1
|
Huang XY, Zhou XX, Yang H, Xu T, Dao JW, Bian L, Wei DX. Directed osteogenic differentiation of human bone marrow mesenchymal stem cells via sustained release of BMP4 from PBVHx-based nanoparticles. Int J Biol Macromol 2024; 265:130649. [PMID: 38453121 DOI: 10.1016/j.ijbiomac.2024.130649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Bone Morphogenetic Protein 4 (BMP4) is crucial for bone and cartilage tissue regeneration, essential in medical tissue engineering, cosmetology, and aerospace. However, its cost and degradation susceptibility pose significant clinical challenges. To enhance its osteogenic activity while reducing dosage and administration frequency, we developed a novel long-acting BMP4 delivery system using poly(3-hydroxybutyrate-co-3-hydroxyvalerate-co-3-hydroxyhexanoate) (PBVHx) nanoparticles with soybean lecithin-modified BMP4 (sBP-NPs). These nanoparticles promote directed osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) through sustained BMP4 release. sBP-NPs exhibited uniform size (100-200 nm) and surface charges, with higher BMP4 entrapment efficiency (82.63 %) compared to controls. After an initial burst release within 24 h, sBP-NPs achieved 80 % cumulative BMP4 release within 20 days, maintaining levels better than control BP-NPs with unmodified BMP4. Co-incubation and nanoparticle uptake experiments confirmed excellent biocompatibility of sBP-NPs, promoting hBMSC differentiation towards osteogenic lineage with increased expression of type I collagen, calcium deposition, and ALP activity (> 20,000 U/g protein) compared to controls. Moreover, hBMSCs treated with sBP-NPs exhibited heightened expression of osteogenic genetic markers, surpassing control groups. Hence, this innovative strategy of sustained BMP4 release from sBP-NPs holds potential to revolutionize bone regeneration in minimally invasive surgery, medical cosmetology or space environments.
Collapse
Affiliation(s)
- Xiao-Yun Huang
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China; Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Xiao-Xiang Zhou
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China
| | - Hui Yang
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China
| | - Tao Xu
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China
| | - Jin-Wei Dao
- Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong 643002, China
| | - Li Bian
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Dai-Xu Wei
- School of Clinical Medicine, Qujing Medical College, Qujing 655000, China; School of Clinical Medicine, Chengdu University, Chengdu, China; Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong 643002, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| |
Collapse
|
2
|
Khazaei S, Varela-Calviño R, Rad-Malekshahi M, Quattrini F, Jokar S, Rezaei N, Balalaie S, Haririan I, Csaba N, Garcia-Fuentes M. Self-assembled peptide/polymer hybrid nanoplatform for cancer immunostimulating therapies. Drug Deliv Transl Res 2024; 14:455-473. [PMID: 37721693 PMCID: PMC10761384 DOI: 10.1007/s13346-023-01410-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2023] [Indexed: 09/19/2023]
Abstract
Integrating peptide epitopes in self-assembling materials is a successful strategy to obtain nanovaccines with high antigen density and improved efficacy. In this study, self-assembling peptides containing MAGE-A3/PADRE epitopes were designed to generate functional therapeutic nanovaccines. To achieve higher stability, peptide/polymer hybrid nanoparticles were formulated by controlled self-assembly of the engineered peptides. The nanoparticles showed good biocompatibility to both human red blood- and dendritic cells. Incubation of the nanoparticles with immature dendritic cells triggered immune effects that ultimately activated CD8 + cells. The antigen-specific and IgG antibody responses of healthy C57BL/6 mice vaccinated with the nanoparticles were analyzed. The in vivo results indicate a specific response to the nanovaccines, mainly mediated through a cellular pathway. This research indicates that the immunogenicity of peptide epitope vaccines can be effectively enhanced by developing self-assembled peptide-polymer hybrid nanostructures.
Collapse
Affiliation(s)
- Saeedeh Khazaei
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ruben Varela-Calviño
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Federico Quattrini
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Safura Jokar
- Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Noemi Csaba
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
3
|
Karahaliloglu Z, Ercan B, Hazer B. Impregnation of polyethylene terephthalate (PET) grafts with BMP-2 loaded functional nanoparticles for reconstruction of anterior cruciate ligament. J Microencapsul 2023; 40:197-215. [PMID: 36881484 DOI: 10.1080/02652048.2023.2188940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Current artificial ligaments based on polyethylene terephthalate (PET) are associated with some disadvantages due to their hydrophobicity and low biocompatibility. In this study, we aimed to modify the surface of PET using polyethylene glycol (PEG)-terminated polystyrene (PS)-linoleic nanoparticles (PLinaS-g-PEG-NPs). We accomplished that BMP-2 in two different concentrations encapsulated in nanoparticles with an efficiency of 99.71 ± 1.5 and 99.95 ± 2.8%. While the dynamic contact angle of plain PET surface reduced from 116° to 115° after a measurement periods of 10 s, that of PLinaS-g-PEG-NPs modified PET from 80° to 17.5° within 0.35 s. According to in vitro BMP2 release study, BMP-2 was released 13.12 ± 1.76% and 45.47 ± 1.78% from 0.05 and 0.1BMP2-PLinaS-g-PEG-NPs modified PET at the end of 20 days, respectively. Findings from this study revealed that BMP2-PLinaS-g-PEG-NPs has a great potential to improve the artificial PET ligaments, and could be effectively applied for ACL reconstruction.
Collapse
Affiliation(s)
| | - Batur Ercan
- Department of Metallurgical and Materials Engineering, Middle East Technical University, Çankaya, Ankara, Turkey
- Biomedical Engineering Program, Middle East Technical University, Çankaya, Ankara, Turkey
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Çankaya, Ankara, Turkey
| | - Baki Hazer
- Department of Aircraft Airframe Engine Maintenance, Kapadokya University, Ürgüp, Nevsehir, Turkey
- Department of Chemistry, Bulent Ecevit University, Zonguldak, Turkey
- Department of Nanotechnology Engineering, Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
4
|
Hoseinzadeh A, Ghoddusi Johari H, Anbardar MH, Tayebi L, Vafa E, Abbasi M, Vaez A, Golchin A, Amani AM, Jangjou A. Effective treatment of intractable diseases using nanoparticles to interfere with vascular supply and angiogenic process. Eur J Med Res 2022; 27:232. [PMID: 36333816 PMCID: PMC9636835 DOI: 10.1186/s40001-022-00833-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is a vital biological process involving blood vessels forming from pre-existing vascular systems. This process contributes to various physiological activities, including embryonic development, hair growth, ovulation, menstruation, and the repair and regeneration of damaged tissue. On the other hand, it is essential in treating a wide range of pathological diseases, such as cardiovascular and ischemic diseases, rheumatoid arthritis, malignancies, ophthalmic and retinal diseases, and other chronic conditions. These diseases and disorders are frequently treated by regulating angiogenesis by utilizing a variety of pro-angiogenic or anti-angiogenic agents or molecules by stimulating or suppressing this complicated process, respectively. Nevertheless, many traditional angiogenic therapy techniques suffer from a lack of ability to achieve the intended therapeutic impact because of various constraints. These disadvantages include limited bioavailability, drug resistance, fast elimination, increased price, nonspecificity, and adverse effects. As a result, it is an excellent time for developing various pro- and anti-angiogenic substances that might circumvent the abovementioned restrictions, followed by their efficient use in treating disorders associated with angiogenesis. In recent years, significant progress has been made in different fields of medicine and biology, including therapeutic angiogenesis. Around the world, a multitude of research groups investigated several inorganic or organic nanoparticles (NPs) that had the potential to effectively modify the angiogenesis processes by either enhancing or suppressing the process. Many studies into the processes behind NP-mediated angiogenesis are well described. In this article, we also cover the application of NPs to encourage tissue vascularization as well as their angiogenic and anti-angiogenic effects in the treatment of several disorders, including bone regeneration, peripheral vascular disease, diabetic retinopathy, ischemic stroke, rheumatoid arthritis, post-ischemic cardiovascular injury, age-related macular degeneration, diabetic retinopathy, gene delivery-based angiogenic therapy, protein delivery-based angiogenic therapy, stem cell angiogenic therapy, and diabetic retinopathy, cancer that may benefit from the behavior of the nanostructures in the vascular system throughout the body. In addition, the accompanying difficulties and potential future applications of NPs in treating angiogenesis-related diseases and antiangiogenic therapies are discussed.
