1
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
2
|
Li Z, Xu P, Shang L, Ma B, Zhang H, Fu L, Ou Y, Mao Y. 3D collagen porous scaffold carrying PLGA-PTX/SDF-1α recruits and promotes neural stem cell differentiation for spinal cord injury repair. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2332-2355. [PMID: 37566099 DOI: 10.1080/09205063.2023.2247715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
Spinal Cord Injury (SCI), one of the major factors of disability, can cause irreversible motor and sensory impairment. There are no effective therapeutic drugs and technologies available in domestic or foreign countries currently. Neural stem cells (NSCs), with the potential for multidirectional differentiation, are a potential treatment for SCI. However, it has been demonstrated that NSCs primarily differentiated into astrocytes rather than neurons due to the inflammatory microenvironment, and the current challenge remains to direct the differentiation of NSCs into neurons in the lesion site. It was reported that the microtubule-stabilizing agent paclitaxel (PTX) was able to promote the differentiation of NSCs into neurons rather than astrocytes after SCI. SDF-1α can recruit NSCs and thus guide the migration of stem cells. In this study, we developed a functional collagen scaffold by loading SDF-1α and nanoparticle-encapsulated PLGA-PTX into a 3D collagen porous scaffold, allowing for slow release of PTX. When the functional scaffolds were implanted into the injury site, it provided a neural regeneration conduit channel for the migration of NSCs and neuronal differentiation. Neural regeneration promoted the recovery of motor function and reduced glial scar formation after SCI. In conclusion, a 3D collagen porous scaffold combined with PLGA-PTX and SDF-1α is a promising therapeutic strategy for SCI repair.
Collapse
Affiliation(s)
- Zhixiang Li
- School of Life Sciences, Bengbu Medical College, Bengbu, China
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Panpan Xu
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Lijun Shang
- School of Life Sciences, Bengbu Medical College, Bengbu, China
| | - Bingxu Ma
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Huihui Zhang
- Department of Oncology, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Liangmin Fu
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yuanyuan Ou
- School of Life Sciences, Bengbu Medical College, Bengbu, China
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| |
Collapse
|
3
|
Eckrich J, Hoormann N, Kersten E, Piradashvili K, Wurm FR, Heller M, Becker S, Anusic T, Brieger J, Strieth S. Surface Modification of Porous Polyethylene Implants with an Albumin-Based Nanocarrier-Release System. Biomedicines 2021; 9:1485. [PMID: 34680602 PMCID: PMC8533240 DOI: 10.3390/biomedicines9101485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Porous polyethylene (PPE) implants are used for the reconstruction of tissue defects but have a risk of rejection in case of insufficient ingrowth into the host tissue. Various growth factors can promote implant ingrowth, yet a long-term gradient is a prerequisite for the mediation of these effects. As modification of the implant surface with nanocarriers may facilitate a long-term gradient by sustained factor release, implants modified with crosslinked albumin nanocarriers were evaluated in vivo. METHODS Nanocarriers from murine serum albumin (MSA) were prepared by an inverse miniemulsion technique encapsulating either a low- or high-molar mass fluorescent cargo. PPE implants were subsequently coated with these nanocarriers. In control cohorts, the implant was coated with the homologue non-encapsulated cargo substance by dip coating. Implants were consequently analyzed in vivo using repetitive fluorescence microscopy utilizing the dorsal skinfold chamber in mice for ten days post implantation. RESULTS Implant-modification with MSA nanocarriers significantly prolonged the presence of the encapsulated small molecules while macromolecules were detectable during the investigated timeframe regardless of the form of application. CONCLUSIONS Surface modification of PPE implants with MSA nanocarriers results in the alternation of release kinetics especially when small molecular substances are used and therefore allows a prolonged factor release for the promotion of implant integration.
Collapse
Affiliation(s)
- Jonas Eckrich
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (N.H.); (M.H.); (J.B.); (S.S.)
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Niklas Hoormann
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (N.H.); (M.H.); (J.B.); (S.S.)
| | - Erik Kersten
- Max Planck Institute for Polymer Research (MPIP), Ackermannweg 10, 55128 Mainz, Germany; (E.K.); (K.P.); (F.R.W.)
| | - Keti Piradashvili
- Max Planck Institute for Polymer Research (MPIP), Ackermannweg 10, 55128 Mainz, Germany; (E.K.); (K.P.); (F.R.W.)
| | - Frederik R. Wurm
- Max Planck Institute for Polymer Research (MPIP), Ackermannweg 10, 55128 Mainz, Germany; (E.K.); (K.P.); (F.R.W.)
- Sustainable Polymer Chemistry, Department of Molecules and Materials, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, Universiteit Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Martin Heller
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (N.H.); (M.H.); (J.B.); (S.S.)
| | - Sven Becker
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076 Tübingen, Germany;
| | - Toni Anusic
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Obere Zahlbacher Str. 69, 55131 Mainz, Germany;
| | - Juergen Brieger
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (N.H.); (M.H.); (J.B.); (S.S.)
| | - Sebastian Strieth
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (N.H.); (M.H.); (J.B.); (S.S.)
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
4
|
Salerno A, Netti PA. Review on Computer-Aided Design and Manufacturing of Drug Delivery Scaffolds for Cell Guidance and Tissue Regeneration. Front Bioeng Biotechnol 2021; 9:682133. [PMID: 34249885 PMCID: PMC8264554 DOI: 10.3389/fbioe.2021.682133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
In the last decade, additive manufacturing (AM) processes have updated the fields of biomaterials science and drug delivery as they promise to realize bioengineered multifunctional devices and implantable tissue engineering (TE) scaffolds virtually designed by using computer-aided design (CAD) models. However, the current technological gap between virtual scaffold design and practical AM processes makes it still challenging to realize scaffolds capable of encoding all structural and cell regulatory functions of the native extracellular matrix (ECM) of health and diseased tissues. Indeed, engineering porous scaffolds capable of sequestering and presenting even a complex array of biochemical and biophysical signals in a time- and space-regulated manner, require advanced automated platforms suitable of processing simultaneously biomaterials, cells, and biomolecules at nanometric-size scale. The aim of this work was to review the recent scientific literature about AM fabrication of drug delivery scaffolds for TE. This review focused on bioactive molecule loading into three-dimensional (3D) porous scaffolds, and their release effects on cell fate and tissue growth. We reviewed CAD-based strategies, such as bioprinting, to achieve passive and stimuli-responsive drug delivery scaffolds for TE and cancer precision medicine. Finally, we describe the authors' perspective regarding the next generation of CAD techniques and the advantages of AM, microfluidic, and soft lithography integration for enhancing 3D porous scaffold bioactivation toward functional bioengineered tissues and organs.
Collapse
Affiliation(s)
| | - Paolo A. Netti
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Interdisciplinary Research Center on Biomaterials, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Chakka JL, Acri T, Laird NZ, Zhong L, Shin K, Elangovan S, Salem AK. Polydopamine functionalized VEGF gene-activated 3D printed scaffolds for bone regeneration. RSC Adv 2021; 11:13282-13291. [PMID: 35423856 PMCID: PMC8697638 DOI: 10.1039/d1ra01193f] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/27/2021] [Indexed: 12/20/2022] Open
Abstract
Bone is a highly vascularized organ and the formation of new blood vessels is essential to regenerate large critical bone defects. In this study, polylactic acid (PLA) scaffolds of 20-80% infill were three-dimensionally (3D) printed using a fused deposition modeling based 3D printer. The PLA scaffolds were coated with polydopamine (PDA) and then were surface-functionalized with polyethyleneimine (PEI) and VEGF-encoding plasmid DNA (pVEGF) nanoplexes (PLA-PDA-PEI-pVEGF). The PLA-PDA-PEI-pVEGF scaffolds with 40% infill demonstrated higher encapsulation efficiency and sustained release of pVEGF than scaffolds with 20, 60 and 80% infill and were therefore used for in vitro and in vivo studies. The PLA-PDA-PEI-pVEGF increased the translation and secretion of VEGF and BMP-2. The PLA-PDA-PEI-pVEGF also yielded a 2- and 4.5-fold change in VEGF and osteocalcin gene expression in vitro, respectively. A tube formation assay using human umbilical vascular endothelial cells (HUVECs) showed a significant increase in tube length when exposed to the PLA-PDA-PEI-pVEGF scaffold, in comparison to PLA and PLA-PDA scaffolds. The PLA-PDA-PEI-pVEGF scaffold in an in vivo rat calvarial critical bone defect model demonstrated 1.6-fold higher new bone formation compared to the PLA-PDA scaffold. H&E and Masson's trichrome staining of bone sections also revealed that the PLA-PDA-PEI-pVEGF scaffold facilitated the formation of more blood vessels in the newly formed bone compared to the PLA and PLA-PDA scaffold groups. Thus, PLA-PDA-PEI-pVEGF might be a potential 3D printed gene activated scaffold for bone regeneration in clinical situations.
Collapse
Affiliation(s)
- Jaidev L Chakka
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| | - Timothy Acri
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| | - Noah Z Laird
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| | - Ling Zhong
- Department of Experimental Research, Sun Yat-sen University Guangzhou PR China
| | - Kyungsup Shin
- Department of Orthodontics, College of Dentistry and Dental Clinics, University of Iowa Iowa City IA-52242 USA
| | - Satheesh Elangovan
- Department of Periodontics, College of Dentistry and Dental Clinics, University of Iowa Iowa City IA-52242 USA
| | - Aliasger K Salem
- Department of Pharmaceutics and Experimental Therapeutics, College of Pharmacy, University of Iowa Iowa City IA-52242 USA +1-319-335-8810
| |
Collapse
|
6
|
He W, Wei D, Zhang J, Huang X, He D, Liu B, Wang Q, Liu M, Liu L, Liu Y, Tian W. Novel bone repairing scaffold consisting of bone morphogenetic Protein-2 and human Beta Defensin-3. J Biol Eng 2021; 15:5. [PMID: 33557881 PMCID: PMC7871609 DOI: 10.1186/s13036-021-00258-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/31/2021] [Indexed: 01/08/2023] Open
Abstract
Background Synthetic biomaterials assist in modulating the vascular response in an injured bone by serving as delivery vehicles of pro-angiogenic molecules to the site of injury or by serving as mimetic platforms which offer support to cell growth and proliferation. Methods This study applied natural phospholipid modified protein technologies together with low temperature three-dimensional printing technology to develop a new model of three-dimensional artificial bone scaffold for potential use in repairing body injuries. The focus was to create a porous structure (PS) scaffold of two components, Bone Morphogenetic Protein-2 and Human Beta Defensin-3 (BMP2 and hBD3), which can synchronously realize directional bone induction, angiogenesis and postoperative antibacterial effects. BMP2 induces osteogenesis, whereas hBD3 is antibacterial. Results Our data showed that in the BMP2-hBD3-PS or hBD3-PS scaffolds, BMP2 had a slow-release rate of about 40% in 30 days, ensuring that BMP2 could penetrate into stem cells for osteogenic differentiation for a long time. The scaffolds promoted cell growth when in combination with BMP2, thus showing its importance in promoting cell growth. Alkaline Phosphatase (ALP) staining showed that the ALP content of BMP2-hBD3-PS and BMP2-PS had a significant increase in samples that contained BMP2, thus showing that these scaffolds promoted osteogenic differentiation. In all the constructs that had hBD3, they displayed antibacterial properties with hBD3, having a slow release of about 35% in 30 days, thus ensuring they provided protection. Conclusion Based on this study, the 3D printed BMP2 scaffolds show a great potential for the development of biodegradable bone implants. Level of evidence Level II, experimental comparative design.
