1
|
Enayati M, Liu W, Madry H, Neisiany RE, Cucchiarini M. Functionalized hydrogels as smart gene delivery systems to treat musculoskeletal disorders. Adv Colloid Interface Sci 2024; 331:103232. [PMID: 38889626 DOI: 10.1016/j.cis.2024.103232] [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: 01/15/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Despite critical advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy based on the delivery of therapeutic genetic sequences has strong value to offer effective, durable options to decisively manage such disorders. Furthermore, scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy, allowing for the spatiotemporal delivery of candidate genes to sites of injury. Among the many scaffolds for musculoskeletal research, hydrogels raised increasing attention in addition to other potent systems (solid, hybrid scaffolds) due to their versatility and competence as drug and cell carriers in tissue engineering and wound dressing. Attractive functionalities of hydrogels for musculoskeletal therapy include their injectability, stimuli-responsiveness, self-healing, and nanocomposition that may further allow to upgrade of them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. Such functionalized hydrogels may also be tuned to successfully transfer therapeutic genes in a minimally invasive manner in order to protect their cargos and allow for their long-term effects. In light of such features, this review focuses on functionalized hydrogels and demonstrates their competence for the treatment of musculoskeletal disorders using gene therapy procedures, from gene therapy principles to hydrogel functionalization methods and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are being discussed in the perspective of translation in patients. STATEMENT OF SIGNIFICANCE: Despite advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy has strong value in offering effective, durable options to decisively manage such disorders. Scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy. Among many scaffolds for musculoskeletal research, hydrogels raised increasing attention. Functionalities including injectability, stimuli-responsiveness, and self-healing, tune them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. This review introduces functionalized hydrogels for musculoskeletal disorder treatment using gene therapy procedures, from gene therapy principles to functionalized hydrogels and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are discussed from the perspective of translation in patients.
Collapse
Affiliation(s)
- Mohammadsaeid Enayati
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Rasoul Esmaeely Neisiany
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland; Department of Polymer Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany.
| |
Collapse
|
2
|
Krasilnikova O, Yakimova A, Ivanov S, Atiakshin D, Kostin AA, Sosin D, Shegay P, Kaprin AD, Klabukov I. Gene-Activated Materials in Regenerative Dentistry: Narrative Review of Technology and Study Results. Int J Mol Sci 2023; 24:16250. [PMID: 38003439 PMCID: PMC10671237 DOI: 10.3390/ijms242216250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Treatment of a wide variety of defects in the oral and maxillofacial regions requires the use of innovative approaches to achieve best outcomes. One of the promising directions is the use of gene-activated materials (GAMs) that represent a combination of tissue engineering and gene therapy. This approach implies that biocompatible materials will be enriched with gene-carrying vectors and implanted into the defect site resulting in transfection of the recipient's cells and secretion of encoded therapeutic protein in situ. GAMs may be presented in various designs depending on the type of material, encoded protein, vector, and way of connecting the vector and the material. Thus, it is possible to choose the most suitable GAM design for the treatment of a particular pathology. The use of plasmids for delivery of therapeutic genes is of particular interest. In the present review, we aimed to delineate the principle of work and various designs of plasmid-based GAMs and to highlight results of experimental and clinical studies devoted to the treatment of periodontitis, jaw bone defects, teeth avulsion, and other pathologies in the oral and maxillofacial regions.
Collapse
Affiliation(s)
- Olga Krasilnikova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Anna Yakimova
- A. Tsyb Medical Radiological Research Centre—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukov St. 10, 249031 Obninsk, Russia
| | - Sergey Ivanov
- A. Tsyb Medical Radiological Research Centre—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukov St. 10, 249031 Obninsk, Russia
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Dmitri Atiakshin
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Andrey A. Kostin
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Dmitry Sosin
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, 119121 Moscow, Russia
| | - Peter Shegay
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Ilya Klabukov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering, National Research Nuclear University MEPhI, Studgorodok 1, 249039 Obninsk, Russia
| |
Collapse
|
3
|
Han Y, Cao L, Li G, Zhou F, Bai L, Su J. Harnessing Nucleic Acids Nanotechnology for Bone/Cartilage Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301996. [PMID: 37116115 DOI: 10.1002/smll.202301996] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/29/2023] [Indexed: 06/19/2023]
Abstract
The effective regeneration of weight-bearing bone defects and critical-sized cartilage defects remains a significant clinical challenge. Traditional treatments such as autologous and allograft bone grafting have not been successful in achieving the desired outcomes, necessitating the need for innovative therapeutic approaches. Nucleic acids have attracted significant attention due to their ability to be designed to form discrete structures and programmed to perform specific functions at the nanoscale. The advantages of nucleic acid nanotechnology offer numerous opportunities for in-cell and in vivo applications, and hold great promise for advancing the field of biomaterials. In this review, the current abilities of nucleic acid nanotechnology to be applied in bone and cartilage regeneration are summarized and insights into the challenges and future directions for the development of this technology are provided.