Collapse
Affiliation(s)
- Ahmad Hoseinzadeh
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Ghoddusi Johari
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Ehsan Vafa
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Golchin
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Poloxamer-Based Scaffolds for Tissue Engineering Applications: A Review. Gels 2022; 8:gels8060360. [PMID: 35735704 PMCID: PMC9222596 DOI: 10.3390/gels8060360] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/28/2022] [Accepted: 06/01/2022] [Indexed: 12/28/2022] Open
Abstract
Poloxamer is a triblock copolymer with amphiphilicity and reversible thermal responsiveness and has wide application prospects in biomedical applications owing to its multifunctional properties. Poloxamer hydrogels play a crucial role in the field of tissue engineering and have been regarded as injectable scaffolds for loading cells or growth factors (GFs) in the last few years. Hydrogel micelles can maintain the integrity and stability of cells and GFs and form an appropriate vascular network at the application site, thus creating an appropriate microenvironment for cell growth, nerve growth, or bone integration. The injectability and low toxicity of poloxamer hydrogels make them a noninvasive method. In addition, they can also be good candidates for bio-inks, the raw material for three-dimensional (3D) printing. However, the potential of poloxamer hydrogels has not been fully explored owing to the complex biological challenges. In this review, the latest progress and cutting-edge research of poloxamer-based scaffolds in different fields of application such as the bone, vascular, cartilage, skin, nervous system, and organs in tissue engineering and 3D printing are reviewed, and the important roles of poloxamers in tissue engineering scaffolds are discussed in depth.
Collapse
|
6
|
Wang J, Xiao L, Wang W, Zhang D, Ma Y, Zhang Y, Wang X. The Auxiliary Role of Heparin in Bone Regeneration and its Application in Bone Substitute Materials. Front Bioeng Biotechnol 2022; 10:837172. [PMID: 35646879 PMCID: PMC9133562 DOI: 10.3389/fbioe.2022.837172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Bone regeneration in large segmental defects depends on the action of osteoblasts and the ingrowth of new blood vessels. Therefore, it is important to promote the release of osteogenic/angiogenic growth factors. Since the discovery of heparin, its anticoagulant, anti-inflammatory, and anticancer functions have been extensively studied for over a century. Although the application of heparin is widely used in the orthopedic field, its auxiliary effect on bone regeneration is yet to be unveiled. Specifically, approximately one-third of the transforming growth factor (TGF) superfamily is bound to heparin and heparan sulfate, among which TGF-β1, TGF-β2, and bone morphogenetic protein (BMP) are the most common growth factors used. In addition, heparin can also improve the delivery and retention of BMP-2 in vivo promoting the healing of large bone defects at hyper physiological doses. In blood vessel formation, heparin still plays an integral part of fracture healing by cooperating with the platelet-derived growth factor (PDGF). Importantly, since heparin binds to growth factors and release components in nanomaterials, it can significantly facilitate the controlled release and retention of growth factors [such as fibroblast growth factor (FGF), BMP, and PDGF] in vivo. Consequently, the knowledge of scaffolds or delivery systems composed of heparin and different biomaterials (including organic, inorganic, metal, and natural polymers) is vital for material-guided bone regeneration research. This study systematically reviews the structural properties and auxiliary functions of heparin, with an emphasis on bone regeneration and its application in biomaterials under physiological conditions.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| | - Weiqun Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| |
Collapse
|
7
|
Vijayan A, C L V, Kumar GSV. Dual growth factor entrapped nanoparticle enriched alginate wafer-based delivery system for suppurating wounds. Int J Biol Macromol 2022; 208:172-181. [PMID: 35304195 DOI: 10.1016/j.ijbiomac.2022.03.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 01/13/2023]
Abstract
We have investigated the wound healing efficiency of calcium alginate wafer embedded with growth factor entrapped PLGA nanoparticle. Herein, vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) entrapped PLGA nanoparticles were synthesized and embedded in a sodium alginate gel by freeze-drying technique. The synthesized dressing exhibited a high degree of swelling and appropriate porosity. The scaffold was characterized by Scanning Electron Microscopy (SEM) showing a highly porous morphology. Also, incorporation of growth factor loaded nanoparticles in a wafer-based delivery system resulted in localized growth factor delivery at the site of the wound in a sustained manner. The biocompatibility of the scaffold was evaluated by MTT assay, which showed a higher cell proliferation in the proposed scaffold as compared to the control. In vivo wound healing efficiency of the scaffold was evaluated using a full thickness murine wound model, which showed improved re-epithelialization, collagen deposition, and angiogenesis. These results suggest the use of the scaffold as a promising wound dressing material.
