1
|
Hao M, Xue L, Wen X, Sun L, Zhang L, Xing K, Hu X, Xu J, Xing D. Advancing bone regeneration: Unveiling the potential of 3D cell models in the evaluation of bone regenerative materials. Acta Biomater 2024; 183:1-29. [PMID: 38815683 DOI: 10.1016/j.actbio.2024.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Bone, a rigid yet regenerative tissue, has garnered extensive attention for its impressive healing abilities. Despite advancements in understanding bone repair and creating treatments for bone injuries, handling nonunions and large defects remains a major challenge in orthopedics. The rise of bone regenerative materials is transforming the approach to bone repair, offering innovative solutions for nonunions and significant defects, and thus reshaping orthopedic care. Evaluating these materials effectively is key to advancing bone tissue regeneration, especially in difficult healing scenarios, making it a critical research area. Traditional evaluation methods, including two-dimensional cell models and animal models, have limitations in predicting accurately. This has led to exploring alternative methods, like 3D cell models, which provide fresh perspectives for assessing bone materials' regenerative potential. This paper discusses various techniques for constructing 3D cell models, their pros and cons, and crucial factors to consider when using these models to evaluate bone regenerative materials. We also highlight the significance of 3D cell models in the in vitro assessments of these materials, discuss their current drawbacks and limitations, and suggest future research directions. STATEMENT OF SIGNIFICANCE: This work addresses the challenge of evaluating bone regenerative materials (BRMs) crucial for bone tissue engineering. It explores the emerging role of 3D cell models as superior alternatives to traditional methods for assessing these materials. By dissecting the construction, key factors of evaluating, advantages, limitations, and practical considerations of 3D cell models, the paper elucidates their significance in overcoming current evaluation method shortcomings. It highlights how these models offer a more physiologically relevant and ethically preferable platform for the precise assessment of BRMs. This contribution is particularly significant for "Acta Biomaterialia" readership, as it not only synthesizes current knowledge but also propels the discourse forward in the search for advanced solutions in bone tissue engineering and regeneration.
Collapse
Affiliation(s)
- Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M139PL, UK
| | - Xiaokun Hu
- Department of Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao 26600, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
2
|
Fois MG, van Griensven M, Giselbrecht S, Habibović P, Truckenmüller RK, Tahmasebi Birgani ZN. Mini-bones: miniaturized bone in vitro models. Trends Biotechnol 2024; 42:910-928. [PMID: 38493050 DOI: 10.1016/j.tibtech.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 03/18/2024]
Abstract
In bone tissue engineering (TE) and regeneration, miniaturized, (sub)millimeter-sized bone models have become a popular trend since they bring about physiological biomimicry, precise orchestration of concurrent stimuli, and compatibility with high-throughput setups and high-content imaging. They also allow efficient use of cells, reagents, materials, and energy. In this review, we describe the state of the art of miniaturized in vitro bone models, or 'mini-bones', describing these models based on their characteristics of (multi)cellularity and engineered extracellular matrix (ECM), and elaborating on miniaturization approaches and fabrication techniques. We analyze the performance of 'mini-bone' models according to their applications for studying basic bone biology or as regeneration models, disease models, and screening platforms, and provide an outlook on future trends, challenges, and opportunities.
Collapse
Affiliation(s)
- Maria Gabriella Fois
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Martijn van Griensven
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Roman K Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| | - Zeinab Niloofar Tahmasebi Birgani
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| |
Collapse
|
3
|
Son Y, Paton CM. A Review of free fatty acid-induced cell signaling, angiopoietin-like protein 4, and skeletal muscle differentiation. Front Physiol 2022; 13:987977. [PMID: 36148297 PMCID: PMC9485487 DOI: 10.3389/fphys.2022.987977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Postnatal skeletal muscle differentiation from quiescent satellite cells is a highly regulated process, although our understanding of the contribution of nutritional factors in myogenesis is limited. Free fatty acids (FFAs) are known to cause detrimental effects to differentiated skeletal muscle cells by increasing oxidative stress which leads to muscle wasting and insulin resistance in skeletal muscle. In addition, FFAs are thought to act as inhibitors of skeletal muscle differentiation. However, the precise molecular mechanisms underlying the effects of FFAs on skeletal muscle differentiation remains to be elucidated. There is a clear relationship between dietary FFAs and their ability to suppress myogenesis and we propose the hypothesis that the FFA-mediated increase in angiopoietin-like protein 4 (ANGPTL4) may play a role in the inhibition of differentiation. This review discusses the role of FFAs in skeletal muscle differentiation to-date and proposes potential mechanisms of FFA-induced ANGPTL4 mediated inhibition of skeletal muscle differentiation.
Collapse
Affiliation(s)
- Yura Son
- Department Nutritional Sciences, Athens, GA, United States
| | - Chad M. Paton
- Department Nutritional Sciences, Athens, GA, United States
- Department of Food Science and Technology, University of Georgia, Athens, GA, United States
- *Correspondence: Chad M. Paton,
| |
Collapse
|
4
|
A hierarchical vascularized engineered bone inspired by intramembranous ossification for mandibular regeneration. Int J Oral Sci 2022; 14:31. [PMID: 35732648 PMCID: PMC9217949 DOI: 10.1038/s41368-022-00179-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/22/2022] Open
Abstract
Mandibular defects caused by injuries, tumors, and infections are common and can severely affect mandibular function and the patient’s appearance. However, mandible reconstruction with a mandibular bionic structure remains challenging. Inspired by the process of intramembranous ossification in mandibular development, a hierarchical vascularized engineered bone consisting of angiogenesis and osteogenesis modules has been produced. Moreover, the hierarchical vascular network and bone structure generated by these hierarchical vascularized engineered bone modules match the particular anatomical structure of the mandible. The ultra-tough polyion complex has been used as the basic scaffold for hierarchical vascularized engineered bone for ensuring better reconstruction of mandible function. According to the results of in vivo experiments, the bone regenerated using hierarchical vascularized engineered bone is similar to the natural mandibular bone in terms of morphology and genomics. The sonic hedgehog signaling pathway is specifically activated in hierarchical vascularized engineered bone, indicating that the new bone in hierarchical vascularized engineered bone underwent a process of intramembranous ossification identical to that of mandible development. Thus, hierarchical vascularized engineered bone has a high potential for clinical application in mandibular defect reconstruction. Moreover, the concept based on developmental processes and bionic structures provides an effective strategy for tissue regeneration.
