201
|
Regeneration of segmental defects in metatarsus of sheep with vascularized and customized 3D-printed calcium phosphate scaffolds. Sci Rep 2020; 10:7068. [PMID: 32341459 PMCID: PMC7184564 DOI: 10.1038/s41598-020-63742-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/31/2020] [Indexed: 11/12/2022] Open
Abstract
Although autografts are considered to be the gold standard treatment for reconstruction of large bone defects resulting from trauma or diseases, donor site morbidity and limited availability restrict their use. Successful bone repair also depends on sufficient vascularization and to address this challenge, novel strategies focus on the development of vascularized biomaterial scaffolds. This pilot study aimed to investigate the feasibility of regenerating large bone defects in sheep using 3D-printed customized calcium phosphate scaffolds with or without surgical vascularization. Pre-operative computed tomography scans were performed to visualize the metatarsus and vasculature and to fabricate customized scaffolds and surgical guides by 3D printing. Critical-sized segmental defects created in the mid-diaphyseal region of the metatarsus were either left empty or treated with the 3D scaffold alone or in combination with an axial vascular pedicle. Bone regeneration was evaluated 1, 2 and 3 months post-implantation. After 3 months, the untreated defect remained non-bridged while the 3D scaffold guided bone regeneration. The presence of the vascular pedicle further enhanced bone formation. Histology confirmed bone growth inside the porous 3D scaffolds with or without vascular pedicle inclusion. Taken together, this pilot study demonstrated the feasibility of precised pre-surgical planning and reconstruction of large bone defects with 3D-printed personalized scaffolds.
Collapse
|
202
|
Three-Dimensional-Printed Poly-L-Lactic Acid Scaffolds with Different Pore Sizes Influence Periosteal Distraction Osteogenesis of a Rabbit Skull. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7381391. [PMID: 32382570 PMCID: PMC7196141 DOI: 10.1155/2020/7381391] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/29/2020] [Accepted: 03/17/2020] [Indexed: 12/25/2022]
Abstract
The repair of bone defects is a big challenge in reconstructive surgery. Periosteal distraction osteogenesis (PDO), as a promising technique used for bone regeneration, forms a space between the periosteum and bone cortex to regenerate the new bone merely by distracting the periosteum. In order to investigate the influence of distractor framework on the PDO, we utilized three-dimensional (3D) printing technology to fabricate three kinds of poly-L-lactic acid (PLLA) scaffolds with different pore sizes in this study. The in vitro experiments showed that the customized PLLA scaffolds had different-sized microchannels with low toxicity, good biocompatibility, and enough mechanical strength. Then, we built up an in vivo bioreactor under the skull periosteum of New Zealand white rabbits. The distractors with different pore sizes all could satisfy the demand of periosteal distraction in the animal experiments. After 8 weeks of consolidation period, the quality and quantity of the newly formed bone were improved with the increasing pore sizes of the distractors. Moreover, the newly formed bone also displayed an increasing degree of vascularization. In conclusion, 3D printing technology could promote the innovation of PDO devices and fabricate optimized scaffolds with appropriate pore sizes, shapes, and structures. It would help us regenerate more functional tissue-engineered bone and provide new ideas for further clinical application of the PDO technique.
Collapse
|
203
|
Injectable Therapeutic Organoids Using Sacrificial Hydrogels. iScience 2020; 23:101052. [PMID: 32353766 PMCID: PMC7191221 DOI: 10.1016/j.isci.2020.101052] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/11/2020] [Accepted: 04/03/2020] [Indexed: 12/28/2022] Open
Abstract
Organoids are becoming widespread in drug-screening technologies but have been used sparingly for cell therapy as current approaches for producing self-organized cell clusters lack scalability or reproducibility in size and cellular organization. We introduce a method of using hydrogels as sacrificial scaffolds, which allow cells to form self-organized clusters followed by gentle release, resulting in highly reproducible multicellular structures on a large scale. We demonstrated this strategy for endothelial cells and mesenchymal stem cells to self-organize into blood-vessel units, which were injected into mice, and rapidly formed perfusing vasculature. Moreover, in a mouse model of peripheral artery disease, intramuscular injections of blood-vessel units resulted in rapid restoration of vascular perfusion within seven days. As cell therapy transforms into a new class of therapeutic modality, this simple method—by making use of the dynamic nature of hydrogels—could offer high yields of self-organized multicellular aggregates with reproducible sizes and cellular architectures. Therapeutic, prevascularized organoids were formed in a sacrificial scaffold The organoids are highly reproducible and grown in a high-throughput manner The organoids rapidly formed perfusing vasculature in healthy mice Therapeutic potential was assessed in a mouse model of peripheral artery disease
Collapse
|
204
|
Pradhan S, Banda OA, Farino CJ, Sperduto JL, Keller KA, Taitano R, Slater JH. Biofabrication Strategies and Engineered In Vitro Systems for Vascular Mechanobiology. Adv Healthc Mater 2020; 9:e1901255. [PMID: 32100473 PMCID: PMC8579513 DOI: 10.1002/adhm.201901255] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The vascular system is integral for maintaining organ-specific functions and homeostasis. Dysregulation in vascular architecture and function can lead to various chronic or acute disorders. Investigation of the role of the vascular system in health and disease has been accelerated through the development of tissue-engineered constructs and microphysiological on-chip platforms. These in vitro systems permit studies of biochemical regulation of vascular networks and parenchymal tissue and provide mechanistic insights into the biophysical and hemodynamic forces acting in organ-specific niches. Detailed understanding of these forces and the mechanotransductory pathways involved is necessary to develop preventative and therapeutic strategies targeting the vascular system. This review describes vascular structure and function, the role of hemodynamic forces in maintaining vascular homeostasis, and measurement approaches for cell and tissue level mechanical properties influencing vascular phenomena. State-of-the-art techniques for fabricating in vitro microvascular systems, with varying degrees of biological and engineering complexity, are summarized. Finally, the role of vascular mechanobiology in organ-specific niches and pathophysiological states, and efforts to recapitulate these events using in vitro microphysiological systems, are explored. It is hoped that this review will help readers appreciate the important, but understudied, role of vascular-parenchymal mechanotransduction in health and disease toward developing mechanotherapeutics for treatment strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Omar A. Banda
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John L. Sperduto
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Keely A. Keller
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Ryan Taitano
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716, USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
205
|
Xie R, Zheng W, Guan L, Ai Y, Liang Q. Engineering of Hydrogel Materials with Perfusable Microchannels for Building Vascularized Tissues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1902838. [PMID: 31559675 DOI: 10.1002/smll.201902838] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/06/2019] [Indexed: 05/23/2023]
Abstract
Vascular systems are responsible for various physiological and pathological processes related to all organs in vivo, and the survival of engineered tissues for enough nutrient supply in vitro. Thus, biomimetic vascularization is highly needed for constructing both a biomimetic organ model and a reliable engineered tissue. However, many challenges remain in constructing vascularized tissues, requiring the combination of suitable biomaterials and engineering techniques. In this review, the advantages of hydrogels on building engineered vascularized tissues are discussed and recent engineering techniques for building perfusable microchannels in hydrogels are summarized, including micromolding, 3D printing, and microfluidic spinning. Furthermore, the applications of these perfusable hydrogels in manufacturing organ-on-a-chip devices and transplantable engineered tissues are highlighted. Finally, current challenges in recapitulating the complexity of native vascular systems are discussed and future development of vascularized tissues is prospected.
Collapse
Affiliation(s)
- Ruoxiao Xie
- MOE Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Wenchen Zheng
- MOE Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Liandi Guan
- MOE Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yongjian Ai
- MOE Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Qionglin Liang
- MOE Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
206
|
Liao X, Yang X, Deng H, Hao Y, Mao L, Zhang R, Liao W, Yuan M. Injectable Hydrogel-Based Nanocomposites for Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 8:251. [PMID: 32296694 PMCID: PMC7136457 DOI: 10.3389/fbioe.2020.00251] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs), including a series of pathological disorders, severely affect millions of people all over the world. To address this issue, several potential therapies have been developed for treating CVDs, including injectable hydrogels as a minimally invasive method. However, the utilization of injectable hydrogel is a bit restricted recently owing to some limitations, such as transporting the therapeutic agent more accurately to the target site and prolonging their retention locally. This review focuses on the advances in injectable hydrogels for CVD, detailing the types of injectable hydrogels (natural or synthetic), especially that complexed with stem cells, cytokines, nano-chemical particles, exosomes, genetic material including DNA or RNA, etc. Moreover, we summarized the mainly prominent mechanism, based on which injectable hydrogel present excellent treating effect of cardiovascular repair. All in all, it is hopefully that injectable hydrogel-based nanocomposites would be a potential candidate through cardiac repair in CVDs treatment.
Collapse
Affiliation(s)
- Xiaoshan Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xushan Yang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hong Deng
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lianzhi Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Rongjun Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Miaomiao Yuan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
207
|
A retrievable implant for the long-term encapsulation and survival of therapeutic xenogeneic cells. Nat Biomed Eng 2020; 4:814-826. [PMID: 32231313 PMCID: PMC8051527 DOI: 10.1038/s41551-020-0538-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 02/18/2020] [Indexed: 12/20/2022]
Abstract
The long-term function of transplanted therapeutic cells typically requires systemic immune suppression. Here, we show that a retrievable implant comprising of a silicone reservoir and a porous polymeric membrane protects human cells encapsulated in it after implant transplantation in the intraperitoneal space of immunocompetent mice. Membranes with pores 1 µm in diameter allowed host macrophages to migrate into the device without the loss of transplanted cells, whereas membranes with pore sizes under 0.8 µm prevented their infiltration by immune cells. A synthetic polymer coating prevented fibrosis and was necessary for the long-term function of the device. For over 130 days the device supported human cells engineered to secrete erythropoietin in immunocompetent mice as well as transgenic human cells carrying an inducible gene circuit for the on-demand secretion of erythropoietin. Pancreatic islets from rats encapsulated in the device and implanted in diabetic mice restored normoglycaemia in the mice for over 75 days. The biocompatible device provides a retrievable solution for the transplantation of engineered cells in the absence of immunosuppression.