Collapse
Affiliation(s)
- Amritha Vijayan
- Nano Drug Delivery Systems (NDDS), Bio-Innovation Centre (BIC), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala 695014, India; Research Centre, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Vipin C L
- Nano Drug Delivery Systems (NDDS), Bio-Innovation Centre (BIC), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala 695014, India; Research Centre, University of Kerala, Thiruvananthapuram, Kerala, India
| | - G S Vinod Kumar
- Nano Drug Delivery Systems (NDDS), Bio-Innovation Centre (BIC), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala 695014, India.
| |
Collapse
|
8
|
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
|
9
|
Cortés H, Hernández-Parra H, Bernal-Chávez SA, Prado-Audelo MLD, Caballero-Florán IH, Borbolla-Jiménez FV, González-Torres M, Magaña JJ, Leyva-Gómez G. Non-Ionic Surfactants for Stabilization of Polymeric Nanoparticles for Biomedical Uses. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3197. [PMID: 34200640 PMCID: PMC8226872 DOI: 10.3390/ma14123197] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022]
Abstract
Surfactants are essential in the manufacture of polymeric nanoparticles by emulsion formation methods and to preserve the stability of carriers in liquid media. The deposition of non-ionic surfactants at the interface allows a considerable reduction of the globule of the emulsion with high biocompatibility and the possibility of oscillating the final sizes in a wide nanometric range. Therefore, this review presents an analysis of the three principal non-ionic surfactants utilized in the manufacture of polymeric nanoparticles; polysorbates, poly(vinyl alcohol), and poloxamers. We included a section on general properties and uses and a comprehensive compilation of formulations with each principal non-ionic surfactant. Then, we highlight a section on the interaction of non-ionic surfactants with biological barriers to emphasize that the function of surfactants is not limited to stabilizing the dispersion of nanoparticles and has a broad impact on pharmacokinetics. Finally, the last section corresponds to a recommendation in the experimental approach for choosing a surfactant applying the systematic methodology of Quality by Design.
Collapse
Affiliation(s)
- Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico; (H.C.); (F.V.B.-J.)
| | - Héctor Hernández-Parra
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico; (H.H.-P.); (I.H.C.-F.)
| | - Sergio A. Bernal-Chávez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - María L. Del Prado-Audelo
- Escuela de Ingeniería y Ciencias, Departamento de Bioingeniería, Tecnológico de Monterrey Campus Ciudad de México, CDMX, Ciudad de México 14380, Mexico;
| | - Isaac H. Caballero-Florán
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico; (H.H.-P.); (I.H.C.-F.)
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Fabiola V. Borbolla-Jiménez
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico; (H.C.); (F.V.B.-J.)
| | - Maykel González-Torres
- CONACyT-Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico;
| | - Jonathan J. Magaña
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico; (H.C.); (F.V.B.-J.)
- Escuela de Ingeniería y Ciencias, Departamento de Bioingeniería, Tecnológico de Monterrey Campus Ciudad de México, CDMX, Ciudad de México 14380, Mexico;
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| |
Collapse
|
10
|
Zhao XH, Peng XL, Gong HL, Wei DX. Osteogenic differentiation system based on biopolymer nanoparticles for stem cells in simulated microgravity. Biomed Mater 2021; 16. [PMID: 33631731 DOI: 10.1088/1748-605x/abe9d1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022]
Abstract
An efficient long-term intracellular growth factor release system in simulated microgravity for osteogenic differentiation was prepared based on polylactic acid (PLA) and polyhydroxyalkanoate (PHA) nanoparticles for loading of bone morphogenetic protein 2 (BMP2) and bone morphogenetic protein 7 (BMP7) (defined as sB2-PLA-NP and sB7-PHA-NP), respectively, associated with osteogenic differentiation of human adipose derived stem cells (hADSCs). On account of soybean lecithin (SL) as biosurfactants, sB2-PLA-NPs and sB7-PHA-NPs had a high encapsulation efficiency (>80%) of BMPs and uniform small size (<100 nm), and showed different slow-release to provide BMP2 in early stage and BMP7 in late stages of osteogenic differentiation within 20 days, due to degradation rate of PLA and PHA in cells. After uptake into hADSCs, by comparison with single sB2-PLA-NP or sB7-PHA-NP, the Mixture NPs, compound of sB2-PLA-NP and sB7-PHA-NP with a mass ratio of 1:1, can well-promote ALP activity, expression of OPN and upregulated related osteo-genes. Directed osteo-differentiation of Mixture NPs was similar to result of sustained free-BMP2 and BMP7-supplying (sFree-B2&B7) in simulated microgravity, which demonstrated the reliability and stability of Mixture NPs as a long-term osteogenic differentiation system in space medicine and biology in future.
Collapse
Affiliation(s)
- Xiao-Hong Zhao
- Northwest University, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of life sciences and medicine, Xi'an, Shaanxi, 710069, CHINA
| | - Xue-Liang Peng
- Northwest University, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of life sciences and medicine, Xi'an, Shaanxi, 710069, CHINA
| | - Hai-Lun Gong
- Northwest University, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of life sciences and medicine, Xi'an, Shaanxi, 710069, CHINA
| | - Dai-Xu Wei
- Northwest University, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of life sciences and medicine, Xi'an, Shaanxi, 710069, CHINA
| |
Collapse
|
11
|
Engineering microenvironment of biodegradable polyester systems for drug stability and release control. Ther Deliv 2021; 12:37-54. [PMID: 33397135 DOI: 10.4155/tde-2020-0113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Polymeric systems made of poly(lactic acid) or poly(lactic-co-glycolic acid) are widely used for long-term delivery of small and large molecules. The advantages of poly(lactic acid)/poly(lactic-co-glycolic acid) systems include biodegradability, safety and a long history of use in US FDA-approved products. However, as drugs delivered by the polymeric systems and their applications become more diverse, the significance of microenvironment change of degrading systems on long-term drug stability and release kinetics has gained renewed attention. In this review, we discuss various issues experienced with acidifying microenvironment of biodegradable polymer systems and approaches to overcome the detrimental effects of polymer degradation on drug stability and release control.
Collapse
|
12
|
Chen R, Yu J, Gong HL, Jiang Y, Xue M, Xu N, Wei DX, Shi C. An easy long-acting BMP7 release system based on biopolymer nanoparticles for inducing osteogenic differentiation of adipose mesenchymal stem cells. J Tissue Eng Regen Med 2020; 14:964-972. [PMID: 32441466 DOI: 10.1002/term.3070] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 12/16/2022]
Abstract
In contrast to the early acting bone morphogenetic protein 2, bone morphogenetic protein 7 (BMP7) plays a decisive role mainly in the late stages of bone formation. To overcome deactivation and degradation of expensive BMP7, we designed a novel long-acting BMP7 release system based on poly(3-hydroxybutyrate-co-4-hydroxybutyrate) (P34HB) nanoparticles to enable the induction of osteogenic differentiation in human adipose mesenchymal stem cells (ADSCs). In order to improve the encapsulation efficiency of BMP7 and avoid damage by organic solvents, BMP7 was modified and protected using the biosurfactant soybean lecithin. In an in vitro test, BMP7-soybean lecithin-P34HB nanoparticles (BMP7-SPNPs) showed a short initial burst of BMP7 release during the first 24h, followed by a steady increase to a cumulative 80% release in 20days. Compared with the rapid release of control P34HB nanoparticles without soybean phospholipids loaded with BMP7 without soybean lecithin, BMP7-SPNPs significantly reduced the initial burst of BMP7 release and stabilized the content of BMP7 to allow long-term osteogenic differentiation during the late phase of bone development. Human ADSCs treated with BMP7-SPNPs showed higher alkaline phosphatase activity and higher expression levels of genetic markers of osteogenic differentiation compared with the control group. Thus, the results indicate that BMP7-SPNPs can be used as a rapid and long-acting BMP7 delivery system for osteogenic differentiation.