Collapse
|
5
|
Tsuji-Tamura K, Tamura M. Basic fibroblast growth factor uniquely stimulates quiescent vascular smooth muscle cells and induces proliferation and dedifferentiation. FEBS Lett 2022; 596:1686-1699. [PMID: 35363891 DOI: 10.1002/1873-3468.14345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 11/11/2022]
Abstract
Blood vessels normally remain stable over the long-term. However, in atherosclerosis, vascular cells leave the quiescent state and enter an activated state. Here, we investigated the factors that trigger breakage of the quiescent state by screening growth factors and cytokines using a vascular smooth muscle cell (SMC) line and an endothelial cell (EC) line. Despite known functions of the tested factors, only basic fibroblast growth factor (bFGF) was identified as a potent trigger of quiescence breakage in SMCs, but not ECs. bFGF disrupted tight SMC-monolayers, and caused morphological changes, proliferation and dedifferentiation. Human primary SMCs, but not ECs, also showed similar results. Aberrant SMC-proliferation is a critical histological event in atherosclerosis. We thus provide further insights into the role of bFGF in vascular pathobiology.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo, 060-8586, Japan
| | - Masato Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-ku, Sapporo, 060-8586, Japan
| |
Collapse
|
6
|
Vakhrushev IV, Nezhurina EK, Karalkin PA, Tsvetkova AV, Sergeeva NS, Majouga AG, Yarygin KN. Heterotypic Multicellular Spheroids as Experimental and Preclinical Models of Sprouting Angiogenesis. BIOLOGY 2021; 11:18. [PMID: 35053016 PMCID: PMC8772844 DOI: 10.3390/biology11010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
Sprouting angiogenesis is the common response of live tissues to physiological and pathological angiogenic stimuli. Its accurate evaluation is of utmost importance for basic research and practical medicine and pharmacology and requires adequate experimental models. A variety of assays for angiogenesis were developed, none of them perfect. In vitro approaches are generally less physiologically relevant due to the omission of essential components regulating the process. However, only in vitro models can be entirely non-xenogeneic. The limitations of the in vitro angiogenesis assays can be partially overcome using 3D models mimicking tissue O2 and nutrient gradients, the influence of the extracellular matrix (ECM), and enabling cell-cell interactions. Here we present a review of the existing models of sprouting angiogenesis that are based on the use of endothelial cells (ECs) co-cultured with perivascular or other stromal cells. This approach provides an excellent in vitro platform for further decoding of the cellular and molecular mechanisms of sprouting angiogenesis under conditions close to the in vivo conditions, as well as for preclinical drug testing and preclinical research in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Igor V. Vakhrushev
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | - Elizaveta K. Nezhurina
- P.A. Hertsen Moscow Oncology Research Center, National Medical Research Radiological Center, 125284 Moscow, Russia;
| | - Pavel A. Karalkin
- Institute for Cluster Oncology, Sechenov University, 119435 Moscow, Russia;
| | | | - Nataliya S. Sergeeva
- Department of Biology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Alexander G. Majouga
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| |
Collapse
|
7
|
Burdis R, Kelly DJ. Biofabrication and bioprinting using cellular aggregates, microtissues and organoids for the engineering of musculoskeletal tissues. Acta Biomater 2021; 126:1-14. [PMID: 33711529 DOI: 10.1016/j.actbio.2021.03.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022]
Abstract
The modest clinical impact of musculoskeletal tissue engineering (TE) can be attributed, at least in part, to a failure to recapitulate the structure, composition and functional properties of the target tissue. This has motivated increased interest in developmentally inspired TE strategies, which seek to recapitulate key events that occur during embryonic and post-natal development, as a means of generating truly biomimetic grafts to replace or regenerate damaged tissues and organs. Such TE strategies can be substantially enabled by emerging biofabrication and bioprinting strategies, and in particular the use of cellular aggregates, microtissues and organoids as 'building blocks' for the development of larger tissues and/or organ precursors. Here, the application of such biological building blocks for the engineering of musculoskeletal tissues, from vascularised bone to zonally organised articular cartilage, will be reviewed. The importance of first scaling-down to later scale-up will be discussed, as this is viewed as a key component of engineering functional grafts using cellular aggregates or microtissues. In the context of engineering anatomically accurate tissues of scale suitable for tissue engineering and regenerative medicine applications, novel bioprinting modalities and their application in controlling the process by which cellular aggregates or microtissues fuse and self-organise will be reviewed. Throughout the paper, we will highlight some of the key challenges facing this emerging field. STATEMENT OF SIGNIFICANCE: The field of bioprinting has grown substantially in recent years, but despite the hype and excitement it has generated, there are relatively few examples of bioprinting strategies producing implants with superior regenerative potential to that achievable with more traditional tissue engineering approaches. This paper provides an up-to-date review of emerging biofabrication and bioprinting strategies which use cellular aggregates and microtissues as 'building blocks' for the development of larger musculoskeletal tissues and/or organ precursors - a field of research that can potentially enable functional regeneration of damaged and diseased tissues. The application of cellular aggregates and microtissues for the engineering of musculoskeletal tissues, from vascularised bone to zonally organised articular cartilage, will be reviewed. In the context of engineering anatomically accurate tissues of scale, novel bioprinting modalities and their application in controlling the process by which cellular aggregates or microtissues self-organise is addressed, as well as key challenges facing this emerging field.
Collapse
|
8
|
Coyle RC, Barrs RW, Richards DJ, Ladd EP, Menick DR, Mei Y. Targeting HIF-α for robust prevascularization of human cardiac organoids. J Tissue Eng Regen Med 2020; 15:189-202. [PMID: 33868541 DOI: 10.1002/term.3165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prevascularized 3D microtissues have been shown to be an effective cell delivery vehicle for cardiac repair. To this end, our lab has explored the development of self-organizing, prevascularized human cardiac organoids by co-seeding human cardiomyocytes with cardiac fibroblasts, endothelial cells, and stromal cells into agarose microwells. We hypothesized that this prevascularization process is facilitated by the endogenous upregulation of hypoxia-inducible factor (HIF) pathway in the avascular 3D microtissues. In this study, we used Molidustat, a selective PHD (prolyl hydroxylase domain enzymes) inhibitor that stabilizes HIF-α, to treat human cardiac organoids, which resulted in 150 ± 61% improvement in endothelial expression (CD31) and 220 ± 20% improvement in the number of lumens per organoids. We hypothesized that the improved endothelial expression seen in Molidustat treated human cardiac organoids was dependent upon upregulation of VEGF, a well-known downstream target of HIF pathway. Through the use of immunofluorescent staining and ELISA assays, we determined that Molidustat treatment improved VEGF expression of non-endothelial cells and resulted in improved co-localization of supporting cell types and endothelial structures. We further demonstrated that Molidustat treated human cardiac organoids maintain cardiac functionality. Lastly, we showed that Molidustat treatment improves survival of cardiac organoids when exposed to both hypoxic and ischemic conditions in vitro. For the first time, we demonstrate that targeted HIF-α stabilization provides a robust strategy to improve endothelial expression and lumen formation in cardiac microtissues, which will provide a powerful framework for prevascularization of various microtissues in developing successful cell transplantation therapies.
Collapse
Affiliation(s)
- Robert C Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ryan W Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan J Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Emma P Ladd
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Donald R Menick
- Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA.,Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA.,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
9
|
Huang S, Kong A, Cao Q, Tong Z, Wang X. The role of blood vessels in broiler chickens with tibial dyschondroplasia. Poult Sci 2020; 98:6527-6532. [PMID: 31433842 PMCID: PMC8913930 DOI: 10.3382/ps/pez497] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
Tibial dyschondroplasia (TD) is an intractable tibiotarsal bone disorder of rapid growing avian species, which leads to huge economic losses and compromised poultry welfare. However, the exact pathogenesis and treatment of TD remain largely unknown. Based on continuous research findings, we propose the TD pathogenesis hypothesis: during skeletal development of TD chickens, due to the absence of vasculature of proximal tibial growth plates (TGP), hypertrophic chondrocytes of the TGP are unable to complete calcification in normal bone development and less dead chondrocytes in the corresponding area can be timely transported through the blood vessels. Moreover, recent studies demonstrate that the TD formation mechanism gradually tends to a large number of dead chondrocytes in the TGP region or apoptosis occur due to various factors (such as, reduction of vascular invasion and blood cells, and increased weight or mechanical force of the tibia), while the reduction of blood vessels is insufficient to remove these chondrocytes and eventually leads to the TD formation. Recognizing the possible role of the blood vessels in the incidence of TD and can propose that the improvement in vasculature might be a novel therapeutic approach for ending TD in chickens.
Collapse
Affiliation(s)
- Shucheng Huang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Anan Kong
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Qinqin Cao
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Zongxi Tong
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Xuebing Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Engineering functional organs starting from stem or progenitor cells holds promise to address the urgent need for organ transplants. However, to date, the development of complex organ structures remains an open challenge. RECENT FINDINGS Among multiple approaches to organ regeneration that are being investigated, two main directions can be identified, namely the patterned deposition of cells to impose specific structures, using bioprinting technologies, and (ii) the spontaneous development of organoids, according to principles of self-organization. In this review, we shortly describe the advantages and limitations of these paradigms and we discuss how they can synergize their positive features to better control and robustly develop organs from stem cells, toward organogenesis by design. SUMMARY The outlined possibilities to bring together tools and concepts of bioprinting and self-organization will be relevant not only to generate implantable organs, but also to dissect fundamental mechanisms of organogenesis and to test therapeutic strategies in modeled pathological settings.