Collapse
Affiliation(s)
- Wei He
- Department of Spine Surgery, Beijing JiShuiTan Hospital, 4th Medical College of Peking University, No.31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Daixu Wei
- Department of Biomaterials and Microorganisms, Northwest University, Xi'an, China
| | - Jun Zhang
- Department of Spine Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical College People's Hospital, Hangzhou, Zhejiang, China
| | - Xiaonan Huang
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Da He
- Department of Spine Surgery, Beijing JiShuiTan Hospital, 4th Medical College of Peking University, No.31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Bo Liu
- Department of Spine Surgery, Beijing JiShuiTan Hospital, 4th Medical College of Peking University, No.31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Qilong Wang
- Department of Spine Surgery, Beijing JiShuiTan Hospital, 4th Medical College of Peking University, No.31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Mingming Liu
- Department of Spine Surgery, Beijing JiShuiTan Hospital, 4th Medical College of Peking University, No.31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Ling Liu
- Department of Gynaecology and Obstetrics, Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yajun Liu
- Department of Spine Surgery, Beijing JiShuiTan Hospital, 4th Medical College of Peking University, No.31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
| | - Wei Tian
- Department of Spine Surgery, Beijing JiShuiTan Hospital, 4th Medical College of Peking University, No.31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
| |
Collapse
|
7
|
Catanzano O, Quaglia F, Boateng JS. Wound dressings as growth factor delivery platforms for chronic wound healing. Expert Opin Drug Deliv 2021; 18:737-759. [PMID: 33338386 DOI: 10.1080/17425247.2021.1867096] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Years of tissue engineering research have clearly demonstrated the potential of integrating growth factors (GFs) into scaffolds for tissue regeneration, a concept that has recently been applied to wound dressings. The old concept of wound dressings that only take a passive role in wound healing has now been overtaken, and advanced dressings which can take an active part in wound healing, are of current research interest.Areas covered: In this review we will focus on the recent strategies for the delivery of GFs to wound sites with an emphasis on the different approaches used to achieve fine tuning of spatial and temporal concentrations to achieve therapeutic efficacy.Expert opinion: The use of GFs to accelerate wound healing and reduce scar formation is now considered a feasible therapeutic approach in patients with a high risk of infections and complications. The integration of micro - and nanotechnologies into wound dressings could be the key to overcome the inherent instability of GFs and offer adequate control over the release rate. Many investigations have led to encouraging outcomes in various in vitro and in vivo wound models, and it is expected that some of these technologies will satisfy clinical needs and will enter commercialization.
Collapse
Affiliation(s)
- Ovidio Catanzano
- Institute for Polymers Composites and Biomaterials (IPCB) - CNR, Pozzuoli, Italy
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Joshua S Boateng
- School of Science, Faculty of Engineering and Science, University of Greenwich, Medway, Central Avenue, Chatham Maritime, Kent, UK
| |
Collapse
|
8
|
Liu H, Du Y, Yang G, Hu X, Wang L, Liu B, Wang J, Zhang S. Delivering Proangiogenic Factors from 3D-Printed Polycaprolactone Scaffolds for Vascularized Bone Regeneration. Adv Healthc Mater 2020; 9:e2000727. [PMID: 32743958 DOI: 10.1002/adhm.202000727] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/10/2020] [Indexed: 01/04/2023]
Abstract
Natural bone is a highly vascularized tissue that relies on the vasculature for blood and nutrients supply to maintain skeletal integrity. Inadequacy of neovascularization may compromise the tissue ingrowth to the implanted scaffolds, and eventually results in failure for the repair. To tackle this issue and enhance self-vascularized bone regeneration, herein a 3D biomimetic selective lasersintering (SLS) derived scaffold, with an angiogenic growth factor immobilized on its surface, that can be released in a controlled manner is proposed. To this end, a porous polycaprolactone/hydroxyapatite (PCL/HA) scaffold is prepared via the SLS technique, which is further modified with vascular endothelial growth factor (VEGF) by coprecipitation with apatite. The resultant scaffold (PCL/HA/VEGF) has an excellent cytocompatibility, and subcutaneous implantation experiment shows that the VEGF-loaded scaffold significantly enhances the blood vessel formation compared with the VEGF-free control. It is further demonstrated that the PCL/HA/VEGF scaffold is able to enhance the in vivo bone regeneration in a rat cranial defect model. Taken together, the current study provides not only a feasible and promising scaffold candidate to enhance the vascularized bone regeneration, but also a general strategy to overcome the inadequate vascularization issue for the repair of other tissue and organs.
Collapse
Affiliation(s)
- Haoming Liu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yingying Du
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Gaojie Yang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xixi Hu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bin Liu
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100037, China
| | - Jianglin Wang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shengmin Zhang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
9
|
Scheiner K, Maas-Bakker RF, Nguyen TT, Duarte AM, Hendriks G, Sequeira L, Duffy GP, Steendam R, Hennink WE, Kok RJ. Sustained Release of Vascular Endothelial Growth Factor from Poly(ε-caprolactone-PEG-ε-caprolactone)- b-Poly(l-lactide) Multiblock Copolymer Microspheres. ACS OMEGA 2019; 4:11481-11492. [PMID: 31460253 PMCID: PMC6681988 DOI: 10.1021/acsomega.9b01272] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/18/2019] [Indexed: 05/14/2023]
Abstract
Vascular endothelial growth factor (VEGF) is the major regulating factor for the formation of new blood vessels, also known as angiogenesis. VEGF is often incorporated in synthetic scaffolds to promote vascularization and to enhance the survival of cells that have been seeded in these devices. Such applications require sustained local delivery of VEGF of around 4 weeks for stable blood vessel formation. Most delivery systems for VEGF only provide short-term release for a couple of days, followed by a release phase with very low VEGF release. We now have developed VEGF-loaded polymeric microspheres that provide sustained release of bioactive VEGF for 4 weeks. Blends of two swellable poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone)-b-poly(l-lactide) ([PCL-PEG-PCL]-b-[PLLA])-based multiblock copolymers with different PEG content and PEG molecular weight were used to prepare the microspheres. Loading of the microspheres was established by a solvent evaporation-based membrane emulsification method. The resulting VEGF-loaded microspheres had average sizes of 40-50 μm and a narrow size distribution. Optimized formulations of a 50:50 blend of the two multiblock copolymers had an average VEGF loading of 0.79 ± 0.09%, representing a high average VEGF loading efficiency of 78 ± 16%. These microspheres released VEGF continuously over 4 weeks in phosphate-buffered saline pH 7.4 at 37 °C. This release profile was preserved after repeated and long-term storage at -20 °C for up to 9 months, thereby demonstrating excellent storage stability. VEGF release was governed by diffusion through the water-filled polymer matrix, depending on PEG molecular weight and PEG content of the polymers. The bioactivity of the released VEGF was retained within the experimental error in the 4-week release window, as demonstrated using a human umbilical vein endothelial cells proliferation assay. Thus, the microspheres prepared in this study are suitable for embedment in polymeric scaffolds with the aim of promoting their functional vascularization.
Collapse
Affiliation(s)
- Karina
C. Scheiner
- Department
of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Roel F. Maas-Bakker
- Department
of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Thanh T. Nguyen
- InnoCore
Pharmaceuticals B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Ana M. Duarte
- InnoCore
Pharmaceuticals B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Gert Hendriks
- InnoCore
Pharmaceuticals B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Lídia Sequeira
- InnoCore
Pharmaceuticals B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Garry P. Duffy
- Discipline
of Anatomy, School of Medicine, National
University of Ireland Galway, University Road, H91 TK33 Galway, Ireland
| | - Rob Steendam
- InnoCore
Pharmaceuticals B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Wim E. Hennink
- Department
of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Robbert J. Kok
- Department
of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- E-mail: . Phone: +31 620275995. Fax: +31 30 251789
| |
Collapse
|
10
|
Fenton OS, Paolini M, Andresen JL, Müller FJ, Langer R. Outlooks on Three-Dimensional Printing for Ocular Biomaterials Research. J Ocul Pharmacol Ther 2019; 36:7-17. [PMID: 31211652 PMCID: PMC6985767 DOI: 10.1089/jop.2018.0142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/07/2019] [Indexed: 10/26/2022] Open
Abstract
Given its potential for high-resolution, customizable, and waste-free fabrication of medical devices and in vitro biological models, 3-dimensional (3D) bioprinting has broad utility within the biomaterials field. Indeed, 3D bioprinting has to date been successfully used for the development of drug delivery systems, the recapitulation of hard biological tissues, and the fabrication of cellularized organ and tissue-mimics, among other applications. In this study, we highlight convergent efforts within engineering, cell biology, soft matter, and chemistry in an overview of the 3D bioprinting field, and we then conclude our work with outlooks toward the application of 3D bioprinting for ocular research in vitro and in vivo.
Collapse
Affiliation(s)
- Owen S. Fenton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Marion Paolini
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jason L. Andresen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Florence J. Müller
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
11
|
Horst M, Eberli D, Gobet R, Salemi S. Tissue Engineering in Pediatric Bladder Reconstruction-The Road to Success. Front Pediatr 2019; 7:91. [PMID: 30984717 PMCID: PMC6449422 DOI: 10.3389/fped.2019.00091] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/01/2019] [Indexed: 12/20/2022] Open
Abstract
Several congenital disorders can cause end stage bladder disease and possibly renal damage in children. The current gold standard therapy is enterocystoplasty, a bladder augmentation using an intestinal segment. However, the use of bowel tissue is associated with numerous complications such as metabolic disturbance, stone formation, urine leakage, chronic infections, and malignancy. Urinary diversions using engineered bladder tissue would obviate the need for bowel for bladder reconstruction. Despite impressive progress in the field of bladder tissue engineering over the past decades, the successful transfer of the approach into clinical routine still represents a major challenge. In this review, we discuss major achievements and challenges in bladder tissue regeneration with a focus on different strategies to overcome the obstacles and to meet the need for living functional tissue replacements with a good growth potential and a long life span matching the pediatric population.