Collapse
Affiliation(s)
- Rui Chen
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jiangming Yu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hai-Lun Gong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuquan Jiang
- Department of Orthopaedics, Joint Logistic Support Force NO.925 Hospital of PLA, Guiyang, China
| | - Mintao Xue
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ning Xu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Dai-Xu Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Changgui Shi
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
13
|
Han Y, Gong T, Zhang C, Dissanayaka WL. HIF-1α Stabilization Enhances Angio-/Vasculogenic Properties of SHED. J Dent Res 2020; 99:804-812. [PMID: 32298193 DOI: 10.1177/0022034520912190] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The outcome of regenerative procedures could be augmented by enhancing the biological performances of stem cells prior to their transplantation. The current study aimed to investigate whether hypoxic preconditioning through stabilization of hypoxia-inducible factor 1α (HIF-1α) could enhance the angio-/vasculogenic properties of stem cells from human exfoliated deciduous teeth (SHED). HIF-1α expression in SHED under normoxia was stabilized by silencing the expression of prolyl hydroxylase domain-containing protein 2 (PHD2) via lentiviral small hairpin RNA. This in turn significantly increased the expression of an angiogenic factor: vascular endothelial growth factor. Conditioned medium of HIF-1α-stabilized SHED increased the migration and proliferation of human umbilical vein endothelial cells (HUVECs), indicating enhanced paracrine signaling of SHED following PHD2 knockdown (P < 0.05). Furthermore, the coculture of HIF-1α-stabilized SHED with HUVECs directly and in fibrin beads demonstrated significantly longer vascular sprouts through juxtacrine and paracrine effects (P < 0.05). When HIF-1α-stabilized SHED were added to a preformed HUVEC vascular tube network on Matrigel, it not only stabilized the vessels, as shown by the increased thickness (P < 0.05) and junctional area (P < 0.01) of tubes, but also gave rise to new sprouting (P < 0.01). This observation, with the morphologic changes and increased CD31 expression, suggested that HIF-1α stabilization enhanced the endothelial differentiation capacity of SHED through autocrine signaling. In vivo Matrigel plug assay demonstrated that HIF-1α-stabilized SHED alone could give rise to a vasculature that was significantly higher than that of control SHED ± HUVECs and similar to that of HIF-1α-stabilized SHED + HUVECs. In addition to vasculogenesis by endothelial differentiation, HIF-1α-stabilized SHED recruited host blood vessels into the implant by exerting a significant paracrine effect. Taken together, our results confirmed that HIF-1α-stabilized SHED could replace the function of HUVECs and act as the sole cell source of vascularization. Thus, targeting PHD2 to stabilize HIF-1α expression is an appealing strategy that enables the use of a single cell source for achieving vascularized tissue regeneration.
Collapse
Affiliation(s)
- Y Han
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR
| | - T Gong
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR
| | - C Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR
| | - W L Dissanayaka
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR
| |
Collapse
|
14
|
Formulation, Colloidal Characterization, and In Vitro Biological Effect of BMP-2 Loaded PLGA Nanoparticles for Bone Regeneration. Pharmaceutics 2019; 11:pharmaceutics11080388. [PMID: 31382552 PMCID: PMC6723283 DOI: 10.3390/pharmaceutics11080388] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/15/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) based on the polymer poly (lactide-co-glycolide) acid (PLGA) have been widely studied in developing delivery systems for drugs and therapeutic biomolecules, due to the biocompatible and biodegradable properties of the PLGA. In this work, a synthesis method for bone morphogenetic protein (BMP-2)-loaded PLGA NPs was developed and optimized, in order to carry out and control the release of BMP-2, based on the double-emulsion (water/oil/water, W/O/W) solvent evaporation technique. The polymeric surfactant Pluronic F68 was used in the synthesis procedure, as it is known to have an effect on the reduction of the size of the NPs, the enhancement of their stability, and the protection of the encapsulated biomolecule. Spherical solid polymeric NPs were synthesized, showing a reproducible multimodal size distribution, with diameters between 100 and 500 nm. This size range appears to allow the protein to act on the cell surface and at the cytoplasm level. The effect of carrying BMP-2 co-adsorbed with bovine serum albumin on the NP surface was analyzed. The colloidal properties of these systems (morphology by SEM, hydrodynamic size, electrophoretic mobility, temporal stability, protein encapsulation, and short-term release profile) were studied. The effect of both BMP2-loaded NPs on the proliferation, migration, and osteogenic differentiation of mesenchymal stromal cells from human alveolar bone (ABSC) was also analyzed in vitro.
Collapse
|
15
|
Martins C, Sousa F, Araújo F, Sarmento B. Functionalizing PLGA and PLGA Derivatives for Drug Delivery and Tissue Regeneration Applications. Adv Healthc Mater 2018; 7. [PMID: 29171928 DOI: 10.1002/adhm.201701035] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/27/2017] [Indexed: 12/16/2022]
Abstract
Poly(lactic-co-glycolic) acid (PLGA) is one of the most versatile biomedical polymers, already approved by regulatory authorities to be used in human research and clinics. Due to its valuable characteristics, PLGA can be tailored to acquire desirable features for control bioactive payload or scaffold matrix. Moreover, its chemical modification with other polymers or bioconjugation with molecules may render PLGA with functional properties that make it the Holy Grail among the synthetic polymers to be applied in the biomedical field. In this review, the physical-chemical properties of PLGA, its synthesis, degradation, and conjugation with other polymers or molecules are revised in detail, as well as its applications in drug delivery and regeneration fields. A particular focus is given to successful examples of products already on the market or at the late stages of trials, reinforcing the potential of this polymer in the biomedical field.