Collapse
|
11
|
Munarin F, Kant RJ, Rupert CE, Khoo A, Coulombe KLK. Engineered human myocardium with local release of angiogenic proteins improves vascularization and cardiac function in injured rat hearts. Biomaterials 2020; 251:120033. [PMID: 32388033 PMCID: PMC8115013 DOI: 10.1016/j.biomaterials.2020.120033] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/27/2022]
Abstract
Heart regeneration after myocardial infarction requires new cardiomyocytes and a supportive vascular network. Here, we evaluate the efficacy of localized delivery of angiogenic factors from biomaterials within the implanted muscle tissue to guide growth of a more dense, organized, and perfused vascular supply into implanted engineered human cardiac tissue on an ischemia/reperfusion injured rat heart. We use large, aligned 3-dimensional engineered tissue with cardiomyocytes derived from human induced pluripotent stem cells in a collagen matrix that contains dispersed alginate microspheres as local protein depots. Release of angiogenic growth factors VEGF and bFGF in combination with morphogen sonic hedgehog from the microspheres into the local microenvironment occurs from the epicardial implant site. Analysis of the 3D vascular network in the engineered tissue via Microfil® perfusion and microCT imaging at 30 days shows increased volumetric network density with a wider distribution of vessel diameters, proportionally increased branching and length, and reduced tortuosity. Global heart function is increased in the angiogenic factor-loaded cardiac implants versus sham. These findings demonstrate for the first time the efficacy of a combined remuscularization and revascularization therapy for heart regeneration after myocardial infarction.
Collapse
Affiliation(s)
- Fabiola Munarin
- Center for Biomedical Engineering, Brown University, 184 Hope St, Providence, RI, 02912, USA
| | - Rajeev J Kant
- Center for Biomedical Engineering, Brown University, 184 Hope St, Providence, RI, 02912, USA
| | - Cassady E Rupert
- Center for Biomedical Engineering, Brown University, 184 Hope St, Providence, RI, 02912, USA
| | - Amelia Khoo
- Center for Biomedical Engineering, Brown University, 184 Hope St, Providence, RI, 02912, USA
| | - Kareen L K Coulombe
- Center for Biomedical Engineering, Brown University, 184 Hope St, Providence, RI, 02912, USA.
| |
Collapse
|
12
|
Tan SY, Leung Z, Wu AR. Recreating Physiological Environments In Vitro: Design Rules for Microfluidic-Based Vascularized Tissue Constructs. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1905055. [PMID: 31913580 DOI: 10.1002/smll.201905055] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/15/2019] [Indexed: 06/10/2023]
Abstract
Vascularization of engineered tissue constructs remains one of the greatest unmet challenges to mimicking the native tissue microenvironment in vitro. The main obstacle is recapitulating the complexity of the physiological environment while providing simplicity in operation and manipulation of the model. Microfluidic technology has emerged as a promising tool that enables perfusion of the tissue constructs through engineered vasculatures and precise control of the vascular microenvironment cues in vitro. The tunable microenvironment includes i) biochemical cues such as coculture, supporting matrix, and growth factors and ii) engineering aspects such as vasculature engineering methods, fluid flow, and shear stress. In this systematic review, the design considerations of the microfluidic-based in vitro model are discussed, with an emphasis on microenvironment control to enhance the development of next-generation vascularized engineered tissues.
Collapse
Affiliation(s)
- Sin Yen Tan
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Ziuwin Leung
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Angela Ruohao Wu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
13
|
Núñez-Toldrà R, Montori S, Bosch B, Hupa L, Atari M, Miettinen S. S53P4 Bioactive Glass Inorganic Ions for Vascularized Bone Tissue Engineering by Dental Pulp Pluripotent-Like Stem Cell Cocultures. Tissue Eng Part A 2019; 25:1213-1224. [DOI: 10.1089/ten.tea.2018.0256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Raquel Núñez-Toldrà
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Barcelona, Spain
| | - Sheyla Montori
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Begoña Bosch
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Leena Hupa
- Johan Gadolin Process Chemistry Centre, Åbo Akademi University, Turku, Finland
| | - Maher Atari
- Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
- Surgery and Oral Implantology Department, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Susanna Miettinen
- Adult Stem Cell Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
14
|
Marx U, Walles H, Hoffmann S, Lindner G, Horland R, Sonntag F, Klotzbach U, Sakharov D, Tonevitsky A, Lauster R. ‘Human-on-a-chip’ Developments: A Translational Cutting-edge Alternative to Systemic Safety Assessment and Efficiency Evaluation of Substances in Laboratory Animals and Man? Altern Lab Anim 2019; 40:235-57. [DOI: 10.1177/026119291204000504] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Uwe Marx
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | | | - Silke Hoffmann
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Gerd Lindner
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Reyk Horland
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Frank Sonntag
- Fraunhofer-Institut für Werkstoff- und Strahltechnik IWS, Dresden, Germany
| | - Udo Klotzbach
- Fraunhofer-Institut für Werkstoff- und Strahltechnik IWS, Dresden, Germany
| | | | | | - Roland Lauster
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| |
Collapse
|
15
|
Zhou L, Sun S, Xu L, Yu Y, Zhang T, Wang M. DExH-Box helicase 58 enhances osteoblast differentiation of osteoblastic cells via Wnt/β-Catenin signaling. Biochem Biophys Res Commun 2019; 511:307-311. [DOI: 10.1016/j.bbrc.2019.02.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/07/2019] [Indexed: 12/24/2022]
|
16
|
Pattanaik S, Arbra C, Bainbridge H, Dennis SG, Fann SA, Yost MJ. Vascular Tissue Engineering Using Scaffold-Free Prevascular Endothelial-Fibroblast Constructs. Biores Open Access 2019; 8:1-15. [PMID: 30637179 PMCID: PMC6327854 DOI: 10.1089/biores.2018.0039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Vascularization remains a substantial limitation to the viability of engineered tissue. By comparing in vivo vascularization dynamics of a self-assembled prevascular endothelial–fibroblast model to avascular grafts, we explore the vascularization rate limitations in implants at early time intervals, during which tissue hypoxia begins to affect cell viability. Scaffold-free prevascular endothelial–fibroblast constructs (SPECs) may serve as a modular and reshapable vascular bed in replacement tissues. SPECs, fibroblast-only spheroids (FOS), and silicone implants were implanted in 54 Sprague Dawley rats and harvested at 6, 12, and 24 h (n = 5 per time point and implant type). We hypothesized that the primary endothelial networks of the SPECs allow earlier anastomosis and increased vessel formation in the interior of the implant compared to FOS and silicone implants within a 24 h window. All constructs were encapsulated by an endothelial lining at 6 h postimplantation and SPEC internal cords inosculated with the host vascular network by this time point. SPECs had a significantly higher microvascular area fraction and branch/junction density of penetrating cords at 6–12 h compared with other constructs. In addition, SPECs demonstrated perivascular cell recruitment, lumen formation, and network remodeling consistent with vessel maturation at 12–24 h; however, these implants were poorly perfused within our observation window, suggesting poor lumen patency. FOS vascular characteristics (microvessel area and penetrating cord density) increased within the 12–24 h period to represent those of the SPEC implants, suggesting a 12 h latency in host response to avascular grafts compared to prevascular grafts. Knowledge of this temporal advantage in in vitro prevascular network self-assembly as well as an understanding of the current limitations of SPEC engraftment builds on our theoretical temporal model of tissue graft vascularization and suggests a crucial time window, during which technological improvements and vascular therapy can improve engineered tissue survival.