Collapse
|
208
|
Shahabipour F, Oskuee RK, Dehghani H, Shokrgozar MA, Aninwene GE, Bonakdar S. Cell-cell interaction in a coculture system consisting of CRISPR/Cas9 mediated GFP knock-in HUVECs and MG-63 cells in alginate-GelMA based nanocomposites hydrogel as a 3D scaffold. J Biomed Mater Res A 2020; 108:1596-1606. [PMID: 32180319 DOI: 10.1002/jbm.a.36928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
The interaction between osteogenic and angiogenic cells through a coculturing system in biocompatible materials has been considered for successfully engineering vascularized bone tissue equivalents. In this study, we developed a hydrogel-blended scaffold consisted of gelatin methacryloyl (GelMA) and alginate enriched with hydroxyapatite nanoparticles (HAP) to model an in vitro prevascularized bone construct. The hydrogel-based scaffold revealed a higher mechanical stiffness than those of pure (GelMA), alginate, and (GelMA+ HAP) hydrogels. In the present study, we generated a green fluorescent protein (GFP) knock-in umbilical vein endothelial cells (HUVECs) cell line using the CRISPR/Cas9 technology. The GFP was inserted into the human-like ROSA locus of HUVECs genome. HUVECs expressing GFP were cocultured with OB-like cells (MG-63) within three-dimensionally (3D) fabricated hydrogel to investigate the response of cocultured osteoblasts and endothelial cells in a 3D structure. Cell viability under the 3D cocultured gel was higher than the 3D monocultured. Compared to the 3D monocultured condition, the cells were aligned and developed into the vessel-like structures. During 14 days of culture periods, the cells displayed actin protrusions by the formation of spike-like filopodia in the 3D cocultured model. Angiogenic and osteogenic-related genes such as CD31, vWF, and osteocalcin showed higher expression in the cocultured versus the monocultured. These results have collectively indicated that the 3D cocultured hydrogel facilitates interaction among cells, thereby having a greater effect on angiogenic and osteogenic properties in the absence of induction media.
Collapse
Affiliation(s)
| | - Reza K Oskuee
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Basic Science, Faculty of Veterinary medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - George E Aninwene
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
209
|
Hoyle HW, Smith LA, Williams RJ, Przyborski SA. Applications of novel bioreactor technology to enhance the viability and function of cultured cells and tissues. Interface Focus 2020; 10:20190090. [PMID: 32194933 DOI: 10.1098/rsfs.2019.0090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
As the field of tissue engineering continues to advance rapidly, so too does the complexity of cell culture techniques used to generate in vitro tissue constructs, with the overall aim of mimicking the in vivo microenvironment. This complexity typically comes at a cost with regards to the size of the equipment required and associated expenses. We have developed a small, low-cost bioreactor system which overcomes some of the issues of typical bioreactor systems while retaining a suitable scale for the formation of complex tissues. Herein, we have tested this system with three cell populations/tissues: the culture of hepatocellular carcinoma cells, where an improved structure and basic metabolic function is seen; the culture of human pluripotent stem cells, in which the cultures can form more heterogeneous tissues resembling the in vivo teratoma and ex vivo liver tissue slices, in which improved maintenance of cellular viability is seen over the 3 days tested. This system has the flexibility to be used for a variety of further uses and has the potential to provide a more accessible alternative to current bioreactor technologies.
Collapse
Affiliation(s)
- H W Hoyle
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - L A Smith
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - R J Williams
- Department of Engineering, Durham University, South Road, Durham DH1 3LE, UK
| | - S A Przyborski
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK.,Reprocell Europe Ltd, NETPark Incubator, Thomas Wright Way, Sedgefield TS21 3FD, UK
| |
Collapse
|
210
|
Tang F, Manz XD, Bongers A, Odell RA, Joukhdar H, Whitelock JM, Lord MS, Rnjak-Kovacina J. Microchannels Are an Architectural Cue That Promotes Integration and Vascularization of Silk Biomaterials in Vivo. ACS Biomater Sci Eng 2020; 6:1476-1486. [DOI: 10.1021/acsbiomaterials.9b01624] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Fengying Tang
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Xue D. Manz
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
- Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam Cardiovascular Sciences (ACS), Amsterdam 1081 HV, The Netherlands
| | - Andre Bongers
- Biological Resources Imaging Laboratory, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Ross A. Odell
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Habib Joukhdar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - John M. Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Megan S. Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
211
|
Mori T, Cukelj R, Prévôt ME, Ustunel S, Story A, Gao Y, Diabre K, McDonough JA, Freeman EJ, Hegmann E, Clements RJ. 3D Porous Liquid Crystal Elastomer Foams Supporting Long-term Neuronal Cultures. Macromol Rapid Commun 2020; 41:e1900585. [PMID: 32009277 DOI: 10.1002/marc.201900585] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/18/2019] [Indexed: 02/05/2023]
Abstract
3D liquid crystal elastomer (3D-LCE) foams are used to support long-term neuronal cultures for over 60 days. Sequential imaging shows that cell density remains relatively constant throughout the culture period while the number of cells per observational area increases. In a subset of samples, retinoic acid is used to stimulate extensive neuritic outgrowth and maturation of proliferated neurons within the LCEs, inducing a threefold increase in length with cells displaying morphologies indicative of mature neurons. Designed LCEs' micro-channels have a similar diameter to endogenous parenchymal arterioles, ensuring that neurons throughout the construct have constant access to growth media during extended experiments. Here it is shown that 3D-LCEs provide a unique environment and simple method to longitudinally study spatial neuronal function, not possible in conventional culture environments, with simplistic integration into existing methodological pipelines.
Collapse
Affiliation(s)
- Taizo Mori
- Advanced Materials and Liquid Crystal Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Richard Cukelj
- Department of Biological Sciences, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Marianne Estelle Prévôt
- Advanced Materials and Liquid Crystal Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Senay Ustunel
- Advanced Materials and Liquid Crystal Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Chemical Physics Interdisciplinary Program, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Anna Story
- Department of Biological Sciences, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Yunxiang Gao
- Advanced Materials and Liquid Crystal Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Karene Diabre
- Advanced Materials and Liquid Crystal Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Jennifer Ann McDonough
- Department of Biological Sciences, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Biomedical Sciences Program, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Brain Health Research Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Ernest Johnson Freeman
- Department of Biological Sciences, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Biomedical Sciences Program, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Brain Health Research Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Elda Hegmann
- Advanced Materials and Liquid Crystal Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Department of Biological Sciences, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Biomedical Sciences Program, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Brain Health Research Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Chemical Physics Interdisciplinary Program, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| | - Robert John Clements
- Department of Biological Sciences, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Biomedical Sciences Program, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA.,Brain Health Research Institute, 1425 Lefton Esplanade, Kent State University, Kent, Ohio, 44242-0001, USA
| |
Collapse
|
212
|
Shahabipour F, Ashammakhi N, Oskuee RK, Bonakdar S, Hoffman T, Shokrgozar MA, Khademhosseini A. Key components of engineering vascularized 3-dimensional bioprinted bone constructs. Transl Res 2020; 216:57-76. [PMID: 31526771 DOI: 10.1016/j.trsl.2019.08.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/16/2022]
Abstract
Vascularization has a pivotal role in engineering successful tissue constructs. However, it remains a major hurdle of bone tissue engineering, especially in clinical applications for the treatment of large bone defects. Development of vascularized and clinically-relevant engineered bone substitutes with sufficient blood supply capable of maintaining implant viability and supporting subsequent host tissue integration remains a major challenge. Since only cells that are 100-200 µm from blood vessels can receive oxygen through diffusion, engineered constructs that are thicker than 400 µm face a challenging oxygenation problem. Following implantation in vivo, spontaneous ingrowth of capillaries in thick engineered constructs is too slow. Thus, it is critical to provide optimal conditions to support vascularization in engineered bone constructs. To achieve this, an in-depth understanding of the mechanisms of angiogenesis and bone development is required. In addition, it is also important to mimic the physiological milieu of native bone to fabricate more successful vascularized bone constructs. Numerous applications of engineered vascularization with cell-and/or microfabrication-based approaches seek to meet these aims. Three-dimensional (3D) printing promises to create patient-specific bone constructs in the future. In this review, we discuss the major components of fabricating vascularized 3D bioprinted bone constructs, analyze their related challenges, and highlight promising future trends.
Collapse
Affiliation(s)
- Fahimeh Shahabipour
- National cell bank of Iran, Pasteur Institute of Iran, Tehran, Iran; Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California; Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California
| | - Reza K Oskuee
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahin Bonakdar
- National cell bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Tyler Hoffman
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | | | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, University of California, Los Angeles, Los Angeles, California; Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
213
|
Gniesmer S, Brehm R, Hoffmann A, de Cassan D, Menzel H, Hoheisel AL, Glasmacher B, Willbold E, Reifenrath J, Ludwig N, Zimmerer R, Tavassol F, Gellrich NC, Kampmann A. Vascularization and biocompatibility of poly(ε-caprolactone) fiber mats for rotator cuff tear repair. PLoS One 2020; 15:e0227563. [PMID: 31929570 PMCID: PMC6957163 DOI: 10.1371/journal.pone.0227563] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Rotator cuff tear is the most frequent tendon injury in the adult population. Despite current improvements in surgical techniques and the development of grafts, failure rates following tendon reconstruction remain high. New therapies, which aim to restore the topology and functionality of the interface between muscle, tendon and bone, are essentially required. One of the key factors for a successful incorporation of tissue engineered constructs is a rapid ingrowth of cells and tissues, which is dependent on a fast vascularization. The dorsal skinfold chamber model in female BALB/cJZtm mice allows the observation of microhemodynamic parameters in repeated measurements in vivo and therefore the description of the vascularization of different implant materials. In order to promote vascularization of implant material, we compared a porous polymer patch (a commercially available porous polyurethane based scaffold from Biomerix™) with electrospun polycaprolactone (PCL) fiber mats and chitosan-graft-PCL coated electrospun PCL (CS-g-PCL) fiber mats in vivo. Using intravital fluorescence microscopy microcirculatory parameters were analyzed repetitively over 14 days. Vascularization was significantly increased in CS-g-PCL fiber mats at day 14 compared to the porous polymer patch and uncoated PCL fiber mats. Furthermore CS-g-PCL fiber mats showed also a reduced activation of immune cells. Clinically, these are important findings as they indicate that the CS-g-PCL improves the formation of vascularized tissue and the ingrowth of cells into electrospun PCL scaffolds. Especially the combination of enhanced vascularization and the reduction in immune cell activation at the later time points of our study points to an improved clinical outcome after rotator cuff tear repair.