Collapse
Affiliation(s)
- Maya Horst
- Laboratory for Urologic Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital, Zurich, Switzerland
- Division of Pediatric Urology, Department of Pediatric Surgery, University Children‘s Hospital, Zurich, Switzerland
| | - Daniel Eberli
- Division of Pediatric Urology, Department of Pediatric Surgery, University Children‘s Hospital, Zurich, Switzerland
| | - Rita Gobet
- Laboratory for Urologic Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital, Zurich, Switzerland
| | - Souzan Salemi
- Division of Pediatric Urology, Department of Pediatric Surgery, University Children‘s Hospital, Zurich, Switzerland
| |
Collapse
|
12
|
Wang X, Yang L, Zhang H, Tian B, Li R, Hou X, Wei F. Fluorescent magnetic PEI-PLGA nanoparticles loaded with paclitaxel for concurrent cell imaging, enhanced apoptosis and autophagy in human brain cancer. Colloids Surf B Biointerfaces 2018; 172:708-717. [PMID: 30245296 DOI: 10.1016/j.colsurfb.2018.09.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/09/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Abstract
Magnetic nanoparticles are regarded as a promising drug delivery vehicle with the improved efficacy and lowered side effects for antitumor therapy. Herein, the poly lactic-co-glycolic acid (PLGA) modified magnetic nanoplatform was synthesized using superparamagnetic γ-Fe2O3 nanoparticles (MNPs) as a core, and then labelled with polyethylenimine (PEI)-conjugated fluorescein isothiocyanate (FITC), and simultaneously loaded with antitumor drug paclitaxel (PTX) for theranostic analysis of antitumor effects investigated in human brain glioblastoma U251 cells. As a result, the prepared PEI-PLGA-MNPs showed a relatively round sphere with an average size of 80 nm approximately, and the FITC-labeling PEI-PLGA-MNPs were efficiently endocytosed by the U251 cells for cellular imaging. Moreover, the fabricated PEI-PLGA-PTX-MNPs also demonstrated an inhibition of the targeted cell proliferation and migration, and a programmed cell death, via both apoptosis modulating by a burst of reactive oxygen species (ROS) and autophagy with accumulation of autophagosomes and LC3-II signals detected in the treated glioblastoma U251 cells after uptaking. Therefore, the constructed nanoplatform could be effectively applied for simultaneous cellular imaging and drug delivery in human brain glioblastoma treatment in future.
Collapse
Affiliation(s)
- Xueqin Wang
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, PR China.
| | - Liping Yang
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, PR China
| | - Huiru Zhang
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, PR China
| | - Baoming Tian
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Ruifang Li
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, PR China
| | - Xuandi Hou
- College of Bioengineering, Henan University of Technology, Zhengzhou, Henan 450001, PR China
| | - Fang Wei
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| |
Collapse
|
13
|
Wang L, Shi Q, Dai J, Gu Y, Feng Y, Chen L. Increased vascularization promotes functional recovery in the transected spinal cord rats by implanted vascular endothelial growth factor-targeting collagen scaffold. J Orthop Res 2018; 36:1024-1034. [PMID: 28786500 DOI: 10.1002/jor.23678] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 08/01/2017] [Indexed: 02/04/2023]
Abstract
Spinal cord injury (SCI) is global health concern. The effective strategies for SCI are relevant to the improvement on nerve regeneration microenvironment. Vascular endothelial growth factor (VEGF) is an important cytokine for inducing angiogenesis and accelerating nerve system function recovery from injury. We proposed that VEGF could improve nerve regeneration in SCI. However, an uncontrolled delivery system target to injury site not only decreases the therapeutic efficacy but also increases the risk of tumor information. We implanted collagen scaffold (CS) targeted with a constructed protein, collagen-binding VEGF (CBD-VEGF), to bridge transected spine cord gap in a rat transected SCI model. Functional and histological examinations were conducted to assess the repair capacity of the delivery system CS/CBD-VEGF. The results indicated that the implantation of CS/CBD-VEGF into the model rats improved the survival rate and exerted beneficial effect on functional recovery. The controlled intervention improved the microenvironment, guided axon growth, and promoted neovascularization at the injury site. Therefore, the delivery system with stable binding of VEGF potentially provides a better therapeutic option for SCI. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1024-1034, 2018.
Collapse
Affiliation(s)
- Lingjun Wang
- Department of Orthopedic, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Qin Shi
- Department of Orthopedic, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Chinese Academy of Sciences, Institute of Genetics and Developmental Biology, Beijing, 100000, P.R. China
| | - Yong Gu
- Department of Orthopedic, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Yu Feng
- Department of Orthopedic, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Liang Chen
- Department of Orthopedic, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| |
Collapse
|
14
|
Wang B, Lv X, Chen S, Li Z, Yao J, Peng X, Feng C, Xu Y, Wang H. Use of heparinized bacterial cellulose based scaffold for improving angiogenesis in tissue regeneration. Carbohydr Polym 2018; 181:948-956. [DOI: 10.1016/j.carbpol.2017.11.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/21/2022]
|
15
|
Lee SJ, Lee JB, Park YW, Lee DY. 3D Bioprinting for Artificial Pancreas Organ. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:355-374. [PMID: 30471043 DOI: 10.1007/978-981-13-0445-3_21] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Type 1 diabetes mellitus (T1DM) results from an autoimmune destruction of insulin-producing beta cells in the islet of the endocrine pancreas. Although islet transplantation has been regarded as an ideal strategy for T1D, transplanted islets are rejected from host immune system. To immunologically protect them, islet encapsulation technology with biocompatible materials is emerged as an immuno-barrier. However, this technology has been limited for clinical trial such as hypoxia in the central core of islet bead, impurity of islet bead and retrievability from the body. Recently, 3D bioprinting has been emerged as an alternative approach to make the artificial pancreas. It can be used to position live cells in a desired location with real scale of human organ. Furthermore, constructing a vascularization of the artificial pancreas is actualized with 3D bioprinting. Therefore, it is possible to create real pancreas-mimic artificial organ for clinical application. In conclusion, 3D bioprinting can become a new leader in the development of the artificial pancreas to overcome the existed islet.
Collapse
Affiliation(s)
- Seon Jae Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Jae Bin Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Young-Woo Park
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea. .,Institute of Nano Science & Technology (INST), Hanyang University, Seoul, South Korea.
| |
Collapse
|
16
|
Tsui JH, Janebodin K, Ieronimakis N, Yama DMP, Yang HS, Chavanachat R, Hays AL, Lee H, Reyes M, Kim DH. Harnessing Sphingosine-1-Phosphate Signaling and Nanotopographical Cues To Regulate Skeletal Muscle Maturation and Vascularization. ACS NANO 2017; 11:11954-11968. [PMID: 29156133 PMCID: PMC6133580 DOI: 10.1021/acsnano.7b00186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Despite possessing substantial regenerative capacity, skeletal muscle can suffer from loss of function due to catastrophic traumatic injury or degenerative disease. In such cases, engineered tissue grafts hold the potential to restore function and improve patient quality of life. Requirements for successful integration of engineered tissue grafts with the host musculature include cell alignment that mimics host tissue architecture and directional functionality, as well as vascularization to ensure tissue survival. Here, we have developed biomimetic nanopatterned poly(lactic-co-glycolic acid) substrates conjugated with sphingosine-1-phosphate (S1P), a potent angiogenic and myogenic factor, to enhance myoblast and endothelial maturation. Primary muscle cells cultured on these functionalized S1P nanopatterned substrates developed a highly aligned and elongated morphology and exhibited higher expression levels of myosin heavy chain, in addition to genes characteristic of mature skeletal muscle. We also found that S1P enhanced angiogenic potential in these cultures, as evidenced by elevated expression of endothelial-related genes. Computational analyses of live-cell videos showed a significantly improved functionality of tissues cultured on S1P-functionalized nanopatterns as indicated by greater myotube contraction displacements and velocities. In summary, our study demonstrates that biomimetic nanotopography and S1P can be combined to synergistically regulate the maturation and vascularization of engineered skeletal muscles.
Collapse
Affiliation(s)
- Jonathan H. Tsui
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Kajohnkiart Janebodin
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Anatomy, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Nicholas Ieronimakis
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, Washington, USA
| | - David M. P. Yama
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Hee Seok Yang
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
| | | | - Aislinn L. Hays
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Haeshin Lee
- Department of Chemistry and the Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Morayma Reyes
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
17
|
Fahimipour F, Rasoulianboroujeni M, Dashtimoghadam E, Khoshroo K, Tahriri M, Bastami F, Lobner D, Tayebi L. 3D printed TCP-based scaffold incorporating VEGF-loaded PLGA microspheres for craniofacial tissue engineering. Dent Mater 2017; 33:1205-1216. [PMID: 28882369 DOI: 10.1016/j.dental.2017.06.016] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/28/2017] [Accepted: 06/22/2017] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Vascularization is a critical process during bone regeneration/repair and the lack of tissue vascularization is recognized as a major challenge in applying bone tissue engineering methods for cranial and maxillofacial surgeries. The aim of our study is to fabricate a vascular endothelial growth factor (VEGF)-loaded gelatin/alginate/β-TCP composite scaffold by 3D printing method using a computer-assisted design (CAD) model. METHODS The paste, composed of (VEGF-loaded PLGA)-containing gelatin/alginate/β-TCP in water, was loaded into standard Nordson cartridges and promptly employed for printing the scaffolds. Rheological characterization of various gelatin/alginate/β-TCP formulations led to an optimized paste as a printable bioink at room temperature. RESULTS The in vitro release kinetics of the loaded VEGF revealed that the designed scaffolds fulfill the bioavailability of VEGF required for vascularization in the early stages of tissue regeneration. The results were confirmed by two times increment of proliferation of human umbilical vein endothelial cells (HUVECs) seeded on the scaffolds after 10 days. The compressive modulus of the scaffolds, 98±11MPa, was found to be in the range of cancellous bone suggesting their potential application for craniofacial tissue engineering. Osteoblast culture on the scaffolds showed that the construct supports cell viability, adhesion and proliferation. It was found that the ALP activity increased over 50% using VEGF-loaded scaffolds after 2 weeks of culture. SIGNIFICANCE The 3D printed gelatin/alginate/β-TCP scaffold with slow releasing of VEGF can be considered as a potential candidate for regeneration of craniofacial defects.
Collapse
Affiliation(s)
- F Fahimipour
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | | | - E Dashtimoghadam
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - K Khoshroo
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - M Tahriri
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - F Bastami
- Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - D Lobner
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - L Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA; Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK.
| |
Collapse
|
18
|
Ji Y, Wang M, Liu W, Chen C, Cui W, Sun T, Feng Q, Guo X. Chitosan/nHAC/PLGA microsphere vehicle for sustained release of rhBMP-2 and its derived synthetic oligopeptide for bone regeneration. J Biomed Mater Res A 2017; 105:1593-1606. [PMID: 27862940 DOI: 10.1002/jbm.a.35962] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/31/2016] [Accepted: 11/08/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Yanhui Ji
- Department of Orthopaedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Mingbo Wang
- Key Laboratory of Biomedical Materials and Implants; Research Institute of Tsinghua University in Shenzhen; Shenzhen 518057 China
| | - Weiqiang Liu
- Key Laboratory of Biomedical Materials and Implants; Research Institute of Tsinghua University in Shenzhen; Shenzhen 518057 China
| | - Changsheng Chen
- Key Laboratory of Biomedical Materials and Implants; Research Institute of Tsinghua University in Shenzhen; Shenzhen 518057 China
| | - Wei Cui
- Department of Orthopaedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Tingfang Sun
- Department of Orthopaedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| | - Qingling Feng
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering; Tsinghua University; Beijing 100084 China
| | - Xiaodong Guo
- Department of Orthopaedics; Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan 430022 China
| |
Collapse
|
19
|
Chen X, Wang M, Xu X, Liu J, Mei B, Fu P, Zhao D, Sun L. Panax ginseng total protein promotes proliferation and secretion of collagen in NIH/3T3 cells by activating extracellular signal-related kinase pathway. J Ginseng Res 2017; 41:411-418. [PMID: 28701885 PMCID: PMC5489768 DOI: 10.1016/j.jgr.2017.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recently, protein from ginseng was studied and used for the treatment of several kinds of diseases. However, the effect of ginseng total protein (GTP) on proliferation and wound healing in fibroblast cells remains unclear. METHODS In this study, cell viability was analyzed using the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. Cell cycle distribution was analyzed by flow cytometer. The levels of transforming growth factor β1, vascular endothelial growth factor, and collagens were analyzed by enzyme-linked immunosorbent assay and immunofluorescence staining. The expressions of cyclin A, phosphorylation of extracellular signal-related kinase (p-ERK1/2), and ERK1/2 were analyzed by Western blotting. RESULTS Our results showed that GTP promoted cell proliferation and increased the percentage of cells in S phase through the upregulation of cyclin A in NIH/3T3 cells. We also found that GTP induced the secretion of type I collagen, and promoted the expression of other factors that regulate the synthesis of collagen such as transforming growth factor β1 and vascular endothelial growth factor. In addition, the phosphorylation of ERK1/2 at Thr202/Tyr204 was also increased by GTP. CONCLUSION Our studies suggest that GTP promoted proliferation and secretion of collagen in NIH/3T3 cells by activating the ERK signal pathway, which shed light on a potential function of GTP in promoting wound healing.