Collapse
Affiliation(s)
- Yafei Han
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Luodian Hospital, Shanghai, 201908, China
| | - Guangfeng Li
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
4
|
Khvorostina M, Mironov A, Nedorubova I, Bukharova T, Vasilyev A, Goldshtein D, Komlev V, Popov V. Osteogenesis Enhancement with 3D Printed Gene-Activated Sodium Alginate Scaffolds. Gels 2023; 9:gels9040315. [PMID: 37102926 PMCID: PMC10137500 DOI: 10.3390/gels9040315] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/28/2023] Open
Abstract
Natural and synthetic hydrogel scaffolds containing bioactive components are increasingly used in solving various tissue engineering problems. The encapsulation of DNA-encoding osteogenic growth factors with transfecting agents (e.g., polyplexes) into such scaffold structures is one of the promising approaches to delivering the corresponding genes to the area of the bone defect to be replaced, providing the prolonged expression of the required proteins. Herein, a comparative assessment of both in vitro and in vivo osteogenic properties of 3D printed sodium alginate (SA) hydrogel scaffolds impregnated with model EGFP and therapeutic BMP-2 plasmids was demonstrated for the first time. The expression levels of mesenchymal stem cell (MSC) osteogenic differentiation markers Runx2, Alpl, and Bglap were evaluated by real-time PCR. Osteogenesis in vivo was studied on a model of a critical-sized cranial defect in Wistar rats using micro-CT and histomorphology. The incorporation of polyplexes comprising pEGFP and pBMP-2 plasmids into the SA solution followed by 3D cryoprinting does not affect their transfecting ability compared to the initial compounds. Histomorphometry and micro-CT analysis 8 weeks after scaffold implantation manifested a significant (up to 46%) increase in new bone volume formation for the SA/pBMP-2 scaffolds compared to the SA/pEGFP ones.
Collapse
Affiliation(s)
- Maria Khvorostina
- Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics", Russian Academy of Sciences, Moscow 108840, Russia
- Research Centre for Medical Genetics, Moscow 115478, Russia
| | - Anton Mironov
- Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics", Russian Academy of Sciences, Moscow 108840, Russia
| | | | | | - Andrey Vasilyev
- Research Centre for Medical Genetics, Moscow 115478, Russia
- Central Research Institute of Dental and Maxillofacial Surgery, Moscow 119021, Russia
| | | | - Vladimir Komlev
- A.A. Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Moscow 119334, Russia
| | - Vladimir Popov
- Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics", Russian Academy of Sciences, Moscow 108840, Russia
| |
Collapse
|
5
|
Agas D, Marchegiani A, Laus F, Gabai V, Sufianov AA, Shneider A, Sabbieti MG. p62/SQSTM1 indirectly mediates remote multipotent mesenchymal cells and rescues bone loss and bone marrow integrity in ovariectomized rats. J Cell Physiol 2023; 238:407-419. [PMID: 36565474 DOI: 10.1002/jcp.30937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Intramuscular administration of p62/SQSTM1 (sequestosome1)-encoding plasmid demonstrated an anticancer effect in rodent models and dogs as well as a high safety profile and the first evidence of clinical benefits in humans. Also, an anti-inflammatory effect of the plasmid was reported in several rodent disease models. Yet, the mechanisms of action for the p62 plasmid remain unknown. Here, we tested a hypothesis that the p62-plasmid can act through the modulation of bone marrow multipotent mesenchymal cells (MSCs). We demonstrated that a p62 plasmid can affect MSCs indirectly by stimulating p62-transfected cells to secrete an active ingredient(s) sensed by untransfected MSCs. When we transfected MSCs with the p62-plasmid, collected their supernatant, and added it to an untransfected MSCs culture, it switched the differentiation state and prompt osteogenic responses of the untransfected MSCs. According to an accepted viewpoint, ovariectomy leads to bone pathology via dysregulation of MSCs, and restoring the MSC homeostasis would restore ovariectomy-induced bone damage. To validate our in vitro observations in a clinically relevant in vivo model, we administered the p62 plasmid to ovariectomized rats. It partially reversed bone loss and notably reduced adipogenesis with concurrent reestablishing of the MSC subpopulation pool within the bone marrow. Overall, our study suggests that remote modulation of progenitor MSCs via administering a p62-encoding plasmid may constitute a mechanism for its previously reported effects and presents a feasible disease-preventing and/or therapeutic strategy.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | - Fulvio Laus