Collapse
Affiliation(s)
- Sarah Gniesmer
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
- NIFE—Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Ralph Brehm
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Andrea Hoffmann
- NIFE—Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
- Department of Orthopedic Surgery, Laboratory for Biomechanics and Biomaterials, Graded Implants and Regenerative Strategies, Hannover Medical School, Hannover, Germany
| | - Dominik de Cassan
- Institute for Technical Chemistry, Braunschweig University of Technology, Braunschweig, Germany
| | - Henning Menzel
- Institute for Technical Chemistry, Braunschweig University of Technology, Braunschweig, Germany
| | - Anna Lena Hoheisel
- NIFE—Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
- Institute of Multiphase Processes, Leibniz University Hannover, Hannover, Germany
| | - Birgit Glasmacher
- NIFE—Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
- Institute of Multiphase Processes, Leibniz University Hannover, Hannover, Germany
| | - Elmar Willbold
- NIFE—Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
- Department of Orthopedic Surgery, Hannover Medical School, Hannover, Germany
| | - Janin Reifenrath
- NIFE—Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
- Department of Orthopedic Surgery, Hannover Medical School, Hannover, Germany
| | - Nils Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Ruediger Zimmerer
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Frank Tavassol
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Nils-Claudius Gellrich
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Andreas Kampmann
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
- NIFE—Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| |
Collapse
|
214
|
Najdahmadi A, Smink AM, de Vos P, Lakey JR, Botvinick E. Non-Invasive Monitoring of Oxygen Tension and Oxygen Transport Inside Subcutaneous Devices After H 2S Treatment. Cell Transplant 2020; 29:963689719893936. [PMID: 32024377 PMCID: PMC7444232 DOI: 10.1177/0963689719893936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 10/21/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
Medical devices for cell therapy can be improved through prevascularization. In this work we study the vascularization of a porous polymer device, previously used by our group for pancreatic islet transplantation with results indicating improved glycemic control. Oxygen partial pressure within such devices was monitored non-invasively using an optical technique. Oxygen-sensitive tubes were fabricated and placed inside devices prior to subcutaneous implantation in nude mice. We tested the hypothesis that vascularization will be enhanced by administration of the pro-angiogenic factor hydrogen sulfide (H2S). We found that oxygen dynamics were unique to each implant and that the administration of H2S does not result in significant changes in perfusion of the devices as compared with control. These observations suggest that vascular perfusion and density are not necessarily correlated, and that the rate of vascularization was not enhanced by the pro-angiogenic agent.
Collapse
Affiliation(s)
- Avid Najdahmadi
- Department of Materials Science and Engineering, University of
California Irvine, Irvine, CA, USA
| | - Alexandra M. Smink
- Department of Pathology and Medical Biology, University Medical
Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical
Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jonathan R.T. Lakey
- Department of Biomedical Engineering, University of California
Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA,
USA
| | - Elliot Botvinick
- Department of Materials Science and Engineering, University of
California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California
Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA,
USA
| |
Collapse
|
215
|
Pang Y, Sutoko S, Wang Z, Horimoto Y, Montagne K, Horiguchi I, Shinohara M, Danoy M, Niino T, Sakai Y. Organization of liver organoids using Raschig ring-like micro-scaffolds and triple co-culture: Toward modular assembly-based scalable liver tissue engineering. Med Eng Phys 2019; 76:69-78. [PMID: 31883633 DOI: 10.1016/j.medengphy.2019.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 10/16/2019] [Accepted: 10/20/2019] [Indexed: 12/26/2022]
Abstract
In order to address the remaining issues of fragile structure and insufficient mass transfer faced in modular assembly-based liver tissue engineering, a Raschig ring-like hollowed micro-scaffold was proposed and fabricated using poly-ε-caprolactone with 60% porosity and 11.4 mm2 effective surface area for cell immobilization. The method of cell inoculation, the types of cells for co-culture and the scalability of the proposed hollowed micro-scaffold in perfusion were all investigated to obtain an optimized organoid made of tissue modules. Extracellular matrix was found necessary to establish a hierarchical co-culture, and the triple co-culture of Human Hepatoma Hep G2 cells, liver sinusoid cell line TMNK-1 cells and fibroblasts (Swiss 3T3 cells) was recognized to be the most efficient to obtain higher cell attachment, proliferation and hepatic function. The equipped intersecting hollow channels provided in the micro-scaffold functioned as flow paths to promote mass transfer to the immobilized cells after the modules have been randomly packed into a bioreactor for perfusion culture, and resulted in enhanced albumin production and high cellular viability. Cell density comparable to those found in vivo were obtained in the perfused construct, which also maintained its rigid structure. Those results suggest that modular tissues made with hollowed micro-scaffold-based organoids hold great potential for scaling up tissue engineered constructs towards implantation.
Collapse
Affiliation(s)
- Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Room A730, Lee Shau Kee science and Technology Building Haidian District, Beijing 100084, China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, China.
| | - Stephanie Sutoko
- Institute of Industrial Science, University of Tokyo, Komaba 4-6-1, Meguro-ku, Tokyo 153-8505, Japan
| | - Zitong Wang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Haidian District, Beijing 100084, China
| | - Yohei Horimoto
- Graduate School of Engineering, Shibaura Institute of Technology, Shibaura 3-9-14, Minato-ku, Tokyo 108-8548, Japan
| | - Kevin Montagne
- Department of Mechanical Engineering, Graduate School of Engineering, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-033, Japan
| | - Ikki Horiguchi
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Japan
| | - Marie Shinohara
- Institute of Industrial Science, University of Tokyo, Komaba 4-6-1, Meguro-ku, Tokyo 153-8505, Japan
| | - Mathieu Danoy
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-033, Japan
| | - Toshiki Niino
- Institute of Industrial Science, University of Tokyo, Komaba 4-6-1, Meguro-ku, Tokyo 153-8505, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-033, Japan
| |
Collapse
|
216
|
Mesenchymal Stem Cells Attract Endothelial Progenitor Cells via a Positive Feedback Loop between CXCR2 and CXCR4. Stem Cells Int 2019; 2019:4197164. [PMID: 31885605 PMCID: PMC6915119 DOI: 10.1155/2019/4197164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 08/04/2019] [Accepted: 09/11/2019] [Indexed: 01/25/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can attract host endothelial progenitor cells (EPCs) to promote vascularization in tissue-engineered constructs (TECs). Nevertheless, the underlying mechanism remains vague. This study is aimed at investigating the roles of CXCR2 and CXCR4 in the EPC migration towards MSCs. In vitro, Transwell assays were performed to evaluate the migration of EPCs towards MSCs. Antagonists and shRNAs targeting CXCR2, CXCR4, and JAK/STAT3 were applied for the signaling blockade. Western blot and RT-PCR were conducted to analyze the molecular events in EPCs. In vivo, TECs were constructed and subcutaneously implanted into GFP+ transgenic mice. Signaling inhibitors were injected in an orientated manner into TECs. Recruitment of host CD34+ cells was evaluated by immunofluorescence. Eventually, we demonstrated that CXCR2 and CXCR4 were both highly expressed in migrated EPCs and indispensable for MSC-induced EPC migration. CXCR2 and CXCR4 strongly correlated with each other in the way that the expression of CXCR2 and CXCR2-mediated migration depends on the activity of CXCR4 and vice versa. Further studies documented that both of CXCR2 and CXCR4 activated STAT3 signaling, which in turn regulated the expression of CXCR2 and CXCR4, as well as cell migration. In summary, we firstly introduced a reciprocal crosstalk between CXCR2 and CXCR4 in the context of EPC migration. This feedback loop plays critical roles in the migration of EPCs towards MSCs.
Collapse
|
217
|
Zucchelli E, Majid QA, Foldes G. New artery of knowledge: 3D models of angiogenesis. VASCULAR BIOLOGY 2019; 1:H135-H143. [PMID: 32923965 PMCID: PMC7439835 DOI: 10.1530/vb-19-0026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022]
Abstract
Angiogenesis and vasculogenesis are complex processes by which new blood vessels are formed and expanded. They play a pivotal role not only in physiological development and growth and tissue and organ repair, but also in a range of pathological conditions, from tumour formation to chronic inflammation and atherosclerosis. Understanding the multistep cell-differentiation programmes and identifying the key molecular players of physiological angiogenesis/vasculogenesis are critical to tackle pathological mechanisms. While many questions are yet to be answered, increasingly sophisticated in vitro, in vivo and ex vivo models of angiogenesis/vasculogenesis, together with cutting-edge imaging techniques, allowed for recent major advances in the field. This review aims to summarise the three-dimensional models available to study vascular network formation and to discuss advantages and limitations of the current systems.
Collapse
Affiliation(s)
| | - Qasim A Majid
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Gabor Foldes
- National Heart and Lung Institute, Imperial College London, London, UK.,Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
218
|
Soucy JR, Askaryan J, Diaz D, Koppes AN, Annabi N, Koppes RA. Glial cells influence cardiac permittivity as evidenced through in vitro and in silico models. Biofabrication 2019; 12:015014. [PMID: 31593932 PMCID: PMC11062241 DOI: 10.1088/1758-5090/ab4c0a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Excitation-contraction (EC) coupling in the heart has, until recently, been solely accredited to cardiomyocytes. The inherent complexities of the heart make it difficult to examine non-muscle contributions to contraction in vivo, and conventional in vitro models fail to capture multiple features and cellular heterogeneity of the myocardium. Here, we report on the development of a 3D cardiac μTissue to investigate changes in the cellular composition of native myocardium in vitro. Cells are encapsulated within micropatterned gelatin-based hydrogels formed via visible light photocrosslinking. This system enables spatial control of the microarchitecture, perturbation of the cellular composition, and functional measures of EC coupling via video microscopy and a custom algorithm to quantify beat frequency and degree of coordination. To demonstrate the robustness of these tools and evaluate the impact of altered cell population densities on cardiac μTissues, contractility and cell morphology were assessed with the inclusion of exogenous non-myelinating Schwann cells (SCs). Results demonstrate that the addition of exogenous SCs alter cardiomyocyte EC, profoundly inhibiting the response to electrical pacing. Computational modeling of connexin-mediated coupling suggests that SCs impact cardiomyocyte resting potential and rectification following depolarization. Cardiac μTissues hold potential for examining the role of cellular heterogeneity in heart health, pathologies, and cellular therapies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, United States of America
| | | | | | | | | | | |
Collapse
|
219
|
Cho WC, Jour G, Aung PP. Role of angiogenesis in melanoma progression: Update on key angiogenic mechanisms and other associated components. Semin Cancer Biol 2019; 59:175-186. [PMID: 31255774 DOI: 10.1016/j.semcancer.2019.06.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/05/2019] [Accepted: 06/26/2019] [Indexed: 01/09/2023]
Abstract
Angiogenesis, the formation of new blood vessels from existing blood vessels, is a complex and highly regulated process that plays a role in a wide variety of physiological and pathological processes. In malignancy, angiogenesis is essential for neoplastic cells to acquire the nutrients and oxygen critical for their continued proliferation. Angiogenesis requires a sequence of well-coordinated events mediated by a number of tightly regulated interactions between pro-angiogenic factors and their corresponding receptors expressed on various vascular components (e.g., endothelial cells and pericytes) and stromal components forming the extracellular matrix. In this review, we discuss the functional roles of key growth factors and cytokines known to promote angiogenesis in cutaneous melanoma and key factors implicated in the extracellular matrix remodeling that acts synergistically with angiogenesis to promote tumor progression in melanoma, incorporating some of the most up-to-date basic science knowledge from recently published in vivo and in vitro experimental studies.