Collapse
Affiliation(s)
- Xuenan Chen
- Research Center of Traditional Chinese Medicine, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Manying Wang
- Research Center of Traditional Chinese Medicine, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaohao Xu
- Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, Jilin, China
| | - Jianzeng Liu
- Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, Jilin, China
- Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Bing Mei
- Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, Changchun, Jilin, China
- College of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Pingping Fu
- China–Japan Union Hospital and First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Daqing Zhao
- Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, Changchun, Jilin, China
- Corresponding author. Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, Jilin Province 130021, China.Research and Development Center of Traditional Chinese Medicine and Biological EngineeringChangchun University of Chinese Medicine1035 Boshuo RoadChangchunJilin Province130021China
| | - Liwei Sun
- Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, Jilin, China
- Corresponding author. Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, 15 Jilin Street, Jilin, Jilin Province 132013, China.Jilin Technology Innovation Center for Chinese Medicine BiotechnologyCollege of Biology and ChemistryBeihua University15 Jilin StreetJilinJilin Province132013China
| |
Collapse
|
20
|
Bayrak ES, Akar B, Somo SI, Lu C, Xiao N, Brey EM, Cinar A. Computational Model-Based Analysis of Strategies to Enhance Scaffold Vascularization. Biores Open Access 2016; 5:342-355. [PMID: 27965914 PMCID: PMC5144865 DOI: 10.1089/biores.2016.0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Stable and extensive blood vessel networks are required for cell function and survival in engineered tissues. A number of different strategies are currently being investigated to enhance biomaterial vascularization with screening primarily through extensive in vitro and in vivo experiments. In this article, we describe an agent-based model (ABM) developed to evaluate various strategies in silico, including design of optimal biomaterial structure, delivery of angiogenic factors, and application of prevascularized biomaterials. The model predictions are evaluated using experimental data. The ABM developed provides insight into different strategies currently applied for scaffold vascularization and will enable researchers to rapidly screen new hypotheses and explore alternative strategies for enhancing vascularization.
Collapse
Affiliation(s)
- Elif Seyma Bayrak
- Department of Chemical and Biological Engineering, Illinois Institute of Technology , Chicago, Illinois
| | - Banu Akar
- Department of Biomedical Engineering, Illinois Institute of Technology , Chicago, Illinois
| | - Sami I Somo
- Department of Biomedical Engineering, Illinois Institute of Technology , Chicago, Illinois
| | - Chenlin Lu
- Department of Chemical and Biological Engineering, Illinois Institute of Technology , Chicago, Illinois
| | - Nan Xiao
- Department of Chemical and Biological Engineering, Illinois Institute of Technology , Chicago, Illinois
| | - Eric M Brey
- Department of Biomedical Engineering, Illinois Institute of Technology , Chicago, Illinois
| | - Ali Cinar
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois.; Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| |
Collapse
|
21
|
Patra S, Young V. A Review of 3D Printing Techniques and the Future in Biofabrication of Bioprinted Tissue. Cell Biochem Biophys 2016; 74:93-8. [DOI: 10.1007/s12013-016-0730-0] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 04/22/2016] [Indexed: 12/21/2022]
|
22
|
Minardi S, Corradetti B, Taraballi F, Sandri M, Martinez JO, Powell ST, Tampieri A, Weiner BK, Tasciotti E. Biomimetic Concealing of PLGA Microspheres in a 3D Scaffold to Prevent Macrophage Uptake. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1479-1488. [PMID: 26797709 DOI: 10.1002/smll.201503484] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Indexed: 06/05/2023]
Abstract
Scaffolds functionalized with delivery systems for the release of growth factors is a robust strategy to enhance tissue regeneration. However, after implantation, macrophages infiltrate the scaffold, eventually initiating the degradation and clearance of the delivery systems. Herein, it is hypothesized that fully embedding the poly(d,l-lactide-co-glycolide acid) microspheres (MS) in a highly structured collagen-based scaffold (concealing) can prevent their detection, preserving the integrity of the payload. Confocal laser microscopy reveals that non-embedded MS are easily internalized; when concealed, J774 and bone marrow-derived macrophages (BMDM) cannot detect them. This is further demonstrated by flow cytometry, as a tenfold decrease is found in the number of MS engulfed by the cells, suggesting that collagen can cloak the MS. This correlates with the amount of nitric oxide and tumor necrosis factor-α produced by J774 and BMDM in response to the concealed MS, comparable to that found for non-functionalized collagen scaffolds. Finally, the release kinetics of a reporter protein is preserved in the presence of macrophages, only when MS are concealed. The data provide detailed strategies for fabricating three dimensional (3D) biomimetic scaffolds able to conceal delivery systems and preserve the therapeutic molecules for release.
Collapse
Affiliation(s)
- Silvia Minardi
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
- Institute of Science and Technology for Ceramics-CNR (ISTEC-CNR), Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Bruna Corradetti
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131, Ancona, Italy
| | - Francesca Taraballi
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Monica Sandri
- Institute of Science and Technology for Ceramics-CNR (ISTEC-CNR), Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Jonathan O Martinez
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Sebastian T Powell
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Anna Tampieri
- Institute of Science and Technology for Ceramics-CNR (ISTEC-CNR), Via Granarolo 64, 48018, Faenza, RA, Italy
| | - Bradley K Weiner
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
- Department of Orthopedic Surgery, Houston Methodist Hospital, 6550 Fannin St., Houston, TX, 77030, USA
| | - Ennio Tasciotti
- Department of Regenerative Medicine, Houston Methodist Research Institute (HMRI), 6670 Bertner Ave., Houston, TX, 77030, USA
| |
Collapse
|
23
|
Cao H, Chen MM, Liu Y, Liu YY, Huang YQ, Wang JH, Chen JD, Zhang QQ. Fish collagen-based scaffold containing PLGA microspheres for controlled growth factor delivery in skin tissue engineering. Colloids Surf B Biointerfaces 2015; 136:1098-106. [PMID: 26618451 DOI: 10.1016/j.colsurfb.2015.10.022] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/10/2015] [Accepted: 10/16/2015] [Indexed: 02/06/2023]
Abstract
To design a scaffold controlled release system for skin tissue engineering, fish collagen/chitosan/chondroitin sulfate scaffolds were fabricated by freeze-drying and incorporated with bFGF-loaded PLGA microspheres (MPs). SEM showed that the scaffolds exhibited an interconnected porous structure, and the spherical MPs were uniformly distributed into the scaffolds. The higher swelling and degradation rate of scaffolds/MPs could lead to a higher diffusion rate of MPs from the scaffolds, causing an increase in the protein release. The release rate of proteins could be adjusted by the size of MPs and the ratio of collagen to chitosan of scaffolds. Circular dichroism spectroscopy and MTT of bFGF after release indicated that the released bFGF retained its structural integrity and bioactivity during preparation. Cell proliferation and in vivo evaluation results suggested that the scaffolds/MPs had a good biocompatibility and an ability to promote fibroblast cell proliferation and skin tissue regeneration. These results demonstrated that this scaffold/MP controlled release system has the potential for skin tissue engineering.
Collapse
Affiliation(s)
- Huan Cao
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou 350002, China
| | - Ming-Mao Chen
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou 350002, China.
| | - Yan Liu
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Yuan-Yuan Liu
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou 350002, China
| | - Yu-Qing Huang
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou 350002, China
| | - Jian-Hua Wang
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou 350002, China
| | - Jing-Di Chen
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou 350002, China
| | - Qi-Qing Zhang
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou 350002, China; Key Laboratory of Biomedical Material of Tianjin, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
24
|
de Alteriis R, Vecchione R, Attanasio C, De Gregorio M, Porzio M, Battista E, Netti PA. A method to tune the shape of protein-encapsulated polymeric microspheres. Sci Rep 2015. [PMID: 26224659 PMCID: PMC4519779 DOI: 10.1038/srep12634] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Protein encapsulation technologies of polymeric microspheres currently in use have been optimized to effectively protect their “protein cargo” from inactivation occurring in biological environments, preserving its bioactivity during release up to several weeks. The scenario of protein delivery would greatly benefit by strategies enabling the production of non-spherical particles. Herein we report an easy and effective stamp-based method to produce poly-lactic-glycolic-acid (PLGA) microparticles encapsulating Vascular Endothelial Growth Factor (VEGF) of different shapes. We demonstrate that PLGA microspheres can be deformed at room temperature exploiting solvent/non-solvent plasticization in order to preserve the properties of the starting microspheres. This gentle method allows the production of shaped particles that provide a prolonged release of VEGF in active form, as verified by an angiogenic assay. The retention of the biological activity of an extremely labile molecule, i.e. VEGF, lets us hypothesize that a wide variety of drug and protein encapsulated polymeric microspheres can be processed using this method.