Collapse
Affiliation(s)
- Sanket Pattanaik
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Chase Arbra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Heather Bainbridge
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Sarah Grace Dennis
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen A. Fann
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Michael J. Yost
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- Address correspondence to: Michael J. Yost, PhD, Department of Surgery, Medical University of South Carolina, 173 Ashley Avenue, Room 605, Charleston, SC 29425,
| |
Collapse
|
17
|
McKenzie JA, Maschhoff C, Liu X, Migotsky N, Silva MJ, Gardner MJ. Activation of hedgehog signaling by systemic agonist improves fracture healing in aged mice. J Orthop Res 2019; 37:51-59. [PMID: 29663560 PMCID: PMC6226344 DOI: 10.1002/jor.24017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/04/2018] [Indexed: 02/04/2023]
Abstract
Fracture healing is a complex process of many coordinated biological pathways. This system can go awry resulting in nonunion, which leads to significant patient morbidity. The Hedgehog (Hh) signaling pathway is upregulated in fracture healing. We hypothesized that the Hh signaling pathway can be pharmacologically modulated to positively affect fracture healing. Diaphyseal femur fractures were created in elderly mice (18 months, C57BL/6 females), which have a blunted and delayed healing response compared to younger mice, and were stabilized with intramedullary pins. To activate the Hh pathway we targeted the receptor Smoothened using an agonist (Hh-Ag1.5 [Hh-Ag]) and compared this to a vehicle control. Expression of Hh target genes were significantly increased in the fracture callus of the agonist group compared to controls, indicating pathway activation. Expression of osteogenic and chondrogenic-related genes was greatly upregulated in fracture callus versus intact femora, although Hh agonist treatment did not consistently enhance this response. Blindly graded, radiographic callus healing scores were significantly higher in the Hh-Ag groups at post operative day (POD) 14, indicating earlier callus bridging. On microCT, Hh-Ag treatment led to greater callus volume (+40%) and bone volume (+25%) at POD21. By day 14, callus vascularity, as assessed by 3D microCT angiography vessel volume, was 85% greater in the Hh-Ag group. Finally, mechanical strength of the calluses in the Hh-Ag groups was significantly greater than in the control groups at POD21. In conclusion, systemic administration of a Hh agonist appears to improve the osseous and vascular healing responses in a mouse fracture healing-impaired model. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
| | | | - Xiaochen Liu
- Washington University Orthopedics, St. Louis, MO
| | | | | | - Michael J. Gardner
- Washington University Orthopedics, St. Louis, MO,Stanford University Orthopedics, Stanford, CA
| |
Collapse
|
18
|
Salybekov AA, Salybekova AK, Pola R, Asahara T. Sonic Hedgehog Signaling Pathway in Endothelial Progenitor Cell Biology for Vascular Medicine. Int J Mol Sci 2018; 19:E3040. [PMID: 30301174 PMCID: PMC6213474 DOI: 10.3390/ijms19103040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 01/08/2023] Open
Abstract
The Hedgehog (HH) signaling pathway plays an important role in embryonic and postnatal vascular development and in maintaining the homeostasis of organs. Under physiological conditions, Sonic Hedgehog (SHH), a secreted protein belonging to the HH family, regulates endothelial cell growth, promotes cell migration and stimulates the formation of new blood vessels. The present review highlights recent advances made in the field of SHH signaling in endothelial progenitor cells (EPCs). The canonical and non-canonical SHH signaling pathways in EPCs and endothelial cells (ECs) related to homeostasis, SHH signal transmission by extracellular vesicles (EVs) or exosomes containing single-strand non-coding miRNAs and impaired SHH signaling in cardiovascular diseases are discussed. As a promising therapeutic tool, the possibility of using the SHH signaling pathway for the activation of EPCs in patients suffering from cardiovascular diseases is further explored.
Collapse
Affiliation(s)
- Amankeldi A Salybekov
- Department of Regenerative Medicine Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 2591193, Japan.
| | - Ainur K Salybekova
- Department of Regenerative Medicine Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 2591193, Japan.
| | - Roberto Pola
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy.
| | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 2591193, Japan.
| |
Collapse
|
19
|
Janke HP, Güvener N, Dou W, Tiemessen DM, YantiSetiasti A, Cremers JGO, Borm PJA, Feitz WFJ, Heerschap A, Kiessling F, Oosterwijk E. Labeling of Collagen Type I Templates with a Naturally Derived Contrast Agent for Noninvasive MR Imaging in Soft Tissue Engineering. Adv Healthc Mater 2018; 7:e1800605. [PMID: 30058274 DOI: 10.1002/adhm.201800605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/26/2018] [Indexed: 12/14/2022]
Abstract
In vivo monitoring of tissue-engineered constructs is important to assess their integrity, remodeling, and degradation. However, this is challenging when the contrast with neighboring tissues is low, necessitating labeling with contrast agents (CAs), but current CAs have limitations (i.e., toxicity, negative contrast, label instability, and/or inappropriate size). Therefore, a naturally derived hemin-L-lysine (HL) complex is used as a potential CA to label collagen-based templates for magnetic resonance imaging (MRI). Labeling does not change the basic characteristics of the collagen templates. When hybrid templates composed of collagen type I reinforced with degradable polymers are subcutaneously implanted in mice, longitudinal visualization by MRI is possible with good contrast and in correlation with template remodeling. In contrast, unlabeled collagen templates are hardly detectable and the fate of these templates cannot be monitored by MRI. Interestingly, tissue remodeling and vascularization are enhanced within HL-labeled templates. Thus, HL labeling is presented as a promising universal imaging marker to label tissue-engineered implants for MRI, which additionally seems to accelerate tissue regeneration.
Collapse
Affiliation(s)
- Heinz P. Janke
- Department of Urology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Geert Grooteplein 28 6525 GA Nijmegen The Netherlands
| | - Nihan Güvener
- Institute for Experimental Molecular Imaging; Center for Biohybrid Medical Systems Uniklinik RWTH and Helmholtz Institute for Biomedical Engineering; RWTH Aachen University; Forckenbeckstr. 55 52074 Aachen Germany
- Nano4Imaging GmbH; Zentrum für Biomedizintechnik (ZBMT); Pauwelsstrasse 17 52074 Aachen Germany
| | - Weiqiang Dou
- Department of Radiology and Nuclear Medicine; Radboud University Medical Center; PO Box 9101 6500 HB Nijmegen The Netherlands
- GE Healthcare; MR Research China; Beijing 100176 China
| | - Dorien M. Tiemessen
- Department of Urology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Geert Grooteplein 28 6525 GA Nijmegen The Netherlands
| | - Anglita YantiSetiasti
- Department of Urology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Geert Grooteplein 28 6525 GA Nijmegen The Netherlands
- Department of Anatomical Pathology; Faculty of Medicine; University of Padjadjaran; Jalan Professor Eyckman No. 38; Bandung 4016 Indonesia
| | - Jozef G. O. Cremers
- Institute for Experimental Molecular Imaging; Center for Biohybrid Medical Systems Uniklinik RWTH and Helmholtz Institute for Biomedical Engineering; RWTH Aachen University; Forckenbeckstr. 55 52074 Aachen Germany
- Nano4Imaging GmbH; Zentrum für Biomedizintechnik (ZBMT); Pauwelsstrasse 17 52074 Aachen Germany
| | - Paul J. A. Borm
- Nano4Imaging GmbH; Zentrum für Biomedizintechnik (ZBMT); Pauwelsstrasse 17 52074 Aachen Germany
| | - Wout F. J. Feitz
- Department of Urology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Geert Grooteplein 28 6525 GA Nijmegen The Netherlands
- Radboudumc Amalia Children's Hospital; Radboud University Medical Center; Geert Grooteplein 28 6525 GA Nijmegen The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine; Radboud University Medical Center; PO Box 9101 6500 HB Nijmegen The Netherlands
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging; Center for Biohybrid Medical Systems Uniklinik RWTH and Helmholtz Institute for Biomedical Engineering; RWTH Aachen University; Forckenbeckstr. 55 52074 Aachen Germany
| | - Egbert Oosterwijk
- Department of Urology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Geert Grooteplein 28 6525 GA Nijmegen The Netherlands
| |
Collapse
|
20
|
Abstract
Craniofacial bones, separate from the appendicular skeleton, bear a significant amount of strain and stress generated from mastication-related muscles. Current research on the regeneration of craniofacial bone focuses on the reestablishment of an elaborate vascular network. In this review, current challenges and efforts particularly in advances of scaffold properties and techniques for vascularization remodeling in craniofacial bone tissue engineering will be discussed. A microenvironment of ischemia and hypoxia in the biomaterial core drives propagation and reorganization of endothelial progenitor cells (EPCs) to assemble into a primitive microvascular framework. Co-culture strategies and delivery of vasculogenic molecules enhance EPCs' differentiation and stimulate the host regenerative response to promote vessel sprouting and strength. To optimize structural and vascular integration, well-designed microstructures of scaffolds are biologically considered. Proper porous structures, matrix stiffness, and surface morphology of scaffolds have a profound influence on cell behaviors and thus affect revascularization. In addition, advanced techniques facilitating angiogenesis and vaculogenesis have also been discussed. Oxygen delivery biomaterials, scaffold-free cell sheet techniques, and arteriovenous loop-induced axial vascularization strategies bring us new understanding and powerful strategies to manage revascularization of large craniofacial bone defects. Although promising histological results have been achieved, the efficient perfusion and functionalization of newly formed vessels are still challenging.
