovascularization and vessel maturation and acts upstream of the most prominent angiogenic growth factor, vascular endothelial growth factor. In this context, a very interesting feature of Shh is that it controls both angiogenic activity and vessel stabilization by mural cells. In parallel, Shh seems to have a direct effect on endothelial cell tube formation and seems to trigger the differentiation process of mesenchymal stem cells toward the osteogenic lineage. In this chapter, we will therefore shortly summarize the multifaceted potential of Shh for bone repair and vascularization according to the current literature. In addition, we will show how coculture models based on outgrowth endothelial cells and primary osteoblasts can be used to reveal some of the relevant mechanisms by which Shh drives and connects bone regeneration and vascularization.
Collapse
Affiliation(s)
- Sabine Fuchs
- Institute of Pathology, Johannes Gutenberg University, Mainz, Germany
| | | | | |
Collapse
|
94
|
Fu X, Wang H. Rapid fabrication of biomimetic nanofiber-enabled skin grafts. Nanomedicine (Lond) 2012. [DOI: 10.1533/9780857096449.3.428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
95
|
Phenotypic and proliferative modulation of human mesenchymal stem cells via crosstalk with endothelial cells. Stem Cell Res 2011; 7:186-97. [DOI: 10.1016/j.scr.2011.05.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 05/26/2011] [Accepted: 05/31/2011] [Indexed: 12/14/2022] Open
|
96
|
Costa-Pinto AR, Reis RL, Neves NM. Scaffolds based bone tissue engineering: the role of chitosan. TISSUE ENGINEERING PART B-REVIEWS 2011; 17:331-47. [PMID: 21810029 DOI: 10.1089/ten.teb.2010.0704] [Citation(s) in RCA: 207] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
As life expectancy increases, malfunction or loss of tissue caused by injury or disease leads to reduced quality of life in many patients at significant socioeconomic cost. Even though major progress has been made in the field of bone tissue engineering, present therapies, such as bone grafts, still have limitations. Current research on biodegradable polymers is emerging, combining these structures with osteogenic cells, as an alternative to autologous bone grafts. Different types of biodegradable materials have been proposed for the preparation of three-dimensional porous scaffolds for bone tissue engineering. Among them, natural polymers are one of the most attractive options, mainly due to their similarities with extracellular matrix, chemical versatility, good biological performance, and inherent cellular interactions. In this review, special attention is given to chitosan as a biomaterial for bone tissue engineering applications. An extensive literature survey was performed on the preparation of chitosan scaffolds and their in vitro biological performance as well as their potential to facilitate in vivo bone regeneration. The present review also aims to offer the reader a general overview of all components needed to engineer new bone tissue. It gives a brief background on bone biology, followed by an explanation of all components in bone tissue engineering, as well as describing different tissue engineering strategies. Moreover, also discussed are the typical models used to evaluate in vitro functionality of a tissue-engineered construct and in vivo models to assess the potential to regenerate bone tissue are discussed.
Collapse
Affiliation(s)
- Ana Rita Costa-Pinto
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine University of Minho, Guimarães, Portugal
| | | | | |
Collapse
|
97
|
Kolbe M, Xiang Z, Dohle E, Tonak M, Kirkpatrick CJ, Fuchs S. Paracrine effects influenced by cell culture medium and consequences on microvessel-like structures in cocultures of mesenchymal stem cells and outgrowth endothelial cells. Tissue Eng Part A 2011; 17:2199-212. [PMID: 21529248 DOI: 10.1089/ten.tea.2010.0474] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSC) from bone marrow and outgrowth endothelial cells (OEC) from peripheral blood are considered as attractive cell types for applications in regenerative medicine aiming to build up complex vascularized tissue-engineered constructs. MSC provide several advantages such as the potential to differentiate to osteoblasts and to support the neovascularization process by release of proangiogenic factors. On the other hand, the neovascularization process can be actively supported by OEC forming perfused vascular structures after co-implantation with other cell types. In this study the formation of angiogenic structures in vitro was investigated in cocultures of MSC and OEC, cultured either in the medium for osteogenic differentiation of MSC (ODM) or in the medium for OEC cultivation endothelial cell growth medium-2 (EGM2 Bullet Kit). After 2 weeks, cocultures in EGM2 formed more microvessel-like structures compared to cocultures in ODM as demonstrated by immunofluorescence staining for the endothelial marker CD31. Increased expression of CD31 and CD146 in quantitative real-time polymerase chain reaction as well as a higher percentage of CD31- and CD146-positive cells in flow cytometry indicated a beneficial influence of EGM2 on endothelial cell growth and function. In addition, the improved formation of vascular structures in EGM2 correlates with higher levels of the proangiogenic factor vascular endothelial growth factor and platelet-derived growth factor in the supernatant of cocultures as well as in monocultures of MSC when cultivated in EGM-2. Nevertheless, ODM was more suitable for the differentiation of MSC to osteoblastic lineages in the cocultures, whereas EGM2 favored factors involved in vessel stabilization by pericytes. In conclusion, this study highlights the importance of medium components for cell interaction triggering the formation of angiogenic structures.