Collapse
Affiliation(s)
- Renato de Alteriis
- 1] Centro di Ricerca Interdipartimentale sui Biomateriali - CRIB, Università degli studi di Napoli Federico II, Napoli, 80125, Italy [2] Center for Advanced Biomaterials for Health Care - IIT@CRIB Istituto Italiano di Tecnologia, Napoli, 80125, Italy
| | - Raffaele Vecchione
- 1] Centro di Ricerca Interdipartimentale sui Biomateriali - CRIB, Università degli studi di Napoli Federico II, Napoli, 80125, Italy [2] Center for Advanced Biomaterials for Health Care - IIT@CRIB Istituto Italiano di Tecnologia, Napoli, 80125, Italy
| | - Chiara Attanasio
- Center for Advanced Biomaterials for Health Care - IIT@CRIB Istituto Italiano di Tecnologia, Napoli, 80125, Italy
| | - Maria De Gregorio
- 1] Centro di Ricerca Interdipartimentale sui Biomateriali - CRIB, Università degli studi di Napoli Federico II, Napoli, 80125, Italy [2] Center for Advanced Biomaterials for Health Care - IIT@CRIB Istituto Italiano di Tecnologia, Napoli, 80125, Italy [3] Dipartimento di Medicina Veterinaria e Produzioni Animali, Napoli, 80137, Italy
| | - Massimiliano Porzio
- Centro di Ricerca Interdipartimentale sui Biomateriali - CRIB, Università degli studi di Napoli Federico II, Napoli, 80125, Italy
| | - Edmondo Battista
- Centro di Ricerca Interdipartimentale sui Biomateriali - CRIB, Università degli studi di Napoli Federico II, Napoli, 80125, Italy
| | - Paolo A Netti
- 1] Centro di Ricerca Interdipartimentale sui Biomateriali - CRIB, Università degli studi di Napoli Federico II, Napoli, 80125, Italy [2] Center for Advanced Biomaterials for Health Care - IIT@CRIB Istituto Italiano di Tecnologia, Napoli, 80125, Italy
| |
Collapse
|
25
|
Improved Angiogenesis in Response to Localized Delivery of Macrophage-Recruiting Molecules. PLoS One 2015; 10:e0131643. [PMID: 26132702 PMCID: PMC4489184 DOI: 10.1371/journal.pone.0131643] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/03/2015] [Indexed: 12/31/2022] Open
Abstract
Successful engineering of complex organs requires improved methods to promote rapid and stable vascularization of artificial tissue scaffolds. Toward this goal, tissue engineering strategies utilize the release of pro-angiogenic growth factors, alone or in combination, from biomaterials to induce angiogenesis. In this study we have used intravital microscopy to define key, dynamic cellular changes induced by the release of pro-angiogenic factors from polyethylene glycol diacrylate hydrogels transplanted in vivo. Our data show robust macrophage recruitment when the potent and synergistic angiogenic factors, PDGFBB and FGF2 were used as compared with VEGF alone and intravital imaging suggested roles for macrophages in endothelial tip cell migration and anastomosis, as well as pericyte-like behavior. Further data from in vivo experiments show that delivery of CSF1 with VEGF can dramatically improve the poor angiogenic response seen with VEGF alone. These studies show that incorporating macrophage-recruiting factors into the design of pro-angiogenic biomaterial scaffolds is a key strategy likely to be necessary for stable vascularization and survival of implanted artificial tissues.
Collapse
|
26
|
Schliephake H, Vucak M, Boven J, Backhaus S, Annen T, Epple M. Solvent free production of porous PDLLA/calcium carbonate composite scaffolds improves the release of bone growth factors. Oral Maxillofac Surg 2015; 19:133-141. [PMID: 25178431 DOI: 10.1007/s10006-014-0463-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/21/2014] [Indexed: 06/03/2023]
Abstract
PURPOSE Incorporation of alkaline nano-/microparticles for neutralization of acidic degradation products into degradable polymer foams requires the use of organic solvents, which may compromise biocompatibility and may be associated with biological hazards. The aim of the present study was to develop and validate a solvent-free method to produce porous poly (DL-lactic acid)/calcium carbonate composite scaffolds (PDLLA/CaCO3) for controlled release of incorporated osteogenic growth factors. METHODS Composite PDLLA/CaCO3 granules were produced using a milling process and compared to composite material fabricated through a solution precipitation process using organic solvents. Particle size and mineral content were comparable in both groups. Supercritical carbon dioxide pressure was used to incorporate rhBMP2 into both composites. RESULTS Gas foaming resulted in comparable pore structures in both groups exhibiting a homogenous distribution of CaCO3 microparticles in the polymer scaffolds. The elasticity modulus of both types of scaffolds was not significantly different whereas the bending strength of the solvent-free produced scaffolds was significantly lower. The pH values remained constant between 6.90 and 7.25 during degradation of both composites. Release of BMP2 was significantly higher and the induction of alkaline phosphatase was more reliable in the group of scaffolds produced without organic solvents. CONCLUSION Solvent-free fabrication of composite PDLLA/CaCO3 scaffolds for controlled release of bone growth factors through gas foaming significantly enhances the release of growth factors and improves the biological efficacy of the incorporated growth factors.
Collapse
Affiliation(s)
- H Schliephake
- Department of Oral and Maxillofacial Surgery, George-Augusta-University, Robert-Koch-Str, 40 37075, Göttingen, Germany,
| | | | | | | | | | | |
Collapse
|
27
|
Quinlan E, López-Noriega A, Thompson EM, Hibbitts A, Cryan SA, O'Brien FJ. Controlled release of vascular endothelial growth factor from spray-dried alginate microparticles in collagen-hydroxyapatite scaffolds for promoting vascularization and bone repair. J Tissue Eng Regen Med 2015; 11:1097-1109. [DOI: 10.1002/term.2013] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Elaine Quinlan
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin; Ireland
- Advanced Materials and BioEngineering Research (AMBER) Centre, RCSI & TCD; Dublin 2 Ireland
| | - Adolfo López-Noriega
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- School of Pharmacy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin; Ireland
- Advanced Materials and BioEngineering Research (AMBER) Centre, RCSI & TCD; Dublin 2 Ireland
| | - Emmet M. Thompson
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin; Ireland
- Advanced Materials and BioEngineering Research (AMBER) Centre, RCSI & TCD; Dublin 2 Ireland
| | - Alan Hibbitts
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin; Ireland
- Advanced Materials and BioEngineering Research (AMBER) Centre, RCSI & TCD; Dublin 2 Ireland
| | - Sally Ann Cryan
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- School of Pharmacy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin; Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin; Ireland
- Advanced Materials and BioEngineering Research (AMBER) Centre, RCSI & TCD; Dublin 2 Ireland
| |
Collapse
|
28
|
Carenza E, Jordan O, Martínez-San Segundo P, Jiřík R, Starčuk jr Z, Borchard G, Rosell A, Roig A. Encapsulation of VEGF165into magnetic PLGA nanocapsules for potential local delivery and bioactivity in human brain endothelial cells. J Mater Chem B 2015; 3:2538-2544. [DOI: 10.1039/c4tb01895h] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
New drug delivery systems based on biodegradable magnetic nanocapsules for targeted delivery of pro-angiogenic proteins, potentially useful in therapeutic angiogenesis, are reported.
Collapse
Affiliation(s)
- E. Carenza
- Institut de Ciència de Materials de Barcelona
- Consejo Superior de Investigaciones Científicas (ICMAB-CSIC)
- Campus de la UAB
- 08193 Bellaterra(Barcelona)
- Spain
| | - O. Jordan
- School of Pharmaceutical Sciences
- University of Geneva
- 1205 Genève
- Switzerland
| | - P. Martínez-San Segundo
- Neurovascular Research Laboratory and Neurovascular Unit
- Vall d'Hebron Institut de Recerca
- Universitat Autònoma de Barcelona
- 119-129 Barcelona
- Spain
| | - R. Jiřík
- Institute of Scientific Instruments
- Academy of Sciences of the Czech Republic
- 612 64 Brno
- Czech Republic
| | - Z. Starčuk jr
- Institute of Scientific Instruments
- Academy of Sciences of the Czech Republic
- 612 64 Brno
- Czech Republic
| | - G. Borchard
- School of Pharmaceutical Sciences
- University of Geneva
- 1205 Genève
- Switzerland
| | - A. Rosell
- Neurovascular Research Laboratory and Neurovascular Unit
- Vall d'Hebron Institut de Recerca
- Universitat Autònoma de Barcelona
- 119-129 Barcelona
- Spain
| | - A. Roig
- Institut de Ciència de Materials de Barcelona
- Consejo Superior de Investigaciones Científicas (ICMAB-CSIC)
- Campus de la UAB
- 08193 Bellaterra(Barcelona)
- Spain
| |
Collapse
|
29
|
Quinlan E, López-Noriega A, Thompson E, Kelly HM, Cryan SA, O'Brien FJ. Development of collagen–hydroxyapatite scaffolds incorporating PLGA and alginate microparticles for the controlled delivery of rhBMP-2 for bone tissue engineering. J Control Release 2015; 198:71-9. [DOI: 10.1016/j.jconrel.2014.11.021] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 12/20/2022]
|
30
|
Shah RB, Schwendeman SP. A biomimetic approach to active self-microencapsulation of proteins in PLGA. J Control Release 2014; 196:60-70. [PMID: 25219750 PMCID: PMC4268178 DOI: 10.1016/j.jconrel.2014.08.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/19/2014] [Accepted: 08/30/2014] [Indexed: 01/10/2023]
Abstract
A biomimetic approach to organic solvent-free microencapsulation of proteins based on the self-healing capacity of poly (DL)-lactic-co-glycolic acid (PLGA) microspheres containing glycosaminoglycan-like biopolymers (BPs), was examined. To screen BPs, aqueous solutions of BP [high molecular weight dextran sulfate (HDS), low molecular weight dextran sulfate (LDS), chondroitin sulfate (CS), heparin (HP), hyaluronic acid (HA), chitosan (CH)] and model protein lysozyme (LYZ) were combined in different molar and mass ratios, at 37 °C and pH7. The BP-PLGA microspheres (20-63 μm) were prepared by a double water-oil-water emulsion method with a range of BP content, and trehalose and MgCO3 to control microclimate pH and to create percolating pores for protein. Biomimetic active self-encapsulation (ASE) of proteins [LYZ, vascular endothelial growth factor165 (VEGF) and fibroblast growth factor (FgF-20)] was accomplished by incubating blank BP-PLGA microspheres in low concentration protein solutions at ~24 °C, for 48 h. Pore closure was induced at 42.5 °C under mild agitation for 42h. Formulation parameters of BP-PLGA microspheres and loading conditions were studied to optimize protein loading and subsequent release. LDS and HP were found to bind >95% LYZ at BP:LYZ>0.125 w/w, whereas HDS and CS bound >80% LYZ at BP:LYZ of 0.25-1 and <0.33, respectively. HA-PLGA microspheres were found to be not ideal for obtaining high protein loading (>2% w/w of LYZ). Sulfated BP-PLGA microspheres were capable of loading LYZ (~2-7% w/w), VEGF (~4% w/w), and FgF-20 (~2% w/w) with high efficiency. Protein loading was found to be dependent on the loading solution concentration, with higher protein loading obtained at higher loading solution concentration within the range investigated. Loading also increased with content of sulfated BP in microspheres. Release kinetics of proteins was evaluated in-vitro with complete release media replacement. Rate and extent of release were found to depend upon volume of release (with non-sink conditions observed <5 ml release volume for ~18 mg loaded BP-PLGA microspheres), ionic strength of release media and loading solution concentration. HDS-PLGA formulations were identified as having ideal loading and release characteristics. These optimal microspheres released ~73-80% of the encapsulated LYZ over 60 days, with >90% of protein being enzymatically active. Nearly 72% of immunoreactive VEGF was similarly released over 42 days, without significant losses in heparin binding affinity in the release medium.