Collapse
Affiliation(s)
- T Tian
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - T Zhang
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Lin
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Cai
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Rademacher S, Verheijen BM, Hensel N, Peters M, Bora G, Brandes G, Vieira de Sá R, Heidrich N, Fischer S, Brinkmann H, van der Pol WL, Wirth B, Pasterkamp RJ, Claus P. Metalloprotease-mediated cleavage of PlexinD1 and its sequestration to actin rods in the motoneuron disease spinal muscular atrophy (SMA). Hum Mol Genet 2018; 26:3946-3959. [PMID: 29016853 DOI: 10.1093/hmg/ddx282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
Cytoskeletal rearrangement during axon growth is mediated by guidance receptors and their ligands which act either as repellent, attractant or both. Regulation of the actin cytoskeleton is disturbed in Spinal Muscular Atrophy (SMA), a devastating neurodegenerative disease affecting mainly motoneurons, but receptor-ligand interactions leading to the dysregulation causing SMA are poorly understood. In this study, we analysed the role of the guidance receptor PlexinD1 in SMA pathogenesis. We showed that PlexinD1 is cleaved by metalloproteases in SMA and that this cleavage switches its function from an attractant to repellent. Moreover, we found that the PlexinD1 cleavage product binds to actin rods, pathological aggregate-like structures which had so far been described for age-related neurodegenerative diseases. Our data suggest a novel disease mechanism for SMA involving formation of actin rods as a molecular sink for a cleaved PlexinD1 fragment leading to dysregulation of receptor signaling.
Collapse
Affiliation(s)
- Sebastian Rademacher
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Bert M Verheijen
- Department of Translational Neuroscience & MIND Facility, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands.,Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Niko Hensel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Miriam Peters
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases Cologne, and Institute of Genetics, University of Cologne, 50931 Cologne, Germany
| | - Gamze Bora
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, 06100 Ankara, Turkey
| | - Gudrun Brandes
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Renata Vieira de Sá
- Department of Translational Neuroscience & MIND Facility, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Natascha Heidrich
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Silke Fischer
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Hella Brinkmann
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - W Ludo van der Pol
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases Cologne, and Institute of Genetics, University of Cologne, 50931 Cologne, Germany
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience & MIND Facility, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany.,Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
22
|
Rocha LA, Sousa RA, Learmonth DA, Salgado AJ. The Role of Biomaterials as Angiogenic Modulators of Spinal Cord Injury: Mimetics of the Spinal Cord, Cell and Angiogenic Factor Delivery Agents. Front Pharmacol 2018; 9:164. [PMID: 29535633 PMCID: PMC5835322 DOI: 10.3389/fphar.2018.00164] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) represents an extremely debilitating condition for which no efficacious treatment is available. One of the main contributors to the inhospitable environment found in SCI is the vascular disruption that happens at the moment of injury that compromises the blood-spinal cord barrier (BSCB) and triggers a cascade of events that includes infiltration of inflammatory cells, ischemia and intraparenchymal hemorrhage. Due to the unsatisfactory nature of revascularization following SCI, restoring vascular perfusion and the BSCB seems an interesting way of modulating the lesion environment into a regenerative phenotype, with a potential increase in functional recovery. Certain biomaterials possess interesting features to enhance SCI therapies, and in fact have been applied as angiogenic promoters in other pathologies. The present mini-review intends to highlight the contribution that biomaterials could make in the development of novel therapeutic solutions able to restore proper vascularization and the BSCB.
Collapse
Affiliation(s)
- Luís A. Rocha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | - Rui A. Sousa
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | | | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
| |
Collapse
|
23
|
Role and regulation of growth plate vascularization during coupling with osteogenesis in tibial dyschondroplasia of chickens. Sci Rep 2018; 8:3680. [PMID: 29487404 PMCID: PMC5829164 DOI: 10.1038/s41598-018-22109-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Tibial dyschondroplasia (TD) is the most-prevalent leg disorder in fast-growing chickens; it is intractable and characterized by abnormal endochondral bone formation of proximal tibial growth-plates (TGPs). Previous studies have shown that bone is a highly vascularized tissue dependent on the coordinated coupling between angiogenesis and osteogenesis, but the underlying mechanisms of bone formation and bone remodeling are poorly defined in TD chickens. Here, we observed that inhibition of vasculogenesis and angiogenesis remarkably impaired vascular invasion in the hypertrophic chondrocyte zone of the TGPs, resulting in the massive death of chondrocytes due to a shortage of blood vessels and nutrients. Moreover, the balance of the OPG (osteoprotegerin)/RANKL (receptor activator of nuclear factor-kB ligand) system is also severely disrupted during the osteogenesis process while coupling with angiogenesis, both of which eventually lead to abnormal endochondral bone formation in TD chickens. Thus, the process of vascular formation in endochondral bone appears to initiate the pathological changes in TD, and improvement of this process during coupling with osteogenesis may be a potential therapeutic approach to treat this intractable disease.
Collapse
|
24
|
Foxm1 controls a pro-stemness microRNA network in neural stem cells. Sci Rep 2018; 8:3523. [PMID: 29476172 PMCID: PMC5824884 DOI: 10.1038/s41598-018-21876-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/12/2018] [Indexed: 01/07/2023] Open
Abstract
Cerebellar neural stem cells (NSCs) require Hedgehog-Gli (Hh-Gli) signalling for their maintenance and Nanog expression for their self-renewal. To identify novel molecular features of this regulatory pathway, we used next-generation sequencing technology to profile mRNA and microRNA expression in cerebellar NSCs, before and after induced differentiation (Diff-NSCs). Genes with higher transcript levels in NSCs (vs. Diff-NSCs) included Foxm1, which proved to be directly regulated by Gli and Nanog. Foxm1 in turn regulated several microRNAs that were overexpressed in NSCs: miR-130b, miR-301a, and members of the miR-15~16 and miR-17~92 clusters and whose knockdown significantly impaired the neurosphere formation ability. Our results reveal a novel Hh-Gli-Nanog-driven Foxm1-microRNA network that controls the self-renewal capacity of NSCs.
Collapse
|
25
|
Tissue Scaffolds As a Local Drug Delivery System for Bone Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:475-493. [DOI: 10.1007/978-981-13-0950-2_25] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
26
|
αvβ3 and α5β1 integrin-specific ligands: From tumor angiogenesis inhibitors to vascularization promoters in regenerative medicine? Biotechnol Adv 2017; 36:208-227. [PMID: 29155160 DOI: 10.1016/j.biotechadv.2017.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 12/30/2022]
Abstract
Integrins are cell adhesion receptors predominantly important during normal and tumor angiogenesis. A sequence present on several extracellular matrix proteins composed of Arg-Gly-Asp (RGD) has attracted attention due to its role in cell adhesion mediated by integrins. The development of ligands that can bind to integrins involved in tumor angiogenesis and brake disease progression has resulted in new investigational drug entities reaching the clinical trial phase in humans. The use of integrin-specific ligands can be useful for the vascularization of regenerative medicine constructs, which remains a major limitation for translation into clinical practice. In order to enhance vascularization, immobilization of integrin-specific RGD peptidomimetics within constructs is a recommended approach, due to their high specificity and selectivity towards certain desired integrins. This review endeavours to address the potential of peptidomimetic-coated biomaterials as vascular network promoters for regenerative medicine purposes. Clinical studies involving molecules tracking active integrins in cancer angiogenesis and reasons for their failure are also addressed.