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | | | - Albert A Sufianov
- Federal Center of Neurosurgery, Tyumen, Russian Federation.,Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Alexander Shneider
- CureLab Oncology Inc., Dedham, Massachusetts, USA.,Department of Molecular Biology, Ariel University, Ariel, Israel
| | | |
Collapse
|
6
|
Gene-Activated Matrix with Self-Assembly Anionic Nano-Device Containing Plasmid DNAs for Rat Cranial Bone Augmentation. MATERIALS 2021; 14:ma14227097. [PMID: 34832496 PMCID: PMC8621468 DOI: 10.3390/ma14227097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/07/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022]
Abstract
We have developed nanoballs, a biocompatible self-assembly nano-vector based on electrostatic interactions that arrange anionic macromolecules to polymeric nanomaterials to create nucleic acid carriers. Nanoballs exhibit low cytotoxicity and high transfection efficiently in vivo. This study investigated whether a gene-activated matrix (GAM) composed of nanoballs containing plasmid (p) DNAs encoding bone morphogenetic protein 4 (pBMP4) could promote bone augmentation with a small amount of DNA compared to that composed of naked pDNAs. We prepared nanoballs (BMP4-nanoballs) constructed with pBMP4 and dendrigraft poly-L-lysine (DGL, a cationic polymer) coated by γ-polyglutamic acid (γ-PGA; an anionic polymer), and determined their biological functions in vitro and in vivo. Next, GAMs were manufactured by mixing nanoballs with 2% atelocollagen and β-tricalcium phosphate (β-TCP) granules and lyophilizing them for bone augmentation. The GAMs were then transplanted to rat cranial bone surfaces under the periosteum. From the initial stage, infiltrated macrophages and mesenchymal progenitor cells took up the nanoballs, and their anti-inflammatory and osteoblastic differentiations were promoted over time. Subsequently, bone augmentation was clearly recognized for up to 8 weeks in transplanted GAMs containing BMP4-nanoballs. Notably, only 1 μg of BMP4-nanoballs induced a sufficient volume of new bone, while 1000 μg of naked pDNAs were required to induce the same level of bone augmentation. These data suggest that applying this anionic vector to the appropriate matrices can facilitate GAM-based bone engineering.
Collapse
|
7
|
Gajiwala M, Paliwal J, Husain SY, Dadarwal A, Kalla R, Sharma V, Sharma M. Influence of surface modification of titanium implants on improving osseointegration: An in vitro study. J Prosthet Dent 2021; 126:405.e1-405.e7. [PMID: 34376285 DOI: 10.1016/j.prosdent.2021.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 11/30/2022]
Abstract
STATEMENT OF PROBLEM The effect of aging and the surface treatment of implants on osseointegration needs to be evaluated. PURPOSE The purpose of this in vitro study was to evaluate the effects of aging and the surface treatment of titanium with ultraviolet (UV) radiation and fibroblast growth factor (FGF) on hydrophilicity and cell growth and thus on osseointegration. MATERIAL AND METHODS A total of 28 specimens were divided into 2 groups to measure hydrophilicity (n=14) and cell growth (n=14). Each group was further divided into 4 groups according to surface modification. These include the control group (CG) (nascent specimens), aged group (AG) (nascent specimens aged for 4 weeks), photofunctionalized group (PG) (aged specimens UV-A treated), and mimed group (MG) (aged specimens UV-A and FGF2 treated). The PG and MG specimens were treated with UV-A light for 40 minutes. The biomimetic surface modification was performed for MG. Hydrophilicity was measured by using the contact angle in relation to the surface of titanium disks with the help of a drop shape analyzing device (KRUSS), and cell growth was measured by calculating the number of stem cells per cm2 with the help of a scanning electron microscope (SEM). The data obtained were subjected to statistical analysis with a statistical software program (α=.05). RESULTS The lowest contact angle values were found in PG (13.52 ±0.90 degrees) and the highest in AG (70.54 ±1.72 degrees). The highest number of cells per cm2 (2880 ±99.33) were found for MG, and the lowest number of cells per cm2 (760 ±9.17) for AG. CONCLUSIONS Aging decreased the hydrophilicity and cell adhesion, migration, and growth on the titanium surface. UV treatment improved the hydrophilicity, cell adhesion, migration, and growth for both CG and AG. FGF2 treatment increased the cell adhesion, migration, and growth for CG, AG, and PG.