Collapse
Affiliation(s)
- Ronak B Shah
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA.
| |
Collapse
|
31
|
Sustained delivery of rhBMP-2 by means of poly(lactic-co-glycolic acid) microspheres: cranial bone regeneration without heterotopic ossification or craniosynostosis. Plast Reconstr Surg 2014; 134:51-59. [PMID: 24622573 DOI: 10.1097/prs.0000000000000287] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Commercially available recombinant human bone morphogenetic protein 2 (rhBMP2) has demonstrated efficacy in bone regeneration, but not without significant side effects. The authors used rhBMP2 encapsulated in poly(lactic-co-glycolic acid) (PLGA) microspheres placed in a rabbit cranial defect model to test whether low-dose, sustained delivery can effectively induce bone regeneration. METHODS The rhBMP2 was encapsulated in 15% PLGA using a double-emulsion, solvent extraction/evaporation technique, and its release kinetics and bioactivity were tested. Two critical-size defects (10 mm) were created in the calvaria of New Zealand white rabbits (5 to 7 months of age, male and female) and filled with a collagen scaffold containing either (1) no implant, (2) collagen scaffold only, (3) PLGA-rhBMP2 (0.1 μg per implant), or (4) free rhBMP2 (0.1 μg per implant). After 6 weeks, the rabbits were killed and defects were analyzed by micro-computed tomography, histology, and finite element analysis. RESULTS The rhBMP2 delivered by means of bioactive PLGA microspheres resulted in higher volumes and surface area coverage of new bone than an equal dose of free rhBMP2 by micro-computed tomography (p=0.025 and p=0.025). Finite element analysis indicated that the mechanical competence using the regional elastic modulus did not differ with rhBMP2 exposure (p=0.70). PLGA-rhBMP2 did not demonstrate heterotopic ossification, craniosynostosis, or seroma formation. CONCLUSIONS Sustained delivery by means of PLGA microspheres can significantly reduce the rhBMP2 dose required for de novo bone formation. Optimization of the delivery system may be a key to reducing the risk for recently reported rhBMP2-related adverse effects.
Collapse
|
32
|
Li W, Lan Y, Guo R, Zhang Y, Xue W, Zhang Y. In vitro and in vivo evaluation of a novel collagen/cellulose nanocrystals scaffold for achieving the sustained release of basic fibroblast growth factor. J Biomater Appl 2014; 29:882-93. [PMID: 25114196 DOI: 10.1177/0885328214547091] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue-engineered dermis is thought to be the best treatment for skin defects; however, slow vascularization of these biomaterial scaffolds limits their clinical application. Exogenous administration of angiogenic growth factors is highly desirable for tissue regeneration. In this study, biodegradable gelatin microspheres (GMs) containing basic fibroblast growth factor (bFGF) were fabricated and incorporated into a porous collagen/cellulose nanocrystals (CNCs) scaffold, as a platform for long-term release and consequent angiogenic boosting. The physicochemical properties of these scaffolds were examined and the in vitro release pattern of bFGF from scaffolds was measured by ELISA. Collagen/CNCs scaffolds with and without bFGF-GMs were incubated with human umbilical vein endothelial cells for 1 week, results showed that the scaffolds with bFGF-GMs significantly augmented cell proliferation. Then, four different groups of scaffolds were implanted subcutaneously into Sprague-Dawley rats to study angiogenesis in vivo via macroscopic observation, and hematoxylin and eosin and immunohistochemical staining. The results suggested that the collagen/CNCs/bFGF-GMs scaffolds had a significantly higher number of newly formed and mature blood vessels, and the fastest degradation rate. This study demonstrated that collagen/CNCs/bFGF-GMs scaffolds have great potential in skin tissue engineering.
Collapse
Affiliation(s)
- Weichang Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Jinan University, Guangzhou, China Department of Chemistry, Jinan University, Guangzhou, China
| | - Yong Lan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Jinan University, Guangzhou, China Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Jinan University, Guangzhou, China Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Yi Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Jinan University, Guangzhou, China Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Jinan University, Guangzhou, China Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Yuanming Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Jinan University, Guangzhou, China Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|
33
|
Banks JM, Mozdzen LC, Harley BAC, Bailey RC. The combined effects of matrix stiffness and growth factor immobilization on the bioactivity and differentiation capabilities of adipose-derived stem cells. Biomaterials 2014; 35:8951-9. [PMID: 25085859 DOI: 10.1016/j.biomaterials.2014.07.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/10/2014] [Indexed: 12/13/2022]
Abstract
Biomaterial designs are increasingly incorporating multiple instructive signals to induce a desired cell response. However, many approaches do not allow orthogonal manipulation of immobilized growth factor signals and matrix stiffness. Further, few methods support patterning of biomolecular signals across a biomaterial in a spatially-selective manner. Here, we report a sequential approach employing carbodiimide crosslinking and benzophenone photoimmobilization chemistries to orthogonally modify the stiffness and immobilized growth factor content of a model collagen-GAG (CG) biomaterial. We subsequently examined the singular and combined effects of bone morphogenetic protein (BMP-2), platelet derived growth factor (PDGF-BB), and CG membrane stiffness on the bioactivity and osteogenic/adipogenic lineage-specific gene expression of adipose derived stem cells, an increasingly popular cell source for regenerative medicine studies. We found that the stiffest substrates direct osteogenic lineage commitment of ASCs regardless of the presence or absence of growth factors, while softer substrates require biochemical cues to direct cell fate. We subsequently describe the use of this approach to create overlapping patterns of growth factors across a single substrate. These results highlight the need for versatile approaches to selectively manipulate the biomaterial microenvironment to identify synergies between biochemical and mechanical cues for a range of regenerative medicine applications.
Collapse
Affiliation(s)
- Jessica M Banks
- Dept. of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Laura C Mozdzen
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Ryan C Bailey
- Dept. of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
34
|
Belair D, Khalil AS, Miller MJ, Murphy WL. Serum-dependence of affinity-mediated VEGF release from biomimetic microspheres. Biomacromolecules 2014; 15:2038-48. [PMID: 24773176 PMCID: PMC4059260 DOI: 10.1021/bm500177c] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 04/18/2014] [Indexed: 12/29/2022]
Abstract
Vascular endothelial growth factor (VEGF) activity is highly regulated via sequestering within the ECM and cell-demanded proteolysis to release the sequestered VEGF. Numerous studies have demonstrated that VEGF activity mediates cellular events leading to angiogenesis and capillary formation in vivo. This has motivated the study of biomaterials to sustain VEGF release, and in many cases, the materials are inspired by the structure and function of the native ECM. However, there remains a need for materials that can bind to VEGF with high specificity, as the in vivo environment is rich in a variety of growth factors (GFs) and GF-binding moieties. Here we describe a strategy to control VEGF release using hydrogel microspheres with tethered peptides derived from VEGF receptor 2 (VEGFR2). Using biomaterials covalently modified with varying concentrations of two distinct VEGFR2-derived peptides with varying serum stability, we analyzed both biomaterial and environmental variables that influence VEGF release and activity. The presence of tethered VEGF-binding peptides (VBPs) resulted in significantly extended VEGF release relative to control conditions, and the resulting released VEGF significantly increased the expansion of human umbilical vein endothelial cells in culture. VEGF release rates were also strongly influenced by the concentration of serum. The presence of Feline McDonough Sarcoma-like tyrosine kinase 1 (sFlt-1), a serum-borne receptor fragment derived from VEGF receptor 1, increased VEGF release rates, although sFlt-1 was not sufficient to recapitulate the release profile of VEGF in serum. Further, the influence of serum on VEGF release was not due to protease activity or nonspecific VEGF interactions in the presence of serum-borne heparin. VEGF release kinetics correlated well with a generalizable mathematical model describing affinity-mediated release of VEGF from hydrogel microspheres in defined conditions. Modeling results suggest a potential mechanism whereby competition between VEGF and multiple VEGF-binding serum proteins including sFlt-1, soluble kinase insert domain receptor (sKDR), and α2-macroglobulin (α2-M) likely influenced VEGF release from microspheres. The materials and mathematical model described in this approach may be useful in a range of applications in which sustained, biologically active GF release of a specific GF is desirable.
Collapse
Affiliation(s)
- David
G. Belair
- Department
of Biomedical Engineering, University of
Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Andrew S. Khalil
- Department
of Biomedical Engineering, University of
Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Michael J. Miller
- Department
of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - William L. Murphy
- Department
of Biomedical Engineering, University of
Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Department
of Orthopedics and Rehabilitation, University
of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
35
|
Thermally triggered release of a pro-osteogenic peptide from a functionalized collagen-based scaffold using thermosensitive liposomes. J Control Release 2014; 187:158-66. [PMID: 24878185 DOI: 10.1016/j.jconrel.2014.05.043] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 05/19/2014] [Accepted: 05/20/2014] [Indexed: 11/24/2022]
Abstract
Collagen is one of the most attractive materials for the development of matrices for tissue engineering, due to its excellent biocompatibility and non-toxic bioresorption. The present work describes a collagen-based externally controlled drug-eluting scaffold which consists of drug encapsulated thermoresponsive liposomes covalently attached to the surface of a functionalized collagen-based scaffold. The model drug used in this work was PTHrP 107-111, a pentapeptide with pro-osteogenic and antiosteoclastic activity. An osteoconductive collagen-hydroxyapatite scaffold, designed specifically for bone repair, was used as a model scaffold. The results demonstrate that it is possible to modify the kinetics of release of the drug from the scaffold with the application of an external thermal stimulus (42°C, 20min). In vitro studies carried out with pre-osteoblastic MC3T3-E1 cells demonstrated that neither the attachment of liposomes to the surface of the scaffolds nor the hyperthermic pulse negatively affected the ability of cells to attach and proliferate on the scaffolds. Importantly, the on-demand release of PTHrP 107-111 had a pro-osteogenic effect, as shown by the enhancement of alkaline phosphatase activity, an early osteogenic marker, which correlated with increased expression of the osteogenic genes osteopontin and osteocalcin. In conclusion, the scaffold-based release system developed in this study has immense potential for tuning the delivery of a diverse range of drugs which can be applied for the regeneration of a variety of tissue types.
Collapse
|
36
|
Farokhi M, Mottaghitalab F, Shokrgozar MA, Ai J, Hadjati J, Azami M. Bio-hybrid silk fibroin/calcium phosphate/PLGA nanocomposite scaffold to control the delivery of vascular endothelial growth factor. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 35:401-10. [PMID: 24411394 DOI: 10.1016/j.msec.2013.11.023] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/24/2013] [Accepted: 11/16/2013] [Indexed: 11/17/2022]
Abstract
This study investigated the efficacy of bio-hybrid silk fibroin/Calcium phosphate/PLGA nanocomposite scaffold as vascular endothelial growth factor (VEGF) delivery system. The scaffold was fabricated using freeze-drying and electrospinning. Here, we highlight the structural changes of the scaffold using scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), Raman spectroscopy and differential scanning calorimetry (DSC). The uniform dispersion of calcium phosohate (CaP) powder within silk fibroin (SF) solution was also confirmed using Zeta potential analysis. Moreover, good biocompatibility of osteoblast cells next to the scaffold was approved by cell adhesion, proliferation and alkaline phosphatase production. The release profile of VEGF during 28 days has established the efficacy of the scaffold as a sustained delivery system. The bioactivity of the released VEGF was maintained about 83%. The histology analysis has shown that the new bone tissue formation happened in the defected site after 10 weeks of implantation. Generally, our data showed that the fabricated scaffold could be considered as an effective scaffold for bone tissue engineering applications.