Collapse
Affiliation(s)
- Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
| | - Flávia Sousa
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- ICBAS - Instituto Ciências Biomédicas Abel Salazar; Universidade do Porto; Rua de Jorge Viterbo Ferreira 228 4050-313 Porto Portugal
| | - Francisca Araújo
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- INEB - Instituto de Engenharia Biomédica; Universidade do Porto; Rua Alfredo Allen 208 4200-393 Porto Portugal
- CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde; Rua Central de Gandra 1317 4585-116 Gandra Portugal
| |
Collapse
|
16
|
Santalices I, Gonella A, Torres D, Alonso MJ. Advances on the formulation of proteins using nanotechnologies. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
17
|
Preparation of poly(lactic- co -glycolic acid) and chitosan composite nanocarriers via electrostatic self assembly for oral delivery of insulin. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 78:420-428. [DOI: 10.1016/j.msec.2017.04.113] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 10/19/2022]
|
18
|
Ortega-Oller I, Del Castillo-Santaella T, Padial-Molina M, Galindo-Moreno P, Jódar-Reyes AB, Peula-García JM. Dual delivery nanosystem for biomolecules. Formulation, characterization, and in vitro release. Colloids Surf B Biointerfaces 2017; 159:586-595. [PMID: 28854415 DOI: 10.1016/j.colsurfb.2017.08.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/18/2017] [Accepted: 08/17/2017] [Indexed: 12/18/2022]
Abstract
Because of the biocompatible and biodegradable properties of poly (lactic-co-glycolic acid) (PLGA), nanoparticles (NPs) based on this polymer have been widely studied for drug/biomolecule delivery and long-term sustained-release. In this work, two different formulation methods for lysozyme-loaded PLGA NPs have been developed and optimized based on the double-emulsion (water/oil/water, W/O/W) solvent evaporation technique. They differ mainly in the phase in which the surfactant (Pluronic® F68) is added: water (W-F68) and oil (O-F68). The colloidal properties of these systems (morphology by SEM and STEM, hydrodynamic size by DLS and NTA, electrophoretic mobility, temporal stability in different media, protein encapsulation, release, and bioactivity) have been analyzed. The interaction surfactant-protein depending on the formulation procedure has been characterized by surface tension and dilatational rheology. Finally, cellular uptake by human mesenchymal stromal cells and cytotoxicity for both systems have been analyzed. Spherical hard NPs are made by the two methods However, in one case, they are monodisperse with diameters of around 120nm (O-F68), and in the other case, a polydisperse system of NPs with diameters between 100 and 500nm is found (W-F68). Protein encapsulation efficiency, release and bioactivity are maintained better by the W-F68 formulation method. This multimodal system is found to be a promising "dual delivery" system for encapsulating hydrophilic proteins with strong biological activity at the cell-surface and cytoplasmic levels.
Collapse
Affiliation(s)
| | | | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, University of Granada, Granada, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, University of Granada, Granada, Spain
| | - Ana Belén Jódar-Reyes
- Biocolloid and Fluid Physics Group, Department of Applied Physics, University of Granada, 18071 Granada, Spain
| | - José Manuel Peula-García
- Biocolloid and Fluid Physics Group, Department of Applied Physics, University of Granada, 18071 Granada, Spain; Department of Applied Physics II, University of Malaga, 29071 Malaga, Spain.
| |
Collapse
|
19
|
López-Cebral R, Civantos A, Ramos V, Seijo B, López-Lacomba JL, Sanz-Casado JV, Sanchez A. Gellan gum based physical hydrogels incorporating highly valuable endogen molecules and associating BMP-2 as bone formation platforms. Carbohydr Polym 2017; 167:345-355. [PMID: 28433171 DOI: 10.1016/j.carbpol.2017.03.049] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 12/20/2022]
Abstract
Physical hydrogels have been designed for a double purpose: as growth factor delivery systems and as scaffolds to support cell colonization and formation of new bone. Specifically, the polysaccharide gellan gum and the ubiquitous endogenous molecules chondroitin, albumin and spermidine have been used as exclusive components of these hydrogels. The mild ionotropic gelation technique was used to preserve the bioactivity of the selected growth factor, rhBMP-2. In vitro tests demonstrated the effective delivery of rhBMP-2 in its bioactive form. In vivo experiments performed in the muscle tissue of Wistar rats provided a proof of concept of the ability of the developed platforms to elicit new bone formation. Furthermore, this biological effect was better than that of a commercial formulation currently used for regenerative purposes, confirming the potential of these hydrogels as new and innovative growth factor delivery platforms and scaffolds for regenerative medicine applications.
Collapse
Affiliation(s)
- Rita López-Cebral
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Campus Sur, 15782 Santiago de Compostela, Spain
| | - Ana Civantos
- Institute of Biofunctional Studies, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Viviana Ramos
- Institute of Biofunctional Studies, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Begoña Seijo
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Campus Sur, 15782 Santiago de Compostela, Spain; Genetics and Biology of the Development of Kidney Diseases Unit, Sanitary Research Institute (IDIS) of the University Hospital Complex of Santiago de Compostela (CHUS), Travesía da Choupana, s/n, 15706 Santiago de Compostela, Spain
| | - José Luis López-Lacomba
- Institute of Biofunctional Studies, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | | | - Alejandro Sanchez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela (USC), Campus Sur, 15782 Santiago de Compostela, Spain; Genetics and Biology of the Development of Kidney Diseases Unit, Sanitary Research Institute (IDIS) of the University Hospital Complex of Santiago de Compostela (CHUS), Travesía da Choupana, s/n, 15706 Santiago de Compostela, Spain.
| |
Collapse
|
20
|
Xu HL, Yu WZ, Lu CT, Li XK, Zhao YZ. Delivery of growth factor-based therapeutics in vascular diseases: Challenges and strategies. Biotechnol J 2017; 12. [PMID: 28296342 DOI: 10.1002/biot.201600243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 01/27/2017] [Accepted: 02/09/2017] [Indexed: 12/18/2022]
Abstract
Either cardiovascular or peripheral vascular diseases have become the major cause of morbidity and mortality worldwide. Recently, growth factors therapeutics, whatever administrated in form of exogenous growth factors or their relevant genes have been discovered to be an effective strategy for the prevention and therapy of vascular diseases, because of their promoting angiogenesis. Besides, as an alternative, stem cell-based therapy has been also developed in view of their paracrine-mediated effect or ability of differentiation toward angiogenesis-related cells under assistance of growth factors. Despite of being specific and potent, no matter growth factors or stem cells-based therapy, their full clinical transformation is limited from bench to bedside. In this review, the potential choices of therapeutic modes based on types of different growth factors or stem cells were firstly summarized for vascular diseases. The confronted various challenges such as lack of non-invasive delivery method, the physiochemical challenge, the short half-life time, and poor cell survival, were carefully analyzed for these therapeutic modes. Various strategies to overcome these limitations are put forward from the perspective of drug delivery. The expertised design of a suitable delivery form will undoubtedly provide valuable insight into their clinical application in the regenerative medicine.
Collapse
Affiliation(s)
- He-Lin Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Wen-Ze Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Cui-Tao Lu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Xiao-Kun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China.,Collaborative Innovation Center of Biomedical Science by Wenzhou University & Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| |
Collapse
|
21
|
Zhang HX, Zhang XP, Xiao GY, Hou Y, Cheng L, Si M, Wang SS, Li YH, Nie L. In vitro and in vivo evaluation of calcium phosphate composite scaffolds containing BMP-VEGF loaded PLGA microspheres for the treatment of avascular necrosis of the femoral head. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 60:298-307. [DOI: 10.1016/j.msec.2015.11.055] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/24/2015] [Accepted: 11/20/2015] [Indexed: 10/22/2022]
|
22
|
Almubarak S, Nethercott H, Freeberg M, Beaudon C, Jha A, Jackson W, Marcucio R, Miclau T, Healy K, Bahney C. Tissue engineering strategies for promoting vascularized bone regeneration. Bone 2016; 83:197-209. [PMID: 26608518 PMCID: PMC4911893 DOI: 10.1016/j.bone.2015.11.011] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/06/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023]
Abstract
This review focuses on current tissue engineering strategies for promoting vascularized bone regeneration. We review the role of angiogenic growth factors in promoting vascularized bone regeneration and discuss the different therapeutic strategies for controlled/sustained growth factor delivery. Next, we address the therapeutic uses of stem cells in vascularized bone regeneration. Specifically, this review addresses the concept of co-culture using osteogenic and vasculogenic stem cells, and how adipose derived stem cells compare to bone marrow derived mesenchymal stem cells in the promotion of angiogenesis. We conclude this review with a discussion of a novel approach to bone regeneration through a cartilage intermediate, and discuss why it has the potential to be more effective than traditional bone grafting methods.