Collapse
Affiliation(s)
- Woo Cheal Cho
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George Jour
- Department of Pathology and Dermatology, NYU Langone Medical Center, New York, NY, USA
| | - Phyu P Aung
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
220
|
Enhanced Host Neovascularization of Prevascularized Engineered Muscle Following Transplantation into Immunocompetent versus Immunocompromised Mice. Cells 2019; 8:cells8121472. [PMID: 31757007 PMCID: PMC6953003 DOI: 10.3390/cells8121472] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/10/2019] [Accepted: 11/18/2019] [Indexed: 01/03/2023] Open
Abstract
: Engineering of functional tissue, by combining either autologous or allogeneic cells with biomaterials, holds promise for the treatment of various diseases and injuries. Prevascularization of the engineered tissue was shown to enhance and improve graft integration and neovascularization post-implantation in immunocompromised mice. However, the neovascularization and integration processes of transplanted engineered tissues have not been widely studied in immunocompetent models. Here, we fabricated a three-dimensional (3D) vascularized murine muscle construct that was transplanted into immunocompetent and immunocompromised mice. Intravital imaging demonstrated enhanced neovascularization in immunocompetent mice compared to immunocompromised mice, 18 days post-implantation, indicating the advantageous effect of an intact immune system on neovascularization. Moreover, construct prevascularization enhanced neovascularization, integration, and myogenesis in both animal models. These findings demonstrate the superiority of implantation into immunocompetent over immunocompromised mice and, therefore, suggest that using autologous cells might be beneficial compared to allogeneic cells and subsequent immunosuppression. Taken together, these observations have the potential to advance the field of regenerative medicine and tissue engineering, ultimately reducing the need for donor organs and tissues.
Collapse
|
221
|
Espona-Noguera A, Ciriza J, Cañibano-Hernández A, Orive G, Hernández RM, Saenz del Burgo L, Pedraz JL. Review of Advanced Hydrogel-Based Cell Encapsulation Systems for Insulin Delivery in Type 1 Diabetes Mellitus. Pharmaceutics 2019; 11:E597. [PMID: 31726670 PMCID: PMC6920807 DOI: 10.3390/pharmaceutics11110597] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022] Open
Abstract
: Type 1 Diabetes Mellitus (T1DM) is characterized by the autoimmune destruction of β-cells in the pancreatic islets. In this regard, islet transplantation aims for the replacement of the damaged β-cells through minimally invasive surgical procedures, thereby being the most suitable strategy to cure T1DM. Unfortunately, this procedure still has limitations for its widespread clinical application, including the need for long-term immunosuppression, the lack of pancreas donors and the loss of a large percentage of islets after transplantation. To overcome the aforementioned issues, islets can be encapsulated within hydrogel-like biomaterials to diminish the loss of islets, to protect the islets resulting in a reduction or elimination of immunosuppression and to enable the use of other insulin-producing cell sources. This review aims to provide an update on the different hydrogel-based encapsulation strategies of insulin-producing cells, highlighting the advantages and drawbacks for a successful clinical application.
Collapse
Affiliation(s)
- Albert Espona-Noguera
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Jesús Ciriza
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Alberto Cañibano-Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01006 Vitoria, Spain
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore
| | - Rosa María Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Laura Saenz del Burgo
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
222
|
Hao D, He C, Ma B, Lankford L, Reynaga L, Farmer DL, Guo F, Wang A. Hypoxic Preconditioning Enhances Survival and Proangiogenic Capacity of Human First Trimester Chorionic Villus-Derived Mesenchymal Stem Cells for Fetal Tissue Engineering. Stem Cells Int 2019; 2019:9695239. [PMID: 31781252 PMCID: PMC6874947 DOI: 10.1155/2019/9695239] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/23/2019] [Accepted: 09/04/2019] [Indexed: 12/19/2022] Open
Abstract
Prenatal stem cell-based regenerative therapies have progressed substantially and have been demonstrated as effective treatment options for fetal diseases that were previously deemed untreatable. Due to immunoregulatory properties, self-renewal capacity, and multilineage potential, autologous human placental chorionic villus-derived mesenchymal stromal cells (CV-MSCs) are an attractive cell source for fetal regenerative therapies. However, as a general issue for MSC transplantation, the poor survival and engraftment is a major challenge of the application of MSCs. Particularly for the fetal transplantation of CV-MSCs in the naturally hypoxic fetal environment, improving the survival and engraftment of CV-MSCs is critically important. Hypoxic preconditioning (HP) is an effective priming approach to protect stem cells from ischemic damage. In this study, we developed an optimal HP protocol to enhance the survival and proangiogenic capacity of CV-MSCs for improving clinical outcomes in fetal applications. Total cell number, DNA quantification, nuclear area test, and cell viability test showed HP significantly protected CV-MSCs from ischemic damage. Flow cytometry analysis confirmed HP did not alter the immunophenotype of CV-MSCs. Caspase-3, MTS, and Western blot analysis showed HP significantly reduced the apoptosis of CV-MSCs under ischemic stimulus via the activation of the AKT signaling pathway that was related to cell survival. ELISA results showed HP significantly enhanced the secretion of vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) by CV-MSCs under an ischemic stimulus. We also found that the environmental nutrition level was critical for the release of brain-derived neurotrophic factor (BDNF). The angiogenesis assay results showed HP-primed CV-MSCs could significantly enhance endothelial cell (EC) proliferation, migration, and tube formation. Consequently, HP is a promising strategy to increase the tolerance of CV-MSCs to ischemia and improve their therapeutic efficacy in fetal clinical applications.
Collapse
Affiliation(s)
- Dake Hao
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Chuanchao He
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Bowen Ma
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Lee Lankford
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Lizette Reynaga
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Diana L. Farmer
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Fuzheng Guo
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
223
|
Kaiser NJ, Bellows JA, Kant RJ, Coulombe KLK. Digital Design and Automated Fabrication of Bespoke Collagen Microfiber Scaffolds. Tissue Eng Part C Methods 2019; 25:687-700. [PMID: 31017039 PMCID: PMC6859695 DOI: 10.1089/ten.tec.2018.0379] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/01/2019] [Indexed: 01/06/2023] Open
Abstract
A great variety of natural and synthetic polymer materials have been utilized in soft tissue engineering as extracellular matrix (ECM) materials. Natural polymers, such as collagen and fibrin hydrogels, have experienced especially broad adoption due to the high density of cell adhesion sites compared to their synthetic counterparts, ready availability, and ease of use. However, these and other hydrogels lack the structural and mechanical anisotropy that define the ECM in many tissues, such as skeletal and cardiac muscle, tendon, and cartilage. Herein, we present a facile, low-cost, and automated method of preparing collagen microfibers, organizing these fibers into precisely controlled mesh designs, and embedding these meshes in a bulk hydrogel, creating a composite biomaterial suitable for a wide variety of tissue engineering and regenerative medicine applications. With the assistance of custom software tools described herein, mesh patterns are designed by a digital graphical user interface and translated into protocols that are executed by a custom mesh collection and organization device. We demonstrate a high degree of precision and reproducibility in both fiber and mesh fabrication, evaluate single fiber mechanical properties, and provide evidence of collagen self-assembly in the microfibers under standard cell culture conditions. This work offers a powerful, flexible platform for the study of tissue engineering and cell material interactions, as well as the development of therapeutic biomaterials in the form of custom collagen microfiber patterns that will be accessible to all through the methods and techniques described here. Impact Statement Collagen microfiber meshes have immediate and broad applications in tissue engineering research and show high potential for later use in clinical therapeutics due to their compositional similarities to native extracellular matrix and tunable structural and mechanical characteristics. Physical and biological characterizations of these meshes demonstrate physiologically relevant mechanical properties, native-like collagen structure, and cytocompatibility. The methods presented herein not only describe a process through which custom collagen microfiber meshes can be fabricated but also provide the reader with detailed device plans and software tools to produce their own bespoke meshes through a precise, consistent, and automated process.
Collapse
Affiliation(s)
- Nicholas J Kaiser
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island
| | - Jessica A Bellows
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island
| | - Rajeev J Kant
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island
| | - Kareen L K Coulombe
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, Rhode Island
| |
Collapse
|
224
|
Johnson DL, Ziemba RM, Shebesta JH, Lipscomb JC, Wang Y, Wu Y, O’Connell KD, Kaltchev MG, van Groningen A, Chen J, Hua X, Zhang W. Design of pectin-based bioink containing bioactive agent-loaded microspheres for bioprinting. Biomed Phys Eng Express 2019. [DOI: 10.1088/2057-1976/ab4dbc] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
225
|
Lim KS, Baptista M, Moon S, Woodfield TB, Rnjak-Kovacina J. Microchannels in Development, Survival, and Vascularisation of Tissue Analogues for Regenerative Medicine. Trends Biotechnol 2019; 37:1189-1201. [DOI: 10.1016/j.tibtech.2019.04.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 11/26/2022]
|
226
|
Katarivas Levy G, Ong J, Birch MA, Justin AW, Markaki AE. Albumin-Enriched Fibrin Hydrogel Embedded in Active Ferromagnetic Networks Improves Osteoblast Differentiation and Vascular Self-Organisation. Polymers (Basel) 2019; 11:polym11111743. [PMID: 31652977 PMCID: PMC6918167 DOI: 10.3390/polym11111743] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/19/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022] Open
Abstract
Porous coatings on prosthetic implants encourage implant fixation. Enhanced fixation may be achieved using a magneto-active porous coating that can deform elastically in vivo on the application of an external magnetic field, straining in-growing bone. Such a coating, made of 444 ferritic stainless steel fibres, was previously characterised in terms of its mechanical and cellular responses. In this work, co-cultures of human osteoblasts and endothelial cells were seeded into a novel fibrin-based hydrogel embedded in a 444 ferritic stainless steel fibre network. Albumin was successfully incorporated into fibrin hydrogels improving the specific permeability and the diffusion of fluorescently tagged dextrans without affecting their Young’s modulus. The beneficial effect of albumin was demonstrated by the upregulation of osteogenic and angiogenic gene expression. Furthermore, mineralisation, extracellular matrix production, and formation of vessel-like structures were enhanced in albumin-enriched fibrin hydrogels compared to fibrin hydrogels. Collectively, the results indicate that the albumin-enriched fibrin hydrogel is a promising bio-matrix for bone tissue engineering and orthopaedic applications.