Collapse
Affiliation(s)
- Mehdi Farokhi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Mottaghitalab
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran.
| | | | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Jamshid Hadjati
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Chen Y, Yang Z, Liu C, Wang C, Zhao S, Yang J, Sun H, Zhang Z, Kong D, Song C. Synthesis, characterization, and evaluation of paclitaxel loaded in six-arm star-shaped poly(lactic-co-glycolic acid). Int J Nanomedicine 2013; 8:4315-26. [PMID: 24235829 PMCID: PMC3825676 DOI: 10.2147/ijn.s51629] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Star-shaped polymers provide more terminal groups, and are promising for application in drug-delivery systems. Methods A new series of six-arm star-shaped poly(lactic-co-glycolic acid) (6-s-PLGA) was synthesized by ring-opening polymerization. The structure and properties of the 6-s-PLGA were characterized by carbon-13 nuclear magnetic resonance spectroscopy, infrared spectroscopy, gel permeation chromatography, and differential scanning calorimetry. Then, paclitaxel-loaded six-arm star-shaped poly(lactic-co-glycolic acid) nanoparticles (6-s-PLGA-PTX-NPs) were prepared under the conditions optimized by the orthogonal testing. High-performance liquid chromatography was used to analyze the nanoparticles’ encapsulation efficiency and drug-loading capacity, dynamic light scattering was used to determine their size and size distribution, and transmission electron microscopy was used to evaluate their morphology. The release performance of the 6-s-PLGA-PTX-NPs in vitro and the cytostatic effect of 6-s-PLGA-PTX-NPs were investigated in comparison with paclitaxel-loaded linear poly(lactic-co-glycolic acid) nanoparticles (L-PLGA-PTX-NPs). Results The results of carbon-13 nuclear magnetic resonance spectroscopy and infrared spectroscopy suggest that the polymerization was successfully initiated by inositol and confirm the structure of 6-s-PLGA. The molecular weights of a series of 6-s-PLGAs had a ratio corresponding to the molar ratio of raw materials to initiator. Differential scanning calorimetry revealed that the 6-s-PLGA had a low glass transition temperature of 40°C–50°C. The 6-s-PLGA-PTX-NPs were monodispersed with an average diameter of 240.4±6.9 nm in water, which was further confirmed by transmission electron microscopy. The encapsulation efficiency of the 6-s-PLGA-PTX-NPs was higher than that of the L-PLGA-PTX-NPs. In terms of the in vitro release of nanoparticles, paclitaxel (PTX) was released more slowly and more steadily from 6-s-PLGA than from linear poly(lactic-co-glycolic acid). In the cytostatic study, the 6-s-PLGA-PTX-NPs and L-PLGA-PTX-NPs were found to have a similar antiproliferative effect, which indicates durable efficacy due to the slower release of the PTX when loaded in 6-s-PLGA. Conclusion The results suggest that 6-s-PLGA may be promising for application in PTX delivery to enhance sustained antiproliferative therapy.
Collapse
Affiliation(s)
- Yongxia Chen
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, People's Republic of China ; Center for Medical Device Evaluation of Tianjin, Tianjin, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Engineering strategies to control vascular endothelial growth factor stability and levels in a collagen matrix for angiogenesis: the role of heparin sodium salt and the PLGA-based microsphere approach. Acta Biomater 2013; 9:7389-98. [PMID: 23523534 DOI: 10.1016/j.actbio.2013.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/12/2013] [Accepted: 03/13/2013] [Indexed: 02/02/2023]
Abstract
New vessel formation is the result of the complex orchestration of various elements, such as cells, signalling molecules and extracellular matrix (ECM). In order to establish the suitable conditions for an effective cell response, the influence of vascular endothelial growth factor (VEGF) complexation with heparin sodium salt (Hp) on its pro-angiogenic activity has been evaluated by an in vitro capillary-like tube formation assay. VEGF with or without Hp was embedded into collagen gels, and the activated matrices were characterized in terms of VEGF activity and release kinetics. Taking into account the crucial role of Hp in VEGF stability and activity, VEGF/Hp complex was then encapsulated into microspheres based on poly(lactide-co-glycolide) (PLGA), and microsphere properties, VEGF/Hp release kinetics and VEGF in vitro activity over time were evaluated. Integrated microsphere/collagen matrices were developed in order to provide a continuous release of active VEGF/Hp inside the matrix but also a VEGF gradient at the boundary, which is an essential condition for endothelial cell attraction and scaffold invasion. The results confirmed a strong influence of Hp on VEGF configuration and, consequently, on its activity, while the encapsulation of VEGF/Hp complex in PLGA-microspheres guaranteed a sustained release of active VEGF for more than 30days. This paper confirms the importance of VEGF stability and signal presentation to cells for an effective proangiogenic activity and highlights how the combination of two stabilizing approaches, namely VEGF/Hp complexation and entrapment within PLGA-based microspheres, may be a very effective strategy to achieve this goal.
Collapse
|
39
|
Abstract
INTRODUCTION Therapeutic angiogenesis is a strategy of inducing new collateral vessels and stimulating new capillaries that enhance tissue oxygen exchange in ischemic cardiovascular disorders, including acute myocardial infarction, chronic cardiac ischemia, peripheral artery disease and stroke. AREAS COVERED Over the last 10 years, promising results of early clinical trials have generated great expectation on the potential of therapeutic angiogenesis. However, even if large randomized placebo-controlled and double-blinded Phase II clinical trials have confirmed the feasibility, safety and potential effectiveness of therapeutic angiogenesis, they provided very limited evidence of its efficacy in terms of clinical benefit. EXPERT OPINION Results of the latest trials on therapeutic angiogenesis have not provided satisfactory results. Much is still unknown about the optimal delivery of angiogenic factors. Trials using alternative growth factors, dose regimens and methods of delivery are needed to enhance the treatment benefit of therapeutic angiogenesis.
Collapse
Affiliation(s)
- Domenico Ribatti
- University of Bari Medical School, National Cancer Institute, Giovanni Paolo II, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Piazza G. Cesare, 11, Policlinico, 70124 Bari, Italy.
| | | |
Collapse
|
40
|
Vargas GE, Haro Durand LA, Cadena V, Romero M, Mesones RV, Mačković M, Spallek S, Spiecker E, Boccaccini AR, Gorustovich AA. Effect of nano-sized bioactive glass particles on the angiogenic properties of collagen based composites. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:1261-1269. [PMID: 23430337 DOI: 10.1007/s10856-013-4892-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 02/10/2013] [Indexed: 06/01/2023]
Abstract
Angiogenesis is essential for tissue regeneration and repair. A growing body of evidence shows that the use of bioactive glasses (BG) in biomaterial-based tissue engineering (TE) strategies may improve angiogenesis and induce increased vascularization in TE constructs. This work investigated the effect of adding nano-sized BG particles (n-BG) on the angiogenic properties of bovine type I collagen/n-BG composites. Nano-sized (20-30 nm) BG particles of nominally 45S5 Bioglass® composition were used to prepare composite films, which were characterized by scanning electron microscopy (SEM) and transmission electron microscopy (TEM). The in vivo angiogenic response was evaluated using the quail chorioallantoic membrane (CAM) as an model of angiogenesis. At 24 h post-implantation, 10 wt% n-BG containing collagen films stimulated angiogenesis by increasing by 41 % the number of blood vessels branch points. In contrast, composite films containing 20 wt% n-BG were found to inhibit angiogenesis. This experimental study provides the first evidence that addition of a limited concentration of n-BG (10 wt%) to collagen films induces an early angiogenic response making selected collagen/n-BG composites attractive matrices for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Gabriela E Vargas
- Interdisciplinary Materials Group-IESIING-UCASAL, INTECIN UBA-CONICET, A4400EDD, Salta, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Liu F, Yu S, Wang Z, Sun X. Biomimetic construction of large engineered bone using hemoperfusion and cyto-capture in traumatic bone defect. Biores Open Access 2013; 1:247-51. [PMID: 23516672 PMCID: PMC3559229 DOI: 10.1089/biores.2012.0247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Due to lack of blood vessel systems, only a few tissues, such as skin, cartilage, and cornea, have been successfully constructed in vivo. Anticoagulative scaffolds have been used in drug-eluting stent systems both in animal studies and clinical therapies, as in the medicinal leech therapy used to salvage venous-congested microvascular free flaps improved perfusion inspired us to tackle this hurdle in bone tissue engineering. We hypothesize that a combination of bone marrow as the blood supply and a heparin/chitosan-coated acellular bone matrix that acts like hirudin, together with a vacuum-assisted closure therapy system, would provide blood perfusion to the scaffold. Using these methods, a biomimetically engineered bone construct would facilitate clinical translation in bone tissue engineering and offer new therapeutic strategies for reconstructing large bone defects if the hypothesis proves to be practical.
Collapse
Affiliation(s)
- Fei Liu
- Department of Orthopedics, 89th Hospital of People's Liberation Army , Weifang, China
| | | | | | | |
Collapse
|
42
|
Turturro MV, Christenson MC, Larson JC, Young DA, Brey EM, Papavasiliou G. MMP-sensitive PEG diacrylate hydrogels with spatial variations in matrix properties stimulate directional vascular sprout formation. PLoS One 2013; 8:e58897. [PMID: 23554954 PMCID: PMC3595229 DOI: 10.1371/journal.pone.0058897] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 02/08/2013] [Indexed: 01/06/2023] Open
Abstract
The spatial presentation of immobilized extracellular matrix (ECM) cues and matrix mechanical properties play an important role in directed and guided cell behavior and neovascularization. The goal of this work was to explore whether gradients of elastic modulus, immobilized matrix metalloproteinase (MMP)-sensitivity, and YRGDS cell adhesion ligands are capable of directing 3D vascular sprout formation in tissue engineered scaffolds. PEGDA hydrogels were engineered with mechanical and biofunctional gradients using perfusion-based frontal photopolymerization (PBFP). Bulk photopolymerized hydrogels with uniform mechanical properties, degradation, and immobilized biofunctionality served as controls. Gradient hydrogels exhibited an 80.4% decrease in elastic modulus and a 56.2% decrease in immobilized YRGDS. PBFP hydrogels also demonstrated gradients in hydrogel degradation with degradation times ranging from 10-12 hours in the more crosslinked regions to 4-6 hours in less crosslinked regions. An in vitro model of neovascularization, composed of co-culture aggregates of endothelial and smooth muscle cells, was used to evaluate the effect of these gradients on vascular sprout formation. Aggregate invasion in gradient hydrogels occurred bi-directionally with sprout alignment observed in the direction parallel to the gradient while control hydrogels with homogeneous properties resulted in uniform invasion. In PBFP gradient hydrogels, aggregate sprout length was found to be twice as long in the direction parallel to the gradient as compared to the perpendicular direction after three weeks in culture. This directionality was found to be more prominent in gradient regions of increased stiffness, crosslinked MMP-sensitive peptide presentation, and immobilized YRGDS concentration.
Collapse
Affiliation(s)
- Michael V. Turturro
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, United States of America
| | - Megan C. Christenson
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, United States of America
| | - Jeffery C. Larson
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, United States of America
| | - Daniel A. Young
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, United States of America
| | - Eric M. Brey
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, United States of America
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois, United States of America
| | - Georgia Papavasiliou
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
43
|
Abstract
Vascularization is crucial for implantation of engineered tissues in reconstructive surgery. Polypeptides encapsulated in microspheres can be efficiently transported to their site of action and released in a sustained dosage. We evaluated the effect of delivering vascular endothelial growth factor (VEGF)-encapsulated microspheres in a lipoaspirate scaffold on vascularization and tissue survival. The VEGF-loaded (n=6) and empty (n=6) poly(lactic-co-glycolic acid) microspheres in human lipoaspirate and the human lipoaspirate alone (n=6) were injected subcutaneously into the flanks of athymic nude mice. Three mice from each group were killed, and grafts were explanted at weeks 3 and 6. Increases in mass and volume of VEGF samples, as well as decreases in empty and lipoaspirate-only samples, were observed at 3 and 6 weeks, reaching statistical significance at 6 weeks. Hematoxylin and eosin and CD31+ imaging demonstrated significantly greater vascularization in VEGF samples than in both the empty and lipoaspirate-only groups at both 3 and 6 weeks.