Collapse
|
27
|
Chen L, Pan H, Zhang YH, Feng K, Kong X, Huang T, Cai YD. Network-Based Method for Identifying Co- Regeneration Genes in Bone, Dentin, Nerve and Vessel Tissues. Genes (Basel) 2017; 8:genes8100252. [PMID: 28974058 PMCID: PMC5664102 DOI: 10.3390/genes8100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/28/2017] [Indexed: 12/26/2022] Open
Abstract
Bone and dental diseases are serious public health problems. Most current clinical treatments for these diseases can produce side effects. Regeneration is a promising therapy for bone and dental diseases, yielding natural tissue recovery with few side effects. Because soft tissues inside the bone and dentin are densely populated with nerves and vessels, the study of bone and dentin regeneration should also consider the co-regeneration of nerves and vessels. In this study, a network-based method to identify co-regeneration genes for bone, dentin, nerve and vessel was constructed based on an extensive network of protein–protein interactions. Three procedures were applied in the network-based method. The first procedure, searching, sought the shortest paths connecting regeneration genes of one tissue type with regeneration genes of other tissues, thereby extracting possible co-regeneration genes. The second procedure, testing, employed a permutation test to evaluate whether possible genes were false discoveries; these genes were excluded by the testing procedure. The last procedure, screening, employed two rules, the betweenness ratio rule and interaction score rule, to select the most essential genes. A total of seventeen genes were inferred by the method, which were deemed to contribute to co-regeneration of at least two tissues. All these seventeen genes were extensively discussed to validate the utility of the method.
Collapse
Affiliation(s)
- Lei Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China.
| | - Hongying Pan
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, USA.
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA 02115, USA.
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Kaiyan Feng
- Department of Computer Science, Guangdong AIB Polytechnic, Guangzhou 510507, Guangdong, China.
| | - XiangYin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
28
|
Kicheva A, Rivron NC. Creating to understand - developmental biology meets engineering in Paris. Development 2017; 144:733-736. [PMID: 28246208 DOI: 10.1242/dev.144915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In November 2016, developmental biologists, synthetic biologists and engineers gathered in Paris for a meeting called 'Engineering the embryo'. The participants shared an interest in exploring how synthetic systems can reveal new principles of embryonic development, and how the in vitro manipulation and modeling of development using stem cells can be used to integrate ideas and expertise from physics, developmental biology and tissue engineering. As we review here, the conference pinpointed some of the challenges arising at the intersection of these fields, along with great enthusiasm for finding new approaches and collaborations.
Collapse
Affiliation(s)
- Anna Kicheva
- Institute of Science and Technology IST Austria, Klosterneuburg 3400, Austria
| | - Nicolas C Rivron
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht 6200 MD, The Netherlands .,Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht 3584 CT, The Netherlands
| |
Collapse
|
29
|
Fennema EM, Tchang LAH, Yuan H, van Blitterswijk CA, Martin I, Scherberich A, de Boer J. Ectopic bone formation by aggregated mesenchymal stem cells from bone marrow and adipose tissue: A comparative study. J Tissue Eng Regen Med 2017; 12:e150-e158. [PMID: 28485099 DOI: 10.1002/term.2453] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 02/16/2017] [Accepted: 05/04/2017] [Indexed: 12/29/2022]
Abstract
Tissue engineered constructs (TECs) based on spheroids of bone marrow mesenchymal stromal cells (BM-MSCs) combined with calcium phosphate microparticles and enveloped in a platelet-rich plasma hydrogel showed that aggregation of MSCs improves their ectopic bone formation potential. The stromal vascular fraction (SVF) and adipose-derived MSCs (ASCs) have been recognized as an interesting MSC source for bone tissue engineering, but their ectopic bone formation is limited. We investigated whether aggregation of ASCs could similarly improve ectopic bone formation by ASCs and SVF cells. The formation of aggregates with BM-MSCs, ASCs and SVF cells was carried out and gene expression was analysed for osteogenic, chondrogenic and vasculogenic genes in vitro. Ectopic bone formation was evaluated after implantation of TECs in immunodeficient mice with six conditions: TECs with ASCs, TECs with BM-MSC, TECs with SVF cells (with and without rhBMP2), no cells and no cells with rhBMP2. BM-MSCs showed consistent compact spheroid formation, ASCs to a lesser extent and SVF showed poor spheroid formation. Aggregation of ASCs induced a significant upregulation of the expression of osteogenic markers like alkaline phosphatase and collagen type I, as compared with un-aggregated ASCs. In vivo, ASC and SVF cells both generated ectopic bone in the absence of added morphogenetic proteins. The highest incidence of bone formation was seen with BM-MSCs (7/9) followed by SVF + rhBMP2 (4/9) and no cells + rhBMP2 (2/9). Aggregation can improve ectopic bone tissue formation by adipose-derived cells, but is less efficient than rhBMP2. A combination of both factors should now be tested to investigate an additive effect.
Collapse
Affiliation(s)
- Eelco M Fennema
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - Laurent A H Tchang
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Huipin Yuan
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands.,Xpand Biotechnology B.V., Bilthoven, the Netherlands
| | - Clemens A van Blitterswijk
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands.,MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jan de Boer
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands.,MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
30
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
31
|
Ng J, Spiller K, Bernhard J, Vunjak-Novakovic G. Biomimetic Approaches for Bone Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:480-493. [PMID: 27912680 DOI: 10.1089/ten.teb.2016.0289] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Although autologous bone grafts are considered a gold standard for the treatment of bone defects, they are limited by donor site morbidities and geometric requirements. We propose that tissue engineering technology can overcome such limitations by recreating fully viable and biological bone grafts. Specifically, we will discuss the use of bone scaffolds and autologous cells with bioreactor culture systems as a tissue engineering paradigm to grow bone in vitro. We will also discuss emergent vascularization strategies to promote graft survival in vivo, as well as the role of inflammation during bone repair. Finally, we will highlight some recent advances and discuss new solutions to bone repair inspired by endochondral ossification.
Collapse
Affiliation(s)
- Johnathan Ng
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Kara Spiller
- 2 School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania
| | - Jonathan Bernhard
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Gordana Vunjak-Novakovic
- 1 Department of Biomedical Engineering, Columbia University , New York, New York.,3 Department of Medicine, Columbia University , New York, New York
| |
Collapse
|
32
|
Rouwkema J, Khademhosseini A. Vascularization and Angiogenesis in Tissue Engineering: Beyond Creating Static Networks. Trends Biotechnol 2016; 34:733-745. [DOI: 10.1016/j.tibtech.2016.03.002] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/10/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
|
33
|
Calcium Phosphates and Angiogenesis: Implications and Advances for Bone Regeneration. Trends Biotechnol 2016; 34:983-992. [PMID: 27481474 DOI: 10.1016/j.tibtech.2016.07.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 01/10/2023]
Abstract
Calcium phosphates (CaPs) are among the most utilized synthetic biomaterials for bone regeneration, largely owing to their established osteoconductive and osteoinductive properties. While angiogenesis is a crucial prerequisite to bone formation, research and applications for CaPs have not appreciated its crucial role. This review discusses how CaPs influence angiogenesis, and highlights promising strategies that address this topic. The objective is to draw attention to the gap in the literature and to highlight the importance of angiogenesis in CaP research, development, and use.
Collapse
|
34
|
Shh mediates PDGF-induced contractile-to-synthetic phenotypic modulation in vascular smooth muscle cells through regulation of KLF4. Exp Cell Res 2016; 345:82-92. [DOI: 10.1016/j.yexcr.2016.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/11/2016] [Accepted: 05/15/2016] [Indexed: 12/31/2022]
|
35
|
Harschnitz O, van den Berg LH, Johansen LE, Jansen MD, Kling S, Vieira de Sá R, Vlam L, van Rheenen W, Karst H, Wierenga CJ, Pasterkamp RJ, van der Pol WL. Autoantibody pathogenicity in a multifocal motor neuropathy induced pluripotent stem cell-derived model. Ann Neurol 2016; 80:71-88. [DOI: 10.1002/ana.24680] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Oliver Harschnitz
- Department of Neurology and Neurosurgery; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
- Department of Translational Neuroscience; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Leonard H. van den Berg
- Department of Neurology and Neurosurgery; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Lill Eva Johansen
- Department of Translational Neuroscience; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Marc D. Jansen
- Department of Neurology and Neurosurgery; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Sandra Kling
- Department of Neurology and Neurosurgery; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Renata Vieira de Sá
- Department of Translational Neuroscience; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Lotte Vlam
- Department of Translational Neuroscience; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Wouter van Rheenen
- Department of Neurology and Neurosurgery; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
- Department of Translational Neuroscience; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Henk Karst
- Department of Translational Neuroscience; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - Corette J. Wierenga
- Division of Cell Biology, Department of Biology, Faculty of Science; Utrecht University; Utrecht the Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| | - W. Ludo van der Pol
- Department of Neurology and Neurosurgery; University Medical Center Utrecht; Brain Center Rudolf Magnus Utrecht the Netherlands
| |
Collapse
|
36
|
Vrij E, Rouwkema J, LaPointe V, van Blitterswijk C, Truckenmüller R, Rivron N. Directed Assembly and Development of Material-Free Tissues with Complex Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:4032-4039. [PMID: 27000493 DOI: 10.1002/adma.201505723] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/19/2016] [Indexed: 06/05/2023]
Abstract
Material-free tissues are assembled using solely cells. Microstructured hydrogel templates and high content screening allow the formation of centimeter-scale tissues with precise architectures. Similar to developing tissues, these contract autonomously, controllably shift shape, self-scaffold by secreting extracellular matrix, and undergo morphogenesis.