Collapse
Affiliation(s)
- Galit Katarivas Levy
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK.
| | - John Ong
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK.
| | - Mark A Birch
- Division of Trauma and Orthopaedic Surgery, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK.
| | - Alexander W Justin
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK.
| | - Athina E Markaki
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, UK.
| |
Collapse
|
227
|
Intermittent application of external positive pressure helps to preserve organ viability during ex vivo perfusion and culture. J Artif Organs 2019; 23:36-45. [PMID: 31617028 PMCID: PMC7046602 DOI: 10.1007/s10047-019-01141-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/05/2019] [Indexed: 12/03/2022]
Abstract
The perfusion of medium through blood vessels allows the preservation of donor organs and culture of bioengineered organs. However, tissue damage due to inadequate perfusion remains a problem. We evaluated whether intermittent external pressurization would improve the perfusion and viability of organs in culture. A bioreactor system was used to perfuse and culture rat small intestine and femoral muscle preparations. Intermittent positive external pressure (10 mmHg) was applied for 20 s at intervals of 20 s. Intermittent pressurization resulted in uniform perfusion of small intestine preparations and minimal tissue damage after 20 h of perfusion, whereas non-pressurized (control) preparations exhibited significantly worse perfusion of the upper surface than the lower surface and histologic evidence of tissue damage. Longer term studies were undertaken in luciferase-expressing rat femoral muscle preparations. Compared with non-pressurized controls, intermittent pressurization led to better perfusion throughout the 14-day experimental period, improved organ viability as indicated by a higher bioluminescence intensity after perfusion with luciferin, and reduced levels of tissue necrosis with better preservation of vascular structures and skeletal muscle nuclei (histologic analyses). Therefore, intermittent application of external positive pressure improved the perfusion of small intestine and skeletal muscle preparations and enhanced tissue viability when compared with controls. We anticipate that this innovative perfusion technique could be used to improve the preservation of donor organs and culture of bioengineered organs.
Collapse
|
228
|
Ji C, Bi L, Li J, Fan J. Salvianolic Acid B-Loaded Chitosan/hydroxyapatite Scaffolds Promotes The Repair Of Segmental Bone Defect By Angiogenesis And Osteogenesis. Int J Nanomedicine 2019; 14:8271-8284. [PMID: 31686820 PMCID: PMC6800558 DOI: 10.2147/ijn.s219105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022] Open
Abstract
Background Salvianolic acid B has been proven as an effective drug to promote osteogenesis and angiogenesis which could be beneficial for bone repair. Purpose The objective of this study was to construct a salvianolic acid B-loaded chitosan/hydroxyapatite (Sal B-CS/HA) bone scaffold with controlled release and effective bioactivity. Methods The characteristics, controlled release behavior and bioactivity of Sal B-CS/HA scaffold were evaluated in vitro. The bone repair effect was evaluated in the rabbit radius defect model. Results The results showed that chemical and physical characteristics of salvianolic acid B and chitosan/hydroxyapatite (CS/HA) material did not obviously change after the drug loading procedure; the drug release of salvianolic acid B was stable and continuous from the Sal B-CS/HA scaffold for 8 weeks in vitro; the biocompatibility of the Sal B-CS/HA was favorable by evaluation of cell morphology and proliferation; the osteogenic and angiogenic bioactivities of the Sal B-CS/HA scaffold were proved to be effective by in vivo and in vitro tests. Conclusion Our results suggest that this salvianolic acid B-loaded bone scaffold has potential to be used for bone defect repair with both osteogenic and angiogenic bioactivities.
Collapse
Affiliation(s)
- Chuanlei Ji
- Department of Orthopedics Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Long Bi
- Department of Orthopedics Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Jing Li
- Department of Orthopedics Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Junjun Fan
- Department of Orthopedics Surgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| |
Collapse
|
229
|
Tao C, Lina X, Changxuan W, Cong L, Xiaolan Y, Tao H, Hong A. Orthogonal test design for the optimization of superparamagnetic chitosan plasmid gelatin microspheres that promote vascularization of artificial bone. J Biomed Mater Res B Appl Biomater 2019; 108:1439-1449. [PMID: 31605570 PMCID: PMC7187448 DOI: 10.1002/jbm.b.34491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/06/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022]
Abstract
The optimal conditions for the preparation of superparamagnetic chitosan plasmid (pReceiver‐M29‐VEGF165/DH5a) gelatin microspheres (SPCPGMs) were determined. Then, the performance of the SPCPGMs during neovascularization was evaluated in vivo. The SPCPGMs were prepared through a cross‐linking curing method and then filled into the hollow scaffold of an artificial bone. Neovascularization at the bone defect position was histologically examined in samples collected 2, 4, 6, and 8 weeks after the operation. The cellular magnetofection rate of superparamagnetic chitosan nanoparticles/plasmid (pReceiver‐M29‐VEGF165/DH5a) complexes reached 1–3% under static magnetic field (SMF). Meanwhile, the optimal conditions for SPCPGM fabrication were 20% Fe3O4 (w/v), 4 mg of plasmid, 5.3 mg of glutaraldehyde, and 500 rpm of emulsification rotate speed. Under oscillating magnetic fields (OMFs), 4–6 μg of plasmids was released daily for 21 days. Under the combined application of SMF and OMF, evident neovascularization occurred at the bone defect position 6 weeks after the operation. This result is expected to provide a new type of angiogenesis strategy for the research of bone tissue engineering.
Collapse
Affiliation(s)
- Chen Tao
- Department of Orthopaedics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy
| | - Xie Lina
- Department of Orthopaedics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy
| | - Wang Changxuan
- Department of Orthopaedics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy
| | - Luo Cong
- Department of Orthopaedics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy
| | - Yang Xiaolan
- Department of Pharmacology, Chongqing Medical University, Yuzhong District, Yixueyuan Road1#, Chongqing, 400016, China
| | - Huang Tao
- Department of Orthopaedics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy
| | - An Hong
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Youyi Road 1#, Chongqing, 400016, China
| |
Collapse
|
230
|
Zambuto SG, Clancy KBH, Harley BAC. A gelatin hydrogel to study endometrial angiogenesis and trophoblast invasion. Interface Focus 2019; 9:20190016. [PMID: 31485309 PMCID: PMC6710659 DOI: 10.1098/rsfs.2019.0016] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
The endometrium is the lining of the uterus and site of blastocyst implantation. Each menstrual cycle, the endometrium cycles through rapid phases of growth, remodelling and breakdown. Significant vascular remodelling is also driven by trophoblast cells that form the outer layer of the blastocyst. Trophoblast invasion and remodelling enhance blood flow to the embryo ahead of placentation. Understanding the mechanisms of endometrial vascular remodelling and trophoblast invasion would provide key insights into endometrial physiology and cellular interactions critical for establishment of pregnancy. The objective of this study was to develop a tissue engineering platform to investigate the processes of endometrial angiogenesis and trophoblast invasion in a three-dimensional environment. We report adaptation of a methacrylamide-functionalized gelatin hydrogel that presents matrix stiffness in the range of the native tissue, supports the formation of endometrial endothelial cell networks with human umbilical vein endothelial cells and human endometrial stromal cells as an artificial endometrial perivascular niche and the culture of an endometrial epithelial cell layer, enables culture of a hormone-responsive stromal compartment and provides the capacity to monitor the kinetics of trophoblast invasion. With these studies, we provide a series of techniques that will instruct researchers in the development of endometrial models of increasing complexity.
Collapse
Affiliation(s)
- Samantha G. Zambuto
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kathryn B. H. Clancy
- Department of Anthropology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
231
|
Alghuwainem A, Alshareeda AT, Alsowayan B. Scaffold-Free 3-D Cell Sheet Technique Bridges the Gap between 2-D Cell Culture and Animal Models. Int J Mol Sci 2019; 20:E4926. [PMID: 31590325 PMCID: PMC6801996 DOI: 10.3390/ijms20194926] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Various tissue engineering techniques have been created in research spanning two centuries, resulting in new opportunities for growing cells in culture and the creation of 3-D tissue-like constructs. These techniques are classified as scaffold-based and scaffold-free techniques. Cell sheet, as a scaffold-free technique, has attracted research interest in the context of drug discovery and tissue repair, because it provides more predictive data for in vivo testing. It is one of the most promising techniques and has the potential to treat degenerative tissues such as heart, kidneys, and liver. In this paper, we argue the advantages of cell sheets as a scaffold-free approach, compared to other techniques, including scaffold-based and scaffold-free techniques such as the classic systemic injection of cell suspension.
Collapse
Affiliation(s)
- Ayidah Alghuwainem
- Stem Cell & Regenerative Medicine Unit, Cellular Therapy and Cancer Research Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia.
| | - Alaa T Alshareeda
- Stem Cell & Regenerative Medicine Unit, Cellular Therapy and Cancer Research Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia.
| | - Batla Alsowayan
- Stem Cell & Regenerative Medicine Unit, Cellular Therapy and Cancer Research Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia.
| |
Collapse
|
232
|
|
233
|
Aleman J, George SK, Herberg S, Devarasetty M, Porada CD, Skardal A, Almeida-Porada G. Deconstructed Microfluidic Bone Marrow On-A-Chip to Study Normal and Malignant Hemopoietic Cell-Niche Interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902971. [PMID: 31464364 PMCID: PMC8011350 DOI: 10.1002/smll.201902971] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/31/2019] [Indexed: 05/23/2023]
Abstract
Human hematopoietic niches are complex specialized microenvironments that maintain and regulate hematopoietic stem and progenitor cells (HSPC). Thus far, most of the studies performed investigating alterations of HSPC-niche dynamic interactions are conducted in animal models. Herein, organ microengineering with microfluidics is combined to develop a human bone marrow (BM)-on-a-chip with an integrated recirculating perfusion system that consolidates a variety of important parameters such as 3D architecture, cell-cell/cell-matrix interactions, and circulation, allowing a better mimicry of in vivo conditions. The complex BM environment is deconvoluted to 4 major distinct, but integrated, tissue-engineered 3D niche constructs housed within a single, closed, recirculating microfluidic device system, and equipped with cell tracking technology. It is shown that this technology successfully enables the identification and quantification of preferential interactions-homing and retention-of circulating normal and malignant HSPC with distinct niches.