Collapse
|
44
|
Baiguera S, Ribatti D. Endothelialization approaches for viable engineered tissues. Angiogenesis 2012; 16:1-14. [PMID: 23010872 DOI: 10.1007/s10456-012-9307-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 09/15/2012] [Indexed: 12/21/2022]
Abstract
One of the main limitation in obtaining thick, 3-dimensional viable engineered constructs is the inability to provide a sufficient and functional blood vessel system essential for the in vitro survival and the in vivo integration of the construct. Different strategies have been proposed to simulate the ingrowth of new blood vessels into engineered tissue, such as the use of growth factors, fabrication scaffold technologies, in vivo prevascularization and cell-based strategies, and it has been demonstrated that endothelial cells play a central role in the neovascularization process and in the control of blood vessel function. In particular, different "environmental" settings (origin, presence of supporting cells, biomaterial surface, presence of hemodynamic forces) strongly influence endothelial cell function, angiogenic potential and the in vivo formation of durable vessels. This review provides an overview of the different techniques developed so far for the vascularization of tissue-engineered constructs (with their advantages and pitfalls), focusing the attention on the recent development in the cell-based vascularization strategy and the in vivo applications.
Collapse
Affiliation(s)
- Silvia Baiguera
- BIOAIRLab, European Center for Thoracic Surgery, University Hospital Careggi, Florence, Italy.
| | | |
Collapse
|
45
|
Functional porous hydrogels to study angiogenesis under the effect of controlled release of vascular endothelial growth factor. Acta Biomater 2012; 8:3294-301. [PMID: 22641106 DOI: 10.1016/j.actbio.2012.05.019] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 05/07/2012] [Accepted: 05/20/2012] [Indexed: 01/11/2023]
Abstract
Angiogenesis occurs through a cascade of events controlled by complex multiple signals that are orchestrated according to specific spatial patterns and temporal sequences. Vascularization is a central issue in most tissue engineering applications. However, only a better insight into spatio-temporal signal presentation can help in controlling and guiding angiogenesis in vivo. To this end, versatile and accessible material platforms are required in order to study angiogenic events in a systematic way. In this work we report a three-dimensional porous polyethylene glycol (PEG) diacrylate hydrogel bioactivated with heparin that is able to deliver vascular endothelial growth factor (VEGF) in a sustained and controlled manner. The efficiency of the material has been tested both in vitro and in vivo. In particular, the VEGF released from the hydrogel induces cell proliferation when tested on HUVECs, retains its bioactivity up to 21days, as demonstrated by Matrigel assay, and, when implanted on a chorion allantoic membrane, the hydrogel shows superior angiogenic potential in stimulating new vessel formation compared with unfunctionalized hydrogels. Moreover, in the light of potential tissue regeneration studies, the proposed hydrogel has been modified with adhesion peptides (RGD) to enable cell colonization. The porous hydrogel reported here can be used as a valid tool to characterize angiogenesis, and, possibly, other biological processes, in different experimental set-ups.
Collapse
|
46
|
Controlled release of vascular endothelial growth factor using poly-lactic-co-glycolic acid microspheres: in vitro characterization and application in polycaprolactone fumarate nerve conduits. Acta Biomater 2012; 8:511-8. [PMID: 22019759 DOI: 10.1016/j.actbio.2011.10.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 09/26/2011] [Accepted: 10/03/2011] [Indexed: 11/21/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a potent angiogenic stimulator. Controlled release of such stimulators may enhance and guide the vascularization process, and when applied in a nerve conduit may play a role in nerve regeneration. We report the fabrication and in vitro characterization of poly-lactic-co-glycolic acid (PLGA) microspheres encapsulating VEGF and the in vivo application of nerve conduits supplemented with VEGF-containing microspheres. PLGA microspheres containing VEGF were prepared by the double emulsion-solvent evaporation technique. This yielded 83.16% of microspheres with a diameter <53 μm. VEGF content measured by ELISA indicated 93.79±10.64% encapsulation efficiency. Release kinetics were characterized by an initial burst release of 67.6±8.25% within the first 24h, followed by consistent release of approximately 0.34% per day for 4 weeks. Bioactivity of the released VEGF was tested by human umbilical vein endothelial cell (HUVEC) proliferation assay. VEGF released at all time points enhanced HUVEC proliferation, confirming that VEGF retained its bioactivity throughout the 4 week time period. When the microsphere delivery system was placed in a biosynthetic nerve scaffold robust nerve regeneration was observed. This study established a novel system for controlled release of growth factors and enables in vivo studies of nerve conduits conditioned with this system.
Collapse
|
47
|
Gomes S, Leonor IB, Mano JF, Reis RL, Kaplan DL. Natural and Genetically Engineered Proteins for Tissue Engineering. Prog Polym Sci 2012; 37:1-17. [PMID: 22058578 PMCID: PMC3207498 DOI: 10.1016/j.progpolymsci.2011.07.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
To overcome the limitations of traditionally used autografts, allografts and, to a lesser extent, synthetic materials, there is the need to develop a new generation of scaffolds with adequate mechanical and structural support, control of cell attachment, migration, proliferation and differentiation and with bio-resorbable features. This suite of properties would allow the body to heal itself at the same rate as implant degradation. Genetic engineering offers a route to this level of control of biomaterial systems. The possibility of expressing biological components in nature and to modify or bioengineer them further, offers a path towards multifunctional biomaterial systems. This includes opportunities to generate new protein sequences, new self-assembling peptides or fusions of different bioactive domains or protein motifs. New protein sequences with tunable properties can be generated that can be used as new biomaterials. In this review we address some of the most frequently used proteins for tissue engineering and biomedical applications and describe the techniques most commonly used to functionalize protein-based biomaterials by combining them with bioactive molecules to enhance biological performance. We also highlight the use of genetic engineering, for protein heterologous expression and the synthesis of new protein-based biopolymers, focusing the advantages of these functionalized biopolymers when compared with their counterparts extracted directly from nature and modified by techniques such as physical adsorption or chemical modification.
Collapse
Affiliation(s)
- Sílvia Gomes
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal
| | | | | | | | | |
Collapse
|
48
|
Singh S, Wu BM, Dunn JCY. Delivery of VEGF using collagen-coated polycaprolactone scaffolds stimulates angiogenesis. J Biomed Mater Res A 2011; 100:720-7. [PMID: 22213643 DOI: 10.1002/jbm.a.34010] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 10/18/2011] [Accepted: 11/01/2011] [Indexed: 11/07/2022]
Abstract
Establishing sufficient vascularization in scaffold remains a challenge for tissue-engineering. To improve angiogenesis, we incorporated vascular endothelial growth factor (VEGF) in collagen-coating over the porous polycaprolactone (PCL) scaffolds. The release kinetics of loaded VEGF from collagen-coated PCL (col-PCL) scaffolds was same as from scaffolds without the collagen. The bioactivity of VEGF delivered by the col-PCL scaffolds was confirmed by human umbilical vein endothelial cell (HUVEC) proliferation and chorioallantoic membrane (CAM) assay. The col-PCL scaffolds were implanted subcutaneously in mice for 7 and 14 days. At day 7, vascularization within scaffolds loaded with VEGF was superior to that in the scaffolds without VEGF. However, the vessel connectivity to host circulatory system was incomplete and restricted to the scaffold edges. At day 14, blood vessels in scaffolds reached density similar to the subcutaneous tissue and were perfusable throughout the implant thickness. Prewashing the scaffolds with saline to remove the unbound growth factor decreased the initial burst release and sustained the VEGF-mediated angiogenesis in vivo. In conclusion, our study demonstrates that physically adsorbed VEGF stimulated angiogenesis in collagen-coated PCL scaffolds.
Collapse
Affiliation(s)
- Shivani Singh
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
49
|
Novosel EC, Kleinhans C, Kluger PJ. Vascularization is the key challenge in tissue engineering. Adv Drug Deliv Rev 2011; 63:300-11. [PMID: 21396416 DOI: 10.1016/j.addr.2011.03.004] [Citation(s) in RCA: 703] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 02/09/2011] [Accepted: 03/02/2011] [Indexed: 12/11/2022]
Abstract
The main limitation in engineering in vitro tissues is the lack of a sufficient blood vessel system - the vascularization. In vivo almost all tissues are supplied by these endothelial cell coated tubular networks. Current strategies to create vascularized tissues are discussed in this review. The first strategy is based on the endothelial cells and their ability to form new vessels known as neoangiogenesis. Herein prevascularization techniques are compared to approaches in which biomolecules, such as growth factors, cytokines, peptides and proteins as well as cells are applied to generate new vessels. The second strategy is focused on scaffold-based techniques. Naturally-derived scaffolds, which contain vessels, are distinguished from synthetically manufactured matrices. Advantages and pitfalls of the approaches to create vascularized tissues in vitro are outlined and feasible future strategies are discussed.
Collapse
|
50
|
Chang CC, Boland ED, Williams SK, Hoying JB. Direct-write bioprinting three-dimensional biohybrid systems for future regenerative therapies. J Biomed Mater Res B Appl Biomater 2011; 98:160-70. [PMID: 21504055 DOI: 10.1002/jbm.b.31831] [Citation(s) in RCA: 269] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 01/19/2011] [Accepted: 02/03/2011] [Indexed: 01/09/2023]
Abstract
Regenerative medicine seeks to repair or replace dysfunctional tissues with engineered biological or biohybrid systems. Current clinical regenerative models utilize simple uniform tissue constructs formed with cells cultured onto biocompatible scaffolds. Future regenerative therapies will require the fabrication of complex three-dimensional constructs containing multiple cell types and extracellular matrices. We believe bioprinting technologies will provide a key role in the design and construction of future engineered tissues for cell-based and regenerative therapies. This review describes the current state-of-the-art bioprinting technologies, focusing on direct-write bioprinting. We describe a number of process and device considerations for successful bioprinting of composite biohybrid constructs. In addition, we have provided baseline direct-write printing parameters for a hydrogel system (Pluronic F127) often used in cardiovascular applications. Direct-write dispensed lines (gels with viscosities ranging from 30 mPa s to greater than 600 × 10⁶ mPa s) were measured following mechanical and pneumatic printing via three commercially available needle sizes (20 ga, 25 ga, and 30 ga). Example patterns containing microvascular cells and isolated microvessel fragments were also bioprinted into composite 3D structures. Cells and vessel fragments remained viable and maintained in vitro behavior after incorporation into biohybrid structures. Direct-write bioprinting of biologicals provides a unique method to design and fabricate complex, multicomponent 3D structures for experimental use. We hope our design insights and baseline parameter descriptions of direct-write bioprinting will provide a useful foundation for colleagues to incorporate this 3D fabrication method into future regenerative therapies.
Collapse
Affiliation(s)
- Carlos C Chang
- Cardiovascular Innovation Institute, 302 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | | | | | | |
Collapse
|