Collapse
Affiliation(s)
- Erik Vrij
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherland
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
| | - Vanessa LaPointe
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherland
| | - Clemens van Blitterswijk
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherland
| | - Roman Truckenmüller
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherland
| | - Nicolas Rivron
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherland
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584, CT, Utrecht, The Netherlands
| |
Collapse
|
37
|
Improving vascularization of engineered bone through the generation of pro-angiogenic effects in co-culture systems. Adv Drug Deliv Rev 2015; 94:116-25. [PMID: 25817732 DOI: 10.1016/j.addr.2015.03.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/27/2015] [Accepted: 03/20/2015] [Indexed: 01/31/2023]
Abstract
One of the major problems with bone tissue engineering is the development of a rapid vascularization after implantation to supply the growing osteoblast cells with the nutrients to grow and survive as well as to remove waste products. It has been demonstrated that capillary-like structures produced in vitro will anastomose rapidly after implantation and become functioning blood vessels. For this reason, in recent years many studies have examined a variety of human osteoblast and endothelial cell co-culture systems in order to distribute osteoblasts on all parts of the bone scaffold and at the same time provide conditions for the endothelial cells to migrate to form a network of capillary-like structures throughout the osteoblast-colonized scaffold. The movement and proliferation of endothelial cells to form capillary-like structures is known as angiogenesis and is dependent on a variety of pro-angiogenic factors. This review summarizes human 2- and 3-D co-culture models to date, the types and origins of cells used in the co-cultures and the proangiogenic factors that have been identified in the co-culture models.
Collapse
|
38
|
Expression pattern of sonic hedgehog signaling and calcitonin gene-related peptide in the socket healing process after tooth extraction. Biochem Biophys Res Commun 2015; 467:21-6. [PMID: 26427874 DOI: 10.1016/j.bbrc.2015.09.139] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 09/25/2015] [Indexed: 12/26/2022]
Abstract
Sonic Hedgehog (SHH), a neural development inducer, plays a significant role in the bone healing process. Calcitonin gene-related peptide (CGRP), a neuropeptide marker of sensory nerves, has been demonstrated to affect bone formation. The roles of SHH signaling and CGRP-positive sensory nerves in the alveolar bone formation process have been unknown. Here we examined the expression patterns of SHH signaling and CGRP in mouse socket by immunohistochemistry and immunofluorescence analysis. We found that the expression level of SHH peaked at day 3 and was then decreased at 5 days after tooth extraction. CGRP, PTCH1 and GLI2 were each expressed in a similar pattern with their highest expression levels at day 5 and day 7 after tooth extraction. CGRP and GLI2 were co-expressed in some inflammatory cells and bone forming cells. In some areas, CGRP-positive neurons expressed GLI2. In conclusion, SHH may affect alveolar bone healing by interacting with CGRP-positive sensory neurons and thus regulate the socket's healing process after tooth extraction.
Collapse
|
39
|
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: 287] [Impact Index Per Article: 31.9] [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
|
40
|
Abstract
Here we describe the preparation of a delivery vehicle for controlled release of Sonic hedgehog (Shh). The vehicle contains a synthetic polycation and heparin which interact by polyvalent charge attraction and rapidly self-assemble into liquid coacervate droplets. The coacervate loads Shh with high efficiency, protects its bioactivity, and provides sustained and localized release at the site of application. Shh coacervate may be injected directly or coated onto a polymeric scaffold for tissue engineering approaches, as described here.
Collapse
Affiliation(s)
- Noah Ray Johnson
- Department of Bioengineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara St.,, Pittsburgh, PA, 15219, USA
| | | |
Collapse
|
41
|
Thompson EM, Matsiko A, Farrell E, Kelly DJ, O'Brien FJ. Recapitulating endochondral ossification: a promising route toin vivobone regeneration. J Tissue Eng Regen Med 2014; 9:889-902. [DOI: 10.1002/term.1918] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/14/2014] [Accepted: 04/24/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Emmet M. Thompson
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| | - Amos Matsiko
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC; University Medical Centre Rotterdam; The Netherlands
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering; Trinity College Dublin; Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| |
Collapse
|
42
|
Current trends in bone tissue engineering. BIOMED RESEARCH INTERNATIONAL 2014; 2014:865270. [PMID: 24804256 PMCID: PMC3997160 DOI: 10.1155/2014/865270] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/09/2014] [Indexed: 01/20/2023]
Abstract
The development of tissue engineering and regeneration constitutes a new platform for translational medical research. Effective therapies for bone engineering typically employ the coordinated manipulation of cells, biologically active signaling molecules, and biomimetic, biodegradable scaffolds. Bone tissue engineering has become increasingly dependent on the merging of innovations from each of these fields, as they continue to evolve independently. This foreword will highlight some of the most recent advances in bone tissue engineering and regeneration, emphasizing the interconnected fields of stem cell biology, cell signaling biology, and biomaterial research. These include, for example, novel methods for mesenchymal stem cell purification, new methods of Wnt signaling pathway manipulation, and cutting edge computer assisted nanoscale design of bone scaffold materials. In the following
special issue, we sought to incorporate these diverse areas of emphasis in order to reflect current trends in the field.
Collapse
|
43
|
Temple JP, Hutton DL, Hung BP, Huri PY, Cook CA, Kondragunta R, Jia X, Grayson WL. Engineering anatomically shaped vascularized bone grafts with hASCs and 3D-printed PCL scaffolds. J Biomed Mater Res A 2014; 102:4317-25. [PMID: 24510413 DOI: 10.1002/jbm.a.35107] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 01/29/2014] [Indexed: 12/11/2022]
Abstract
The treatment of large craniomaxillofacial bone defects is clinically challenging due to the limited availability of transplantable autologous bone grafts and the complex geometry of the bones. The ability to regenerate new bone tissues that faithfully replicate the anatomy would revolutionize treatment options. Advances in the field of bone tissue engineering over the past few decades offer promising new treatment alternatives using biocompatible scaffold materials and autologous cells. This approach combined with recent advances in three-dimensional (3D) printing technologies may soon allow the generation of large, bioartificial bone grafts with custom, patient-specific architecture. In this study, we use a custom-built 3D printer to develop anatomically shaped polycaprolactone (PCL) scaffolds with varying internal porosities. These scaffolds are assessed for their ability to support induction of human adipose-derived stem cells (hASCs) to form vasculature and bone, two essential components of functional bone tissue. The development of functional tissues is assessed in vitro and in vivo. Finally, we demonstrate the ability to print large mandibular and maxillary bone scaffolds that replicate fine details extracted from patient's computed tomography scans. The findings of this study illustrate the capabilities and potential of 3D printed scaffolds to be used for engineering autologous, anatomically shaped, vascularized bone grafts.
Collapse
Affiliation(s)
- Joshua P Temple
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21231; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21231
| | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Abstract
Osteochondral defects are difficult to treat because the articular cartilage and the subchondral bone have dissimilar characteristics and abilities to regenerate. Bioinspired scaffolds are designed to mimic structural and biological cues of the native osteochondral unit, supporting both cartilaginous and subchondral bone repair and the integration of the newly formed osteochondral matrix with the surrounding tissues. The aim of this review is to outline fundamental requirements and strategies for the development of biomimetic scaffolds reproducing the unique and multifaceted anatomical structure of the osteochondral unit. Recent progress in preclinical animal studies using bilayer and multilayer scaffolds, together with continuous gradient scaffolds will be discussed and placed in a translational perspective with data emerging from their clinical application to treat osteochondral defects in patients.