Collapse
Affiliation(s)
- Julio Aleman
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Sunil K George
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Samuel Herberg
- Department of Opthamology, State University of New York Upstate Medical University, 4609 Institute for Human Performance, Syracuse, NY, 13210, USA
| | - Mahesh Devarasetty
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| |
Collapse
|
234
|
Salgado CL, Teixeira BIB, Monteiro FJM. Biomimetic Composite Scaffold With Phosphoserine Signaling for Bone Tissue Engineering Application. Front Bioeng Biotechnol 2019; 7:206. [PMID: 31552233 PMCID: PMC6743420 DOI: 10.3389/fbioe.2019.00206] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/12/2019] [Indexed: 01/05/2023] Open
Abstract
In guided bone tissue engineering, successful ingrowth of MSCs depends primarily on the nature of the scaffold. It is well-known that only seconds after implantation, biomaterials are coated by a layer of adsorbed proteins/peptides which modulates the subsequent cell/scaffold interactions, especially at early times after implantation. In this work, nanohydroxyapatite and collagen based composite materials (Coll/nanoHA) were modified with phosphorylated amino acid (O-phospho-L-serine-OPS) to mimic bone tissue, and induce cell differentiation. The choice for this phosphorylated amino acid is due to the fact that osteopontin is a serine-rich glycol-phosphoprotein and has been associated to the early stages of bone formation, and regeneration. Several concentrations of OPS were added to the Coll/nanoHA scaffold and physico-chemical, mechanical, and in vitro cell behavior were evaluated. Afterwards, the composite scaffold with stronger mechanical and best cellular behavior was tested in vivo, with or without previous in vitro culture of human MSC's (bone tissue engineering). The OPS signaling of the biocomposite scaffolds showed similar cellular adhesion and proliferation, but higher ALP enzyme activity (HBMSC). In vivo bone ectopic formation studies allowed for a thorough evaluation of the materials for MSC's osteogenic differentiation. The OPS-scaffolds results showed that the material could modulated mesenchymal cells behavior in favor of osteogenic differentiation into late osteoblasts that gave raised to their ECM with human bone proteins (osteopontin) and calcium deposits. Finally, OPS-modified scaffolds enhanced cell survival, engraftment, migration, and spatial distribution within the 3D matrix that could be used as a cell-loaded scaffold for tissue engineering applications and accelerate bone regeneration processes.
Collapse
Affiliation(s)
- Christiane Laranjo Salgado
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB–Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia (FEUP), DEMM, Universidade do Porto, Porto, Portugal
| | - Beatriz Isabel Brites Teixeira
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB–Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Institute of Health Sciences (ICS), Universidade Católica Portuguesa, Viseu, Portugal
| | - Fernando Jorge Mendes Monteiro
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB–Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia (FEUP), DEMM, Universidade do Porto, Porto, Portugal
| |
Collapse
|
235
|
Pang Y, Sutoko S, Horimoto Y, Weng D, Montagne K, Komori K, Takano K, Shirakashi R, Anzai M, Niino T, Sakai Y. Biodegradable and hollowed micro-scaffolds for improved modular assembly-based tissue engineering: Design, 3D fabrication, and feasibility in randomly packed perfusion culture. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.107239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
236
|
Patel DB, Luthers CR, Lerman MJ, Fisher JP, Jay SM. Enhanced extracellular vesicle production and ethanol-mediated vascularization bioactivity via a 3D-printed scaffold-perfusion bioreactor system. Acta Biomater 2019; 95:236-244. [PMID: 30471476 DOI: 10.1016/j.actbio.2018.11.024] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/12/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (EVs) have garnered significant interest in the biotechnology field due to their intrinsic therapeutic properties as well as their ability to serve as vehicles for bioactive cargo. However, the lack of an established biomanufacturing platform and limited potency of EVs in vivo remain critical bottlenecks for clinical translation. In this study, we utilized a 3D-printed scaffold-perfusion bioreactor system to assess the response of dynamic culture on extracellular vesicle production from endothelial cells (ECs). We also investigated whether ethanol conditioning, which was previously shown to enhance vascularization bioactivity of EC-derived EVs produced in standard 2D culture conditions, could be employed successfully for the same purpose in a 3D production system. Our results indicate that dynamic culture in a perfusion bioreactor significantly enhances EV production from human ECs. Moreover, the use of ethanol conditioning in conjunction with dynamic culture induces pro-vascularization bioactivity of EC-derived EVs that is correlated with increased EV levels of pro-angiogenic lncRNAs HOTAIR and MALAT1. Thus, this study represents one of the first reports of rationally-designed EV potency enhancement that is conserved between static 2D and dynamic 3D EV production systems, increasing the potential for scalable biomanufacturing of therapeutic EC-derived EVs for a variety of applications. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) have substantial therapeutic potential in a variety of applications. However, translation of EV-based therapies may be hindered by biomanufacturing challenges. EV production to date has predominantly involved the use of tissue culture flasks. Here, we report, for the first time, the use of a tubular perfusion bioreactor system with an integrated 3D-printed biomaterial scaffold for EV production from human endothelial cells. This system increases EV yield by over 100-fold compared to conventional tissue culture systems. Further, we show that an ethanol-conditioning approach that our group previously developed in 2D culture for enhancing EV potency is compatible with this new system. Thus, potency enhancement of EVs for vascularization applications is possible even with significantly increased production rate.
Collapse
Affiliation(s)
- Divya B Patel
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States
| | - Christopher R Luthers
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States
| | - Max J Lerman
- Department of Materials Science and Engineering, University of Maryland 4418 Stadium Drive, College Park, MD 20742, United States; Center for Engineering Complex Tissues, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States; Surface and Trace Chemical Analysis Group, Materials Measurement Science Division National Institute of Standards and Technology 100 Bureau Drive, Mailstop 6431, Gaithersburg, MD 20899, United States
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States; Center for Engineering Complex Tissues, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland 3102 A. James Clark Hall, College Park, MD 20742, United States; Greenbaum Comprehensive Cancer Center, University of Maryland - Baltimore, Baltimore, MD 21201, United States; Program in Molecular and Cell Biology, University of Maryland College Park, MD 20742, United States.
| |
Collapse
|
237
|
Zhang FM, Zhou L, Zhou ZN, Dai C, Fan L, Li CH, Xiao CR, Ning CY, Liu Y, Du JQ, Tan GX. Bioactive glass functionalized chondroitin sulfate hydrogel with proangiogenic properties. Biopolymers 2019; 110:e23328. [PMID: 31454076 DOI: 10.1002/bip.23328] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/10/2019] [Accepted: 08/13/2019] [Indexed: 12/23/2022]
Abstract
Blood vessels play an important role in bone defect repair and growth, and a critical challenge of bone defect repair is the promotion of blood vessel formation. Most of the current methods promote vascularization by adding specific growth factors, which are costly and easy to inactivate. In this study, we developed a covalently cross-linked aminated bioactive glass nanoparticle-chondroitin sulfate methacrylate (ABGN-CSMA) organic-inorganic composite hydrogel with angiogenic properties. The amino groups of the ABGNs form covalent bonds with the carboxyl groups on CSMA. Surface amination modification of BGNs not only improved the dispersion of BGNs in CSMA but also significantly improved the mechanical properties of the composite hydrogel. The largest storage modulus (1200 Pa), the largest loss modulus (560 Pa) and the strongest resistance to deformation of the hydrogel are seen at 10% concentration of ABGNs. Simultaneously, the local pH stability and sustained ion release of the composite hydrogel are conducive to cell adhesion, proliferation, and angiogenesis. This work provides evidence for the development of covalently cross-linked organic-inorganic composite hydrogels with angiogenic properties.
Collapse
Affiliation(s)
- Feng-Miao Zhang
- Department of Applied Chemistry, Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, China
| | - Lei Zhou
- Department of Applied Chemistry, Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, China.,School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
| | - Zheng-Nan Zhou
- Department of Applied Chemistry, Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, China
| | - Cong Dai
- Department of Applied Chemistry, Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, China
| | - Lei Fan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
| | - Chang-Hao Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
| | - Cai-Rong Xiao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
| | - Cheng-Yun Ning
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
| | - Yi Liu
- Orthopedics Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jian-Qiang Du
- Department of Nuclear Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guo-Xin Tan
- Department of Applied Chemistry, Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, China
| |
Collapse
|
238
|
Tomasina C, Bodet T, Mota C, Moroni L, Camarero-Espinosa S. Bioprinting Vasculature: Materials, Cells and Emergent Techniques. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2701. [PMID: 31450791 PMCID: PMC6747573 DOI: 10.3390/ma12172701] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022]
Abstract
Despite the great advances that the tissue engineering field has experienced over the last two decades, the amount of in vitro engineered tissues that have reached a stage of clinical trial is limited. While many challenges are still to be overcome, the lack of vascularization represents a major milestone if tissues bigger than approximately 200 µm are to be transplanted. Cell survival and homeostasis is to a large extent conditioned by the oxygen and nutrient transport (as well as waste removal) by blood vessels on their proximity and spontaneous vascularization in vivo is a relatively slow process, leading all together to necrosis of implanted tissues. Thus, in vitro vascularization appears to be a requirement for the advancement of the field. One of the main approaches to this end is the formation of vascular templates that will develop in vitro together with the targeted engineered tissue. Bioprinting, a fast and reliable method for the deposition of cells and materials on a precise manner, appears as an excellent fabrication technique. In this review, we provide a comprehensive background to the fields of vascularization and bioprinting, providing details on the current strategies, cell sources, materials and outcomes of these studies.