Collapse
Affiliation(s)
- Marlen Kolbe
- REPAIR-Lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
98
|
Kirkpatrick CJ, Fuchs S, Unger RE. Co-culture systems for vascularization--learning from nature. Adv Drug Deliv Rev 2011; 63:291-9. [PMID: 21281686 DOI: 10.1016/j.addr.2011.01.009] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 01/11/2011] [Accepted: 01/19/2011] [Indexed: 12/24/2022]
Abstract
The endothelial cell (EC) is practically ubiquitous in the human body and forms the inner cellular lining of the entire cardiovascular system. Following tissue injury, the microcirculation becomes the stage for both the inflammatory response and the subsequent healing reaction to restore physiological function to the damaged tissue. The advent of the multidisciplinary field of Regenerative Medicine (RegMed), of which Tissue Engineering (TE) and drug delivery using modern stimuli-responsive or interactive biomaterials are important components, has opened up new approaches to the acceleration of the healing response. A central and rate-limiting role in the latter is played by the process of vascularization or neovascularization, so that it is not surprising that in RegMed concepts have been developed for the drug- and gene-delivery of potent stimuli such as vascular-endothelial growth factor (VEGF) to promote neovessel development. However, not all of these novel materials can be tested in vivo, and in vitro co-culture model systems using human primary cells are being developed to pre-evaluate and determine which of the RegMed concepts exhibit the most promising potential for success after implantation. This review describes some of the growing number of in vitro co-cultures model systems that are being used to study cell-cell and cell-material interactions at the cellular and molecular levels to determine which materials are best suited to integrate into the host, promote a rapid vascularization and fit into the regenerative process without disturbing or slowing the normal healing steps.
Collapse
|
99
|
Kolbe M, Dohle E, Katerla D, Kirkpatrick CJ, Fuchs S. Enrichment of outgrowth endothelial cells in high and low colony-forming cultures from peripheral blood progenitors. Tissue Eng Part C Methods 2011; 16:877-86. [PMID: 19891540 DOI: 10.1089/ten.tec.2009.0492] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
An effective isolation protocol for outgrowth endothelial cells (OEC) resulting in higher cell numbers and a reduced expansion time would facilitate the therapeutical application. In this study a standard protocol based on the isolation of mononuclear cells from adult peripheral blood was modified by adding a passaging step 7 days after the isolation. OEC colonies gained by both protocols were evaluated after 28 days and resulted in different frequencies of OEC colonies depending on the donor and culture protocol. Accordingly, we defined two groups, namely, high colony-forming cultures (HCC) and low colony-forming cultures (LCC) for further analysis. LCC revealed no increase in OEC colonies by the modified protocol, whereas in HCC the frequency of OEC colonies was significantly improved by the passaging step. Quantitative real-time polymerase chain reaction, flow cytometry, and immunofluorescence for endothelial markers indicated an enrichment of OEC by protocol modification in HCC. In addition, HCC revealed higher expression of CD34 and CD133 compared to LCC and resulted in higher numbers of OEC gained per donor, which was further improved by the modified protocol. We conclude that the modified protocol supports the selection of OEC from adult peripheral blood with a high clonogenic potential and results in a better efficacy in OEC isolation.
Collapse
Affiliation(s)
- Marlen Kolbe
- Institute of Pathology, Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | |
Collapse
|
100
|
Li H, Daculsi R, Grellier M, Bareille R, Bourget C, Remy M, Amedee J. The role of vascular actors in two dimensional dialogue of human bone marrow stromal cell and endothelial cell for inducing self-assembled network. PLoS One 2011; 6:e16767. [PMID: 21304816 PMCID: PMC3033416 DOI: 10.1371/journal.pone.0016767] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 01/14/2011] [Indexed: 11/25/2022] Open
Abstract
Angiogenesis is very important for vascularized tissue engineering. In this study, we found that a two-dimensional co-culture of human bone marrow stromal cell (HBMSC) and human umbical vein endothelial cell (HUVEC) is able to stimulate the migration of co-cultured HUVEC and induce self-assembled network formation. During this process, expression of vascular endothelial growth factor (VEGF165) was upregulated in co-cultured HBMSC. Meanwhile, VEGF165-receptor2 (KDR) and urokinase-type plasminogen activator (uPA) were upregulated in co-cultured HUVEC. Functional studies show that neutralization of VEGF165 blocked the migration and the rearrangement of the cells and downregulated the expression of uPA and its receptor. Blocking of vascular endothelial-cadherin (VE-cad) did not affect the migration of co-cultured HUVEC but suppressed the self-assembled network formation. In conclusion, co-cultures upregulated the expression of VEGF165 in co-cultured HBMSC; VEGF165 then activated uPA in co-cultured HUVEC, which might be responsible for initiating the migration and the self-assembled network formation with the participation of VE-cad. All of these results indicated that only the direct contact of HBMSC and HUVEC and their respective dialogue are sufficient to stimulate secretion of soluble factors and to activate molecules that are critical for self-assembled network formation which show a great application potential for vascularization in tissue engineering.
Collapse
Affiliation(s)
- Haiyan Li
- INSERM U577, Bordeaux and University Victor Segalen Bordeaux 2, Bordeaux, France.
| | | | | | | | | | | | | |
Collapse
|