Collapse
Affiliation(s)
- Sarah Almubarak
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Hubert Nethercott
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Marie Freeberg
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Caroline Beaudon
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Amit Jha
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Wesley Jackson
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Kevin Healy
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Chelsea Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States.
| |
Collapse
|
23
|
Bone Regeneration from PLGA Micro-Nanoparticles. BIOMED RESEARCH INTERNATIONAL 2015; 2015:415289. [PMID: 26509156 PMCID: PMC4609778 DOI: 10.1155/2015/415289] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/04/2015] [Indexed: 12/19/2022]
Abstract
Poly-lactic-co-glycolic acid (PLGA) is one of the most widely used synthetic polymers for development of delivery systems for drugs and therapeutic biomolecules and as component of tissue engineering applications. Its properties and versatility allow it to be a reference polymer in manufacturing of nano- and microparticles to encapsulate and deliver a wide variety of hydrophobic and hydrophilic molecules. It additionally facilitates and extends its use to encapsulate biomolecules such as proteins or nucleic acids that can be released in a controlled way. This review focuses on the use of nano/microparticles of PLGA as a delivery system of one of the most commonly used growth factors in bone tissue engineering, the bone morphogenetic protein 2 (BMP2). Thus, all the needed requirements to reach a controlled delivery of BMP2 using PLGA particles as a main component have been examined. The problems and solutions for the adequate development of this system with a great potential in cell differentiation and proliferation processes under a bone regenerative point of view are discussed.
Collapse
|
24
|
Elliott Donaghue I, Shoichet MS. Controlled release of bioactive PDGF-AA from a hydrogel/nanoparticle composite. Acta Biomater 2015; 25:35-42. [PMID: 26257128 DOI: 10.1016/j.actbio.2015.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/28/2015] [Accepted: 08/05/2015] [Indexed: 12/23/2022]
Abstract
Polymer excipients, such as low molar mass poly(ethylene glycol) (PEG), have shown contradictory effects on protein stability when co-encapsulated in polymeric nanoparticles. To gain further insight into these effects, platelet-derived growth factor (PDGF-AA) was encapsulated in polymeric nanoparticles with vs. without PEG. PDGF-AA is a particularly compelling protein, as it has been demonstrated to promote cell survival and induce the oligodendrocyte differentiation of neural stem/progenitor cells (NSPCs) both in vitro and in vivo. Here we show, for the first time, the controlled release of bioactive PDGF-AA from an injectable nanoparticle/hydrogel drug delivery system (DDS). PDGF-AA was encapsulated, with high efficiency, in poly(lactide-co-glycolide) nanoparticles, and its release from the drug delivery system was followed over 21 d. Interestingly, the co-encapsulation of low molecular weight poly(ethylene glycol) increased the PDGF-AA loading but, unexpectedly, accelerated the aggregation of PDGF-AA, resulting in reduced activity and detection by enzyme-linked immunosorbent assay (ELISA). In the absence of PEG, released PDGF-AA remained bioactive as demonstrated with NSPC oligodendrocyte differentiation, similar to positive controls, and significantly different from untreated controls. This work presents a novel delivery method for differentiation factors, such as PDGF-AA, and provides insights into the contradictory effects reported in the literature of excipients, such as PEG, on the loading and release of proteins from polymeric nanoparticles. STATEMENT OF SIGNIFICANCE Previously, the polymer poly(ethylene glycol) (PEG) has been used in many biomaterials applications, from surface coatings to the encapsulation of proteins. In this work, we demonstrate that, unexpectedly, low molecular weight PEG has a deleterious effect on the release of the encapsulated protein platelet-derived growth factor AA (PDGF-AA). We also demonstrate release of bioactive PDGF-AA (in the absence of PEG). Specifically, we demonstrate the differentiation of neural stem and progenitor cells to oligodendrocytes, similar to what is observed with the addition of fresh PDGFAA. A differentiated oligodendrocyte population is a key strategy in central nervous system regeneration. This work is the first demonstration of controlled PDGF-AA release, and also brings new insights to the broader field of protein encapsulation.
Collapse
Affiliation(s)
- Irja Elliott Donaghue
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada; Department of Chemistry, University of Toronto, Canada.
| |
Collapse
|
25
|
Tu C, Das S, Baker AB, Zoldan J, Suggs LJ. Nanoscale strategies: treatment for peripheral vascular disease and critical limb ischemia. ACS NANO 2015; 9:3436-52. [PMID: 25844518 PMCID: PMC5494973 DOI: 10.1021/nn507269g] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Peripheral vascular disease (PVD) is one of the most prevalent vascular diseases in the U.S. afflicting an estimated 8 million people. Obstruction of peripheral arteries leads to insufficient nutrients and oxygen supply to extremities, which, if not treated properly, can potentially give rise to a severe condition called critical limb ischemia (CLI). CLI is associated with extremely high morbidities and mortalities. Conventional treatments such as angioplasty, atherectomy, stent implantation and bypass surgery have achieved some success in treating localized macrovascular disease but are limited by their invasiveness. An emerging alternative is the use of growth factor (delivered as genes or proteins) and cell therapy for PVD treatment. By delivering growth factors or cells to the ischemic tissue, one can stimulate the regeneration of functional vasculature network locally, re-perfuse the ischemic tissue, and thus salvage the limb. Here we review recent advance in nanomaterials, and discuss how their application can improve and facilitate growth factor or cell therapies. Specifically, nanoparticles (NPs) can serve as drug carrier and target to ischemic tissues and achieve localized and sustained release of pro-angiogenic proteins. As nonviral vectors, NPs can greatly enhance the transfection of target cells with pro-angiogenic genes with relatively fewer safety concern. Further, NPs may also be used in combination with cell therapy to enhance cell retention, cell survival and secretion of angiogenic factors. Lastly, nano/micro fibrous vascular grafts can be engineered to better mimic the structure and composition of native vessels, and hopefully overcome many complications/limitations associated with conventional synthetic grafts.