Collapse
Affiliation(s)
- Clarissa Tomasina
- MERLN Institute for Technology-inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands
| | - Tristan Bodet
- MERLN Institute for Technology-inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands
| | - Carlos Mota
- MERLN Institute for Technology-inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands
| | - Lorenzo Moroni
- MERLN Institute for Technology-inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands.
| | - Sandra Camarero-Espinosa
- MERLN Institute for Technology-inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands.
| |
Collapse
|
239
|
A facile approach for engineering tissue constructs with vessel-like channels by cell-laden hydrogel fibers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:370-379. [DOI: 10.1016/j.msec.2019.03.094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/20/2019] [Accepted: 03/25/2019] [Indexed: 01/21/2023]
|
240
|
Agarwal T, Maiti TK, Behera B, Ghosh SK, Apoorva A, Padmavati M. Biofunctionalized cellulose paper matrix for cell delivery applications. Int J Biol Macromol 2019; 139:114-127. [PMID: 31374267 DOI: 10.1016/j.ijbiomac.2019.07.201] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023]
Abstract
The present study delineates the preparation, characterization, and application of (3-Aminopropyl)triethoxysilane (APTES)/Caprine liver-derived extracellular matrix (CLECM) coated paper matrix for cell delivery. Here, we exploited positively charged surface of the paper matrix (as imparted by APTES derivatization) to improve the biological responses of the cells. Our results demonstrated that the functionalized paper matrixes favored the adhesion, growth, and proliferation of multiple cell types including normal, transformed, cancerous, and stem cells as compared to the pristine paper matrix. Upon implantation into the mice model, the developed paper matrix supported infiltration of the host cells and vasculature without showing any evidence of significant systemic toxicity. Moreover, the cells cultured on the paper matrix, when delivered to the CAM and mouse models, showed an enhanced vascular network around the substrate, thereby confirming its potential to deliver the cells in vivo. Together, the study confirms that the reported paper-based platform is easy to fabricate, cheap, portable and could efficiently be applied to cell delivery applications for either tissue repair or the development of humanized animal model.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India.
| | - Birendra Behera
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India; Department of Biotechnology and Bioinformatics, Sambalpur University, Odisha 768019, India.
| | - Sudip Kumar Ghosh
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India.
| | - Anupam Apoorva
- School of Biosciences, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Manchikanti Padmavati
- Rajiv Gandhi School of Intellectual Property Law, Indian Institute of Technology, Kharagpur, West Bengal 721302, India.
| |
Collapse
|
241
|
Schmid J, Schwarz S, Meier-Staude R, Sudhop S, Clausen-Schaumann H, Schieker M, Huber R. A Perfusion Bioreactor System for Cell Seeding and Oxygen-Controlled Cultivation of Three-Dimensional Cell Cultures. Tissue Eng Part C Methods 2019; 24:585-595. [PMID: 30234443 PMCID: PMC6208160 DOI: 10.1089/ten.tec.2018.0204] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Bioreactor systems facilitate three-dimensional (3D) cell culture by coping with limitations of static cultivation techniques. To allow for the investigation of proper cultivation conditions and the reproducible generation of tissue-engineered grafts, a bioreactor system, which comprises the control of crucial cultivation parameters in independent-operating parallel bioreactors, is beneficial. Furthermore, the use of a bioreactor as an automated cell seeding tool enables even cell distributions on stable scaffolds. In this study, we developed a perfusion microbioreactor system, which enables the cultivation of 3D cell cultures in an oxygen-controlled environment in up to four independent-operating bioreactors. Therefore, perfusion microbioreactors were designed with the help of computer-aided design, and manufactured using the 3D printing technologies stereolithography and fused deposition modeling. A uniform flow distribution in the microbioreactor was shown using a computational fluid dynamics model. For oxygen measurements, microsensors were integrated in the bioreactors to measure the oxygen concentration (OC) in the geometric center of the 3D cell cultures. To control the OC in each bioreactor independently, an automated feedback loop was developed, which adjusts the perfusion velocity according to the oxygen sensor signal. Furthermore, an automated cell seeding protocol was implemented to facilitate the even distribution of cells within a stable scaffold in a reproducible way. As proof of concept, the human mesenchymal stem cell line SCP-1 was seeded on bovine cancellous bone matrix of 1 cm3 and cultivated in the developed microbioreactor system at different oxygen levels. The oxygen control was capable to maintain preset oxygen levels ±0.5% over a cultivation period of several days. Using the automated cell seeding procedure resulted in evenly distributed cells within a stable scaffold. In summary, the developed microbioreactor system enables the cultivation of 3D cell cultures in an automated and thus reproducible way by providing up to four independently operating, oxygen-controlled bioreactors. In combination with the automated cell seeding procedure, the bioreactor system opens up new possibilities to conduct more reproducible experiments to investigate optimal cultivation parameters and to generate tissue-engineering grafts in an oxygen-controlled environment.
Collapse
Affiliation(s)
- Jakob Schmid
- 1 Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences Munich , Munich, Germany .,2 Laboratory of Experimental Surgery and Regenerative Medicine (ExperiMed), Ludwig-Maximilians University Munich , Munich, Germany .,3 Department of Industrial Engineering and Management, University of Applied Sciences Munich , Munich, Germany
| | - Sascha Schwarz
- 1 Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences Munich , Munich, Germany .,4 Department of Mechanical Engineering, Technical University Munich , Garching, Germany
| | - Robert Meier-Staude
- 3 Department of Industrial Engineering and Management, University of Applied Sciences Munich , Munich, Germany
| | - Stefanie Sudhop
- 1 Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences Munich , Munich, Germany .,5 Center for Nanoscience (CeNS), Ludwig-Maximilians University Munich , Munich, Germany
| | - Hauke Clausen-Schaumann
- 1 Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences Munich , Munich, Germany .,5 Center for Nanoscience (CeNS), Ludwig-Maximilians University Munich , Munich, Germany
| | - Matthias Schieker
- 2 Laboratory of Experimental Surgery and Regenerative Medicine (ExperiMed), Ludwig-Maximilians University Munich , Munich, Germany
| | - Robert Huber
- 1 Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences Munich , Munich, Germany .,3 Department of Industrial Engineering and Management, University of Applied Sciences Munich , Munich, Germany
| |
Collapse
|
242
|
Sämfors S, Karlsson K, Sundberg J, Markstedt K, Gatenholm P. Biofabrication of bacterial nanocellulose scaffolds with complex vascular structure. Biofabrication 2019; 11:045010. [DOI: 10.1088/1758-5090/ab2b4f] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
243
|
Deegan AJ, Hendrikson WJ, El Haj AJ, Rouwkema J, Yang Y. Regulation of endothelial cell arrangements within hMSC - HUVEC co-cultured aggregates. Biomed J 2019; 42:166-177. [PMID: 31466710 PMCID: PMC6717755 DOI: 10.1016/j.bj.2019.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/25/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background Micro-mass culturing or cellular aggregation is an effective method used to form mineralised bone tissue. Poor core cell viability, however, is often an impeding characteristic of large micro-mass cultures, and equally for large tissue-engineered bone grafts. Because of this, efforts are being made to enhance large graft perfusion, often through pre-vascularisation, which involves the co-culture of endothelial cells and bone cells or stem cells. Methods This study investigated the effects of different aggregation techniques and culture conditions on endothelial cell arrangements in mesenchymal stem cell and human umbilical vein endothelial cell co-cultured aggregates when endothelial cells constituted just 5%. Two different cellular aggregation techniques, i.e. suspension culture aggregation and pellet culture aggregation, were applied alongside two subsequent culturing techniques, i.e. hydrostatic loading and static culturing. Endothelial cell arrangements were assessed under such conditions to indicate potential pre-vascularisation. Results Our study found that the suspension culture aggregates cultured under hydrostatic loading offered the best environment for enhanced endothelial cell regional arrangements, closely followed by the pellet culture aggregates cultured under hydrostatic loading, the suspension culture aggregates cultured under static conditions, and the pellet culture aggregates cultured under static conditions. Conclusions The combination of particular aggregation techniques with dynamic culturing conditions appeared to have a synergistic effect on the cellular arrangements within the co-cultured aggregates.
Collapse
Affiliation(s)
- Anthony J Deegan
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Wim J Hendrikson
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, AE, the Netherlands
| | - Alicia J El Haj
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Ying Yang
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom.
| |
Collapse
|
244
|
Fetah K, Tebon P, Goudie MJ, Eichenbaum J, Ren L, Barros N, Nasiri R, Ahadian S, Ashammakhi N, Dokmeci MR, Khademhosseini A. The emergence of 3D bioprinting in organ-on-chip systems. ACTA ACUST UNITED AC 2019. [DOI: 10.1088/2516-1091/ab23df] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
245
|
Nakamura H, Koziolová E, Chytil P, Etrych T, Haratake M, Maeda H. Superior Penetration and Cytotoxicity of HPMA Copolymer Conjugates of Pirarubicin in Tumor Cell Spheroid. Mol Pharm 2019; 16:3452-3459. [PMID: 31294568 DOI: 10.1021/acs.molpharmaceut.9b00248] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
N-(2-Hydroxypropyl)methacrylamide copolymer conjugates of pirarubicin (THP), P-THP, accumulates selectively in solid tumor tissue by the enhanced permeability and retention (EPR) effect. Despite of high accumulation in solid tumors, some macromolecular antitumor agents show poor therapeutic outcome because of poor tissue diffusion into the tumor as well as obstructed tumor blood flow. Here, we confirmed that cellular uptake of P-THP was 25 times less than that of free THP at 1-4 h incubation time in vitro. The passage of P-THP through the confluent tight-monolayer cells junction was 12 times higher than free THP, and P-THP penetrated deeper into the tumor cell spheroid (1.3-1.7-fold) than free THP in 4 h. In addition, P-THP showed cytotoxicity comparable to that of free THP to tumor-cells in spheroid form, despite of 7 times lower cytotoxicity of P-THP to the monolayer cells to that of free THP in vitro. These results indicate that P-THP administration can exhibit deeper diffusion into the tumor cell spheroid than free THP. As a consequence, P-THP exhibits more efficient antitumor activity than free THP in vivo, which is also supported by better pharmacokinetics and tumor accumulation of P-THP than free THP.