Collapse
Affiliation(s)
- Silvia Lopa
- 1 Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute , Milan, Italy
| | | |
Collapse
|
46
|
Overexpressing sonic hedgehog peptide restores periosteal bone formation in a murine bone allograft transplantation model. Mol Ther 2013; 22:430-439. [PMID: 24089140 PMCID: PMC3916037 DOI: 10.1038/mt.2013.222] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/20/2013] [Indexed: 12/18/2022] Open
Abstract
Although activation of hedgehog (Hh) signaling has been shown to induce osteogenic differentiation in vitro and bone formation in vivo, the underlying mechanisms and the potential use of Hh-activated mesenchymal progenitors in bone defect repair remain elusive. In this study, we demonstrated that implantation of periosteal-derived mesenchymal progenitor cells (PDMPCs) that overexpressed an N-terminal sonic hedgehog peptide (ShhN) via an adenoviral vector (Ad-ShhN) restored periosteal bone collar formation in a 4-mm segmental bone allograft model in immunodeficient mice. Ad-ShhN enhanced donor cell survival and microvessel formation in collagen scaffold at 2 weeks after surgery and induced donor cell-dependent bone formation at 6 weeks after surgery. Fluorescence-activated cell sorting analysis further showed that Ad-ShhN-PDMPC-seeded scaffold contained a twofold more CD45(-)Sca-1(+)CD34(+)VEGFR2(+) endothelial progenitors than Ad-LacZ-PDMPC-seeded scaffold at day 7 after surgery. Ad-ShhN-transduced PDMPCs induced a 1.8-fold more CD31(+) microvessel formation than Ad-LacZ-transduced PDMPCs in a coculture of endothelial progenitors and PDMPCs. Taken together, our data show that overexpression of ShhN in mesenchymal progenitors improves bone defect reconstruction by enhancing donor progenitor cell survival, differentiation, and scaffold revascularization at the site of compromised periosteum. Hh agonist-based therapy, therefore, merits further investigation in tissue engineering-based applications aimed at enhancing bone defect repair and reconstruction.
Collapse
|
47
|
Boyd AL, Salci KR, Shapovalova Z, McIntyre BAS, Bhatia M. Nonhematopoietic cells represent a more rational target of in vivo hedgehog signaling affecting normal or acute myeloid leukemia progenitors. Exp Hematol 2013; 41:858-869.e4. [PMID: 23747997 DOI: 10.1016/j.exphem.2013.05.287] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 05/21/2013] [Accepted: 05/24/2013] [Indexed: 11/30/2022]
Abstract
Recent work has shown that leukemic stem cell self-renewal in chronic myeloid leukemia is dependent on cell-intrinsic hedgehog (Hh) signaling, and early clinical trials suggest that targeting this pathway is also therapeutic in patients with acute myeloid leukemia (AML). In this study, we aimed to better understand Hh signaling in normal hematopoiesis and AML by molecularly and functionally analyzing more than 200 primary human AML patient samples compared with nonleukemic controls. Gene expression analysis indicated that Hh pathway transcripts were similarly regulated in AML and nonleukemic controls, regardless of whether samples were purified based on primitive phenotypes. Consistent with these results, pharmacologic inhibition of Smoothened (SMO) did not preferentially reduce in vitro colony formation of AML versus normal progenitors. Using a unique analytic approach, messenger RNA expression of membrane receptor SMO was found to be unexpectedly rare within all hematopoietic samples analyzed, which is indicative of heterogeneity at the level of Hh signaling machinery. In contrast, abundant SMO expression could be readily detected in the nonhematopoietic fraction of human and murine bone marrow (BM) cells. Our predictions of increased SMO(+) cell frequencies within nonhematopoietic BM fractions were further supported by single-cell protein analyses. Although we did not find support for cell-autonomous sensitivity of AML cells to Hh pathway inhibition, we alternatively suggest that nonhematopoietic BM cells represent an indirect target through which primitive normal and leukemic cells can be modulated. These findings suggest current approaches to applying Hh inhibition should be carefully reevaluated to account for BM niche cell regulation that might be selectively Hh responsive.
Collapse
Affiliation(s)
- Allison L Boyd
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | | | | | | | | |
Collapse
|
48
|
Maeda Y, Hojo H, Shimohata N, Choi S, Yamamoto K, Takato T, Chung UI, Ohba S. Bone healing by sterilizable calcium phosphate tetrapods eluting osteogenic molecules. Biomaterials 2013; 34:5530-7. [PMID: 23623228 DOI: 10.1016/j.biomaterials.2013.03.089] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 03/29/2013] [Indexed: 01/12/2023]
Abstract
Although bone grafts and prosthetic implants have shown some clinical success in the treatment of bone defects, the graft availability, biocompatibility, function, and longevity still remain to be improved. One possible solution to these problems is to develop bone implants acting on host cells to induce rapid bone regeneration. Here, we demonstrate bone healing by means of a sterilizable and osteogenic molecule-eluting implant system in which two small molecules, a smoothened agonist (SAG) and a helioxanthin derivative (TH), are loaded onto tetrapod-shaped calcium phosphate granules (Tetrabone). We succeeded in directing progenitor cells toward mature osteoblasts with the combined application of the two small molecules acting on different stages of osteogenesis. Tetrabone released SAG and TH for prolonged periods when loaded with these molecules. EOG sterilization did not affect the osteogenic activity of the SAG- and TH-loaded Tetrabones. The combinatorial use of SAG- and TH-loaded Tetrabones achieved bone healing without cell transplantation in a rat femur bone defect model within two weeks. This system will allow us to vary the combination rate of implants loaded with different osteogenic factors depending on the types and sizes of defects, potentially allowing full temporal and spatial control of the bone regeneration.
Collapse
Affiliation(s)
- Yujiro Maeda
- Department of Sensory and Motor System Medicine, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lo KWH, Ashe KM, Kan HM, Laurencin CT. The role of small molecules in musculoskeletal regeneration. Regen Med 2013; 7:535-49. [PMID: 22817627 DOI: 10.2217/rme.12.33] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The uses of bone morphogenetic proteins and parathyroid hormone therapeutics are fraught with several fundamental problems, such as cost, protein stability, immunogenicity, contamination and supraphysiological dosage. These downsides may effectively limit their more universal use. Therefore, there is a clear need for alternative forms of biofactors to obviate the drawbacks of protein-based inductive factors for bone repair and regeneration. Our group has studied small molecules with the capacity to regulate osteoblast differentiation and mineralization because their inherent physical properties minimize limitations observed in protein growth factors. For instance, in general, small molecule inducers are usually more stable, highly soluble, nonimmunogenic, more affordable and require lower dosages. Small molecules with the ability to induce osteoblastic differentiation may represent the next generation of bone regenerative medicine. This review describes efforts to develop small molecule-based biofactors for induction, paying specific attention to their novel roles in bone regeneration.
Collapse
Affiliation(s)
- Kevin W-H Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | | | | | | |
Collapse
|
50
|
Fennema E, Rivron N, Rouwkema J, van Blitterswijk C, de Boer J. Spheroid culture as a tool for creating 3D complex tissues. Trends Biotechnol 2013; 31:108-15. [PMID: 23336996 DOI: 10.1016/j.tibtech.2012.12.003] [Citation(s) in RCA: 676] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 12/10/2012] [Accepted: 12/13/2012] [Indexed: 01/16/2023]
Abstract
3D cell culture methods confer a high degree of clinical and biological relevance to in vitro models. This is specifically the case with the spheroid culture, where a small aggregate of cells grows free of foreign materials. In spheroid cultures, cells secrete the extracellular matrix (ECM) in which they reside, and they can interact with cells from their original microenvironment. The value of spheroid cultures is increasing quickly due to novel microfabricated platforms amenable to high-throughput screening (HTS) and advances in cell culture. Here, we review new possibilities that combine the strengths of spheroid culture with new microenvironment fabrication methods that allow for the creation of large numbers of highly reproducible, complex tissues.
Collapse
Affiliation(s)
- Eelco Fennema
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | |
Collapse
|