Collapse
|
26
|
Place LW, Kelly SM, Kipper MJ. Synthesis and Characterization of Proteoglycan-Mimetic Graft Copolymers with Tunable Glycosaminoglycan Density. Biomacromolecules 2014; 15:3772-80. [DOI: 10.1021/bm501045k] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Laura W. Place
- School of Biomedical Engineering and ‡Department of
Chemical and Biological
Engineering, Colorado State University, 1370 Campus Delivery, Fort Collins, Colorado 80523, United States
| | - Sean M. Kelly
- School of Biomedical Engineering and ‡Department of
Chemical and Biological
Engineering, Colorado State University, 1370 Campus Delivery, Fort Collins, Colorado 80523, United States
| | - Matt J. Kipper
- School of Biomedical Engineering and ‡Department of
Chemical and Biological
Engineering, Colorado State University, 1370 Campus Delivery, Fort Collins, Colorado 80523, United States
| |
Collapse
|
27
|
Development and evaluation of paclitaxel loaded PLGA:poloxamer blend nanoparticles for cancer chemotherapy. Int J Biol Macromol 2014; 69:393-9. [DOI: 10.1016/j.ijbiomac.2014.05.067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/12/2014] [Accepted: 05/19/2014] [Indexed: 11/18/2022]
|
28
|
Reguera-Nuñez E, Roca C, Hardy E, de la Fuente M, Csaba N, Garcia-Fuentes M. Implantable controlled release devices for BMP-7 delivery and suppression of glioblastoma initiating cells. Biomaterials 2014; 35:2859-67. [DOI: 10.1016/j.biomaterials.2013.12.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/08/2013] [Indexed: 01/04/2023]
|
29
|
Losi P, Briganti E, Errico C, Lisella A, Sanguinetti E, Chiellini F, Soldani G. Fibrin-based scaffold incorporating VEGF- and bFGF-loaded nanoparticles stimulates wound healing in diabetic mice. Acta Biomater 2013; 9:7814-21. [PMID: 23603001 DOI: 10.1016/j.actbio.2013.04.019] [Citation(s) in RCA: 223] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 04/02/2013] [Accepted: 04/09/2013] [Indexed: 01/13/2023]
Abstract
Diabetic skin ulcers are difficult to heal spontaneously due to the reduced levels and activity of endogenous growth factors. Recombinant human vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are known to stimulate cell proliferation and accelerate wound healing. Direct delivery of VEGF and bFGF at the wound site in a sustained and controllable way without loss of bioactivity would enhance their biological effects. The aim of this study was to develop a poly(ether)urethane-polydimethylsiloxane/fibrin-based scaffold containing poly(lactic-co-glycolic acid) (PLGA) nanoparticles loaded with VEGF and bFGF (scaffold/GF-loaded NPs) and to evaluate its wound healing properties in genetically diabetic mice (db/db). The scaffold application on full-thickness dorsal skin wounds significantly accelerated wound closure at day 15 compared to scaffolds without growth factors (control scaffold) or containing unloaded PLGA nanoparticles (scaffold/unloaded NPs). However, the closure rate was similar to that observed in mice treated with scaffolds containing free VEGF and bFGF (scaffold/GFs). Both scaffolds containing growth factors induced complete re-epithelialization, with enhanced granulation tissue formation/maturity and collagen deposition compared to the other groups, as revealed by histological analysis. The ability of the scaffold/GF-loaded NPs to promote wound healing in a diabetic mouse model suggests its potential use as a dressing in patients with diabetic foot ulcers.
Collapse
Affiliation(s)
- Paola Losi
- Laboratory of Biomaterials & Graft Technology, Institute of Clinical Physiology, National Research Council, Via Aurelia Sud, 54100 Massa, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
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: 1.9] [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
|
31
|
Fabiilli ML, Wilson CG, Padilla F, Martín-Saavedra FM, Fowlkes JB, Franceschi RT. Acoustic droplet-hydrogel composites for spatial and temporal control of growth factor delivery and scaffold stiffness. Acta Biomater 2013; 9:7399-409. [PMID: 23535233 DOI: 10.1016/j.actbio.2013.03.027] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 01/29/2013] [Accepted: 03/13/2013] [Indexed: 10/27/2022]
Abstract
Wound healing is regulated by temporally and spatially restricted patterns of growth factor signaling, but there are few delivery vehicles capable of the "on-demand" release necessary for recapitulating these patterns. Recently we described a perfluorocarbon double emulsion that selectively releases a protein payload upon exposure to ultrasound through a process known as acoustic droplet vaporization (ADV). In this study, we describe a delivery system composed of fibrin hydrogels doped with growth factor-loaded double emulsion for applications in tissue regeneration. Release of immunoreactive basic fibroblast growth factor (bFGF) from the composites increased up to 5-fold following ADV and delayed release was achieved by delaying exposure to ultrasound. Releasates of ultrasound-treated materials significantly increased the proliferation of endothelial cells compared to sham controls, indicating that the released bFGF was bioactive. ADV also triggered changes in the ultrastructure and mechanical properties of the fibrin as bubble formation and consolidation of the fibrin in ultrasound-treated composites were accompanied by up to a 22-fold increase in shear stiffness. ADV did not reduce the viability of cells suspended in composite scaffolds. These results demonstrate that an acoustic droplet-hydrogel composite could have broad utility in promoting wound healing through on-demand control of growth factor release and/or scaffold architecture.
Collapse
|
32
|
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
|
33
|
Formiga FR, Tamayo E, Simón-Yarza T, Pelacho B, Prósper F, Blanco-Prieto MJ. Angiogenic therapy for cardiac repair based on protein delivery systems. Heart Fail Rev 2013; 17:449-73. [PMID: 21979836 DOI: 10.1007/s10741-011-9285-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cardiovascular diseases remain the first cause of morbidity and mortality in the developed countries and are a major problem not only in the western nations but also in developing countries. Current standard approaches for treating patients with ischemic heart disease include angioplasty or bypass surgery. However, a large number of patients cannot be treated using these procedures. Novel curative approaches under investigation include gene, cell, and protein therapy. This review focuses on potential growth factors for cardiac repair. The role of these growth factors in the angiogenic process and the therapeutic implications are reviewed. Issues including aspects of growth factor delivery are presented in relation to protein stability, dosage, routes, and safety matters. Finally, different approaches for controlled growth factor delivery are discussed as novel protein delivery platforms for cardiac regeneration.