Collapse
Affiliation(s)
- Mohini Gajiwala
- Post Graduate student, Department of Prosthodontics, Crown & bridge, R.U.H.S. College of Dental Sciences, Jaipur, India
| | - Jyoti Paliwal
- Professor and Head, Department of Prosthodontics, Crown & bridge, R.U.H.S. College of Dental Sciences, Jaipur, India.
| | - Syed Yawer Husain
- Assistant Professor, Department of Dental Materials, R.U.H.S. College of Dental Sciences, Jaipur, India
| | - Ashish Dadarwal
- Senior Demonstrator, Department of Prosthodontics, Crown & bridge, R.U.H.S. College of Dental Sciences, Jaipur, India
| | - Rajni Kalla
- Assistant Professor, Department of Prosthodontics, Crown & bridge, R.U.H.S. College of Dental Sciences, Jaipur, India
| | - Vineet Sharma
- Medical Officer (Dental), Department of Prosthodontics, Crown & bridge, R.U.H.S. College of Dental Sciences, Jaipur, India
| | - Meenakshi Sharma
- Assistant Professor, Department of Pedodontics, R.U.H.S. College of Dental Sciences, Jaipur, India
| |
Collapse
|
8
|
Osteogenic effects of microRNA-335-5p/lipidoid nanoparticles coated on titanium surface. Arch Oral Biol 2021; 129:105207. [PMID: 34273868 DOI: 10.1016/j.archoralbio.2021.105207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/26/2021] [Accepted: 07/03/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE In this study, we aimed to investigate the therapeutic potential of miR-335-5p lipidoid nanocomplexes coated on Titanium (Ti) SLActive surface by lyophilization. DESIGN In our model, we coated miR-335-5p/Lipidoid nanoparticles on titanium implant, seeded GFP-labelled mouse bone marrow stromal cells (BMSCs) onto the functionalized Ti implant surface, and analyzed the transfection efficiency, cell adhesion, proliferation, and osteogenic activity of the bone-implant interface. RESULTS The Ti SLActive surface displayed a suitable hydrophilicity ability and provided a large surface area for miRNA loading, enabling spatial retention of the miRNAs within the nanopores until cellular delivery. We demonstrated a high transfection efficiency of miR-335-5p lipidoid nanoparticles in BMSCs seeded onto the Ti SLActive surface, even after 14 days. Alkaline phosphatase (ALP) activity and cell vitality were significantly increased in BMSCs transfected with miR-335-5p at 7 and 14 days as opposed to cells transfected with negative controls. When miR-335-5p transfected BMSCs were induced to undergo osteogenic differentiation, we detected increased mRNA expression of osteogenic markers including Alkaline phosphatase (ALP), collagen I (COL1), osteocalcin (OCN) and bone sialoprotein (BSP) at 7 and 14 days as compared with negative controls. CONCLUSION MiR-335-5p lipidoid nanoparticles could be used as a new cost-effective methodology to increase the osteogenic capacity of biomedical Ti implants.
Collapse
|
9
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
10
|
Gantenbein B, Tang S, Guerrero J, Higuita-Castro N, Salazar-Puerta AI, Croft AS, Gazdhar A, Purmessur D. Non-viral Gene Delivery Methods for Bone and Joints. Front Bioeng Biotechnol 2020; 8:598466. [PMID: 33330428 PMCID: PMC7711090 DOI: 10.3389/fbioe.2020.598466] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Viral carrier transport efficiency of gene delivery is high, depending on the type of vector. However, viral delivery poses significant safety concerns such as inefficient/unpredictable reprogramming outcomes, genomic integration, as well as unwarranted immune responses and toxicity. Thus, non-viral gene delivery methods are more feasible for translation as these allow safer delivery of genes and can modulate gene expression transiently both in vivo, ex vivo, and in vitro. Based on current studies, the efficiency of these technologies appears to be more limited, but they are appealing for clinical translation. This review presents a summary of recent advancements in orthopedics, where primarily bone and joints from the musculoskeletal apparatus were targeted. In connective tissues, which are known to have a poor healing capacity, and have a relatively low cell-density, i.e., articular cartilage, bone, and the intervertebral disk (IVD) several approaches have recently been undertaken. We provide a brief overview of the existing technologies, using nano-spheres/engineered vesicles, lipofection, and in vivo electroporation. Here, delivery for microRNA (miRNA), and silencing RNA (siRNA) and DNA plasmids will be discussed. Recent studies will be summarized that aimed to improve regeneration of these tissues, involving the delivery of bone morphogenic proteins (BMPs), such as BMP2 for improvement of bone healing. For articular cartilage/osteochondral junction, non-viral methods concentrate on targeted delivery to chondrocytes or MSCs for tissue engineering-based approaches. For the IVD, growth factors such as GDF5 or GDF6 or developmental transcription factors such as Brachyury or FOXF1 seem to be of high clinical interest. However, the most efficient method of gene transfer is still elusive, as several preclinical studies have reported many different non-viral methods and clinical translation of these techniques still needs to be validated. Here we discuss the non-viral methods applied for bone and joint and propose methods that can be promising in clinical use.
Collapse
Affiliation(s)
- Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Shirley Tang
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Julien Guerrero
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ana I Salazar-Puerta
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Andreas S Croft
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Devina Purmessur
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
11
|
Mendrek B