Collapse
Affiliation(s)
- Hideaki Nakamura
- Faculty of Pharmaceutical Sciences , Sojo University , Ikeda 4-22-1, Nishi-ku , Kumamoto 860-0082 , Japan
| | - Eva Koziolová
- Institute of Macromolecular Chemistry , Czech Academy of Sciences , Heyrovsky Sq. 2 , 162 06 Prague 6 , Czech Republic
| | - Petr Chytil
- Institute of Macromolecular Chemistry , Czech Academy of Sciences , Heyrovsky Sq. 2 , 162 06 Prague 6 , Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry , Czech Academy of Sciences , Heyrovsky Sq. 2 , 162 06 Prague 6 , Czech Republic
| | - Mamoru Haratake
- Faculty of Pharmaceutical Sciences , Sojo University , Ikeda 4-22-1, Nishi-ku , Kumamoto 860-0082 , Japan
| | - Hiroshi Maeda
- Department of Microbiology , Kumamoto University School of Medical Sciences , Kumamoto , Japan.,BioDynamics Research Foundation , Kuwamizu 1-24-6, Chuo-ku , Kumamoto 862-0954 , Japan
| |
Collapse
|
246
|
Loureiro J, Torres AL, Neto T, Aguiar P, Barrias CC, Pinto MT, Amaral IF. Conjugation of the T1 sequence from CCN1 to fibrin hydrogels for therapeutic vascularization. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109847. [PMID: 31500049 DOI: 10.1016/j.msec.2019.109847] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/13/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022]
Abstract
Hydrogel matrices with angiogenic properties are much desirable for therapeutic vascularization strategies, namely to provide vascular supply to ischemic areas, transplanted cells, or bioengineered tissues. Here we report the pro-angiogenic effect of fibrin (Fb) functionalization with the T1 sequence from the angiogenic inducer CCN1, forseeing its use in the injured brain and spinal cord. Fibrin functionalization with 40 μM of T1 peptide effectively improved cellular sprouting of human brain microvascular endothelial cells (hCMEC/D3) in the absence of vascular endothelial growth factor (VEGF), without impacting the viscoelastic properties of Fb, cell viability, or proliferation. The pro-angiogenic effect of immobilized T1 was potentiated in the presence of VEGF and partially mediated through α6β1 integrin. The tethering of T1 also enhanced sprouting of human cord blood-derived outgrowth endothelial cells (OEC). Still, to elicit such effect, a higher input T1 concentration was required (60 μM), in line with the lower protein levels of α6 and β1 integrin subunits found in OEC comparing to hCMEC/D3, prior to embedment in Fb gel. Finally, the ability of T1-functionalized Fb in inducing cappilary invasion in vivo was assessed using the CAM assay, which evidenced a significant increase in the number of newly formed vessels at sites of implantation of T1-functionalized Fb, in the absence of soluble angiogenic factors. Overall these results demonstrate the potential of T1 peptide-presenting gels for use in therapeutic vascularization approaches. Considering T1 neurite-extension promoting capability and pro-angiogenic properties, T1-functionalized Fb hydrogels are particularly promising for application in the injured central nervous system.
Collapse
Affiliation(s)
- Joana Loureiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto (UPorto), Portugal; INEB - Instituto de Engenharia Biomédica, UPorto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, UPorto, Portugal; FEUP - Faculdade de Engenharia, UPorto, Portugal
| | - Ana Luísa Torres
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto (UPorto), Portugal; INEB - Instituto de Engenharia Biomédica, UPorto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, UPorto, Portugal
| | - Tânia Neto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto (UPorto), Portugal; INEB - Instituto de Engenharia Biomédica, UPorto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, UPorto, Portugal; FEUP - Faculdade de Engenharia, UPorto, Portugal
| | - Paulo Aguiar
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto (UPorto), Portugal; INEB - Instituto de Engenharia Biomédica, UPorto, Portugal
| | - Cristina Carvalho Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto (UPorto), Portugal; INEB - Instituto de Engenharia Biomédica, UPorto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, UPorto, Portugal
| | - Marta Teixeira Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto (UPorto), Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology, UPorto, Portugal
| | - Isabel Freitas Amaral
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto (UPorto), Portugal; INEB - Instituto de Engenharia Biomédica, UPorto, Portugal; FEUP - Faculdade de Engenharia, UPorto, Portugal.
| |
Collapse
|
247
|
Ashammakhi N, Darabi MA, Kehr NS, Erdem A, Hu SK, Dokmeci MR, Nasr AS, Khademhosseini A. Advances in Controlled Oxygen Generating Biomaterials for Tissue Engineering and Regenerative Therapy. Biomacromolecules 2019; 21:56-72. [DOI: 10.1021/acs.biomac.9b00546] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
| | - Mohammad Ali Darabi
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
| | - Nermin Seda Kehr
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Physikalisches Institut
and Center for Soft Nanoscience, Westfälische Wilhelms-Universität Münster, Busse-Peus-Strasse 10, 48149 Münster, Germany
| | - Ahmet Erdem
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry, Kocaeli University, Umuttepe Campus, 41380 Kocaeli, Turkey
- Department of Biomedical Engineering, Kocaeli University, Umuttepe Campus, 41380 Kocaeli, Turkey
| | - Shu-kai Hu
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Physikalisches Institut
and Center for Soft Nanoscience, Westfälische Wilhelms-Universität Münster, Busse-Peus-Strasse 10, 48149 Münster, Germany
| | - Mehmet R. Dokmeci
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
| | - Ali S. Nasr
- Division of Cardiothoracic Surgery, Department of Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
| | - Ali Khademhosseini
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Chemical Engineering, University of California−Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
248
|
Kaur A, Midha S, Giri S, Mohanty S. Functional Skin Grafts: Where Biomaterials Meet Stem Cells. Stem Cells Int 2019; 2019:1286054. [PMID: 31354835 PMCID: PMC6636521 DOI: 10.1155/2019/1286054] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/21/2019] [Indexed: 12/22/2022] Open
Abstract
Skin tissue engineering has attained several clinical milestones making remarkable progress over the past decades. Skin is inhabited by a plethora of cells spatiotemporally arranged in a 3-dimensional (3D) matrix, creating a complex microenvironment of cell-matrix interactions. This complexity makes it difficult to mimic the native skin structure using conventional tissue engineering approaches. With the advent of newer fabrication strategies, the field is evolving rapidly. However, there is still a long way before an artificial skin substitute can fully mimic the functions and anatomical hierarchy of native human skin. The current focus of skin tissue engineers is primarily to develop a 3D construct that maintains the functionality of cultured cells in a guided manner over a period of time. While several natural and synthetic biopolymers have been translated, only partial clinical success is attained so far. Key challenges include the hierarchical complexity of skin anatomy; compositional mismatch in terms of material properties (stiffness, roughness, wettability) and degradation rate; biological complications like varied cell numbers, cell types, matrix gradients in each layer, varied immune responses, and varied methods of fabrication. In addition, with newer biomaterials being adopted for fabricating patient-specific skin substitutes, issues related to escalating processing costs, scalability, and stability of the constructs under in vivo conditions have raised some concerns. This review provides an overview of the field of skin regenerative medicine, existing clinical therapies, and limitations of the current techniques. We have further elaborated on the upcoming tissue engineering strategies that may serve as promising alternatives for generating functional skin substitutes, the pros and cons associated with each technique, and scope of their translational potential in the treatment of chronic skin ailments.
Collapse
Affiliation(s)
- Amtoj Kaur
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Swati Midha
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Shibashish Giri
- Department of Cell Techniques and Applied Stem Cell Biology, Centre for Biotechnology and Biomedicine, University of Leipzig, Deutscher Platz 5, D-04103 Leipzig, Germany
- Department of Plastic Surgery and Hand Surgery, University Hospital Rechts der Isar, Technische Universität München, Munich, Germany
| | - Sujata Mohanty
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
249
|
Min S, Ko IK, Yoo JJ. State-of-the-Art Strategies for the Vascularization of Three-Dimensional Engineered Organs. Vasc Specialist Int 2019; 35:77-89. [PMID: 31297357 PMCID: PMC6609020 DOI: 10.5758/vsi.2019.35.2.77] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023] Open
Abstract
Engineering three-dimensional (3D) implantable tissue constructs is a promising strategy for replacing damaged or diseased tissues and organs with functional replacements. However, the efficient vascularization of new 3D organs is a major scientific and technical challenge since large tissue constructs or organs require a constant blood supply to survive in vivo. Current approaches to solving this problem generally fall into the following three major categories: (a) cell-based, (b) angiogenic factor-based, and (c) scaffold-based. In this review, we summarize state-of-the-art technologies that are used to develop complex, stable, and functional vasculature for engineered 3D tissue constructs and organs; additionally, we have suggested directions for future research.
Collapse
Affiliation(s)
- Sangil Min
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
250
|
Hornung R, Grünberger A, Westerwalbesloh C, Kohlheyer D, Gompper G, Elgeti J. Quantitative modelling of nutrient-limited growth of bacterial colonies in microfluidic cultivation. J R Soc Interface 2019; 15:rsif.2017.0713. [PMID: 29445038 DOI: 10.1098/rsif.2017.0713] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/22/2018] [Indexed: 11/12/2022] Open
Abstract
Nutrient gradients and limitations play a pivotal role in the life of all microbes, both in their natural habitat as well as in artificial, microfluidic systems. Spatial concentration gradients of nutrients in densely packed cell configurations may locally affect the bacterial growth leading to heterogeneous micropopulations. A detailed understanding and quantitative modelling of cellular behaviour under nutrient limitations is thus highly desirable. We use microfluidic cultivations to investigate growth and microbial behaviour of the model organism Corynebacterium glutamicum under well-controlled conditions. With a reaction-diffusion-type model, parameters are extracted from steady-state experiments with a one-dimensional nutrient gradient. Subsequently, we employ particle-based simulations with these parameters to predict the dynamical growth of a colony in two dimensions. Comparing the results of those simulations with microfluidic experiments yields excellent agreement. Our modelling approach lays the foundation for a better understanding of dynamic microbial growth processes, both in nature and in applied biotechnology.
Collapse
Affiliation(s)
- Raphael Hornung
- Theoretical Soft Matter and Biophysics, Institute of Complex Systems and Institute for Advanced Simulation, Forschungszentrum Jülich and JARA, 52425 Jülich, Germany
| | - Alexander Grünberger
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany.,Multiscale Bioengineering, Bielefeld University, Universitätsstrasse 25, Bielefeld 33615, Germany
| | - Christoph Westerwalbesloh
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Dietrich Kohlheyer
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, 52425 Jülich, Germany.,Aachener Verfahrenstechnik (AVT.MSB), RWTH Aachen University, 52056 Aachen, Germany
| | - Gerhard Gompper
- Theoretical Soft Matter and Biophysics, Institute of Complex Systems and Institute for Advanced Simulation, Forschungszentrum Jülich and JARA, 52425 Jülich, Germany
| | - Jens Elgeti
- Theoretical Soft Matter and Biophysics, Institute of Complex Systems and Institute for Advanced Simulation, Forschungszentrum Jülich and JARA, 52425 Jülich, Germany
| |
Collapse
|