Collapse
Affiliation(s)
- F R Formiga
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Yalcin M, Lin HY, Sudha T, Bharali DJ, Meng R, Tang HY, Davis FB, Stain SC, Davis PJ, Mousa SA. Response of Human Pancreatic Cancer Cell Xenografts to Tetraiodothyroacetic Acid Nanoparticles. Discov Oncol 2013; 4:176-85. [DOI: 10.1007/s12672-013-0137-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 02/08/2013] [Indexed: 01/01/2023] Open
|
35
|
Chung E, Ricles LM, Stowers RS, Nam SY, Emelianov SY, Suggs LJ. Multifunctional nanoscale strategies for enhancing and monitoring blood vessel regeneration. NANO TODAY 2012; 7:514-531. [PMID: 28989343 PMCID: PMC5630157 DOI: 10.1016/j.nantod.2012.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Nanomedicine has great potential in biomedical applications, and specifically in regenerative medicine and vascular tissue engineering. Designing nanometer-sized therapeutic and diagnostic devices for tissue engineering applications is critical because cells experience and respond to stimuli on this spatial scale. For example, nanoscaffolds, including nanoscalestructured or nanoscale surface-modified vascular scaffolds, can influence cell alignment, adhesion, and differentiation to promote better endothelization. Furthermore, nanoscale contrast agents can be extended to the field of biomedical imaging to monitor and track stem cells to better understand the process of neovascularization. In addition, nanoscale systems capable of delivering biomolecules (e.g. peptides and angiogenic genes/proteins) can influence cell behavior, function, and phenotype to promote blood vessel regeneration. This review will focus on nanomedicine and nanoscale strategies applied to vascular tissue engineering. In particular, some of the latest research and potential applications pertaining to nanoscaffolds, biomedical imaging and cell tracking using nanoscale contrast agents, and nanodelivery systems of bioactive molecules applied to blood vessel regeneration will be discussed. In addition, the overlap between these three areas and their synergistic effects will be examined as related to vascular tissue engineering.
Collapse
Affiliation(s)
- Eunna Chung
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, C0800, Austin, TX 78712-0238, USA
| | - Laura M. Ricles
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, C0800, Austin, TX 78712-0238, USA
| | - Ryan S. Stowers
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, C0800, Austin, TX 78712-0238, USA
| | - Seung Yun Nam
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, C0800, Austin, TX 78712-0238, USA
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX 78712-0238, USA
| | - Stanislav Y. Emelianov
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, C0800, Austin, TX 78712-0238, USA
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX 78712-0238, USA
| | - Laura J. Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, C0800, Austin, TX 78712-0238, USA
| |
Collapse
|
36
|
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: 6.8] [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
|
37
|
Santo VE, Gomes ME, Mano JF, Reis RL. From nano- to macro-scale: nanotechnology approaches for spatially controlled delivery of bioactive factors for bone and cartilage engineering. Nanomedicine (Lond) 2012; 7:1045-66. [DOI: 10.2217/nnm.12.78] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The field of biomaterials has advanced towards the molecular and nanoscale design of bioactive systems for tissue engineering, regenerative medicine and drug delivery. Spatial cues are displayed in the 3D extracellular matrix and can include signaling gradients, such as those observed during chemotaxis. Architectures range from the nanometer to the centimeter length scales as exemplified by extracellular matrix fibers, cells and macroscopic shapes. The main focus of this review is the application of a biomimetic approach by the combination of architectural cues, obtained through the application of micro- and nanofabrication techniques, with the ability to sequester and release growth factors and other bioactive agents in a spatiotemporal controlled manner for bone and cartilage engineering.
Collapse
Affiliation(s)
- Vítor E Santo
- 3B’s Research Group - Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal
| | - Manuela E Gomes
- 3B’s Research Group - Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal
| | - João F Mano
- 3B’s Research Group - Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal
| | - Rui L Reis
- 3B’s Research Group - Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal
| |
Collapse
|
38
|
Dry powders based on PLGA nanoparticles for pulmonary delivery of antibiotics: Modulation of encapsulation efficiency, release rate and lung deposition pattern by hydrophilic polymers. J Control Release 2012; 157:149-59. [DOI: 10.1016/j.jconrel.2011.08.010] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 08/02/2011] [Accepted: 08/05/2011] [Indexed: 11/18/2022]
|
39
|
Abstract
Ischemic disease causes a large number of deaths and significant clinical problems worldwide. Therapeutic angiogenesis, strengthened by advances in growth-factor-based therapies, is a promising solution to ischemic pathologies. Major challenges in therapeutic angiogenesis are the lack of stability of native angiogenic proteins and also providing sustained delivery of biologically active proteins at the ischemic sites. This paper will discuss various protein engineering strategies to develop stabilized proangiogenic proteins and several biomaterial technologies used to amplify the angiogenic outcome by delivering biologically active growth factors in a sustained manner.
Collapse
Affiliation(s)
- Rituparna Sinha Roy
- Department of Biological and Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur Campus, P O BCKV Campus Mail Office, West Bengal 741252, India.
| | | | | |
Collapse
|
40
|
Ranjan AP, Zeglam K, Mukerjee A, Thamake S, Vishwanatha JK. A sustained release formulation of chitosan modified PLCL:poloxamer blend nanoparticles loaded with optical agent for animal imaging. NANOTECHNOLOGY 2011; 22:295104. [PMID: 21693801 DOI: 10.1088/0957-4484/22/29/295104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The objective of this study was to develop optical imaging agent loaded biodegradable nanoparticles with indocynanine green (ICG) using chitosan modified poly(L-lactide-co-epsilon-caprolactone) (PLCL):poloxamer (Pluronic F68) blended polymer. Nanoparticles were formulated with an emulsification solvent diffusion technique using PLCL and poloxamer as blend-polymers. Polyvinyl alcohol (PVA) and chitosan were used as stabilizers. The particle size, shape and zeta potential of the formulated nanoparticles and the release kinetics of ICG from these nanoparticles were determined. Further, biodistribution of these nanoparticles was studied in mice at various time points until 24 h following intravenous administration, using a non-invasive imaging system. The average particle size of the nanoparticles was found to be 146 ± 3.7 to 260 ± 4.5 nm. The zeta potential progressively increased from - 41.6 to + 25.3 mV with increasing amounts of chitosan. Particle size and shape of the nanoparticles were studied using transmission electron microscopy (TEM) which revealed the particles to be smooth and spherical in shape. These nanoparticles were efficiently delivered to the cytoplasm of the cells, as observed in prostate and breast cancer cells using confocal laser scanning microscopy. In vitro release studies indicated sustained release of ICG from the nanoparticles over a period of seven days. Nanoparticle distribution results in mice showing improved uptake and accumulation with chitosan modified nanoparticles in various organs and slower clearance at different time points over a 24 h period as compared to unmodified nanoparticles. The successful formulation of such cationically modified nanoparticles for encapsulating optical agents may lead to a potential deep tissue imaging technique for tumor detection, diagnosis and therapy.
Collapse
Affiliation(s)
- Amalendu P Ranjan
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | | | | | |
Collapse
|
41
|
Amsden BG. Delivery approaches for angiogenic growth factors in the treatment of ischemic conditions. Expert Opin Drug Deliv 2011; 8:873-90. [DOI: 10.1517/17425247.2011.577412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
42
|
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: 683] [Impact Index Per Article: 52.5] [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
|
43
|
McClure M, Wolfe P, Rodriguez I, Bowlin G. Bioengineered vascular grafts: improving vascular tissue engineering through scaffold design. J Drug Deliv Sci Technol 2011. [DOI: 10.1016/s1773-2247(11)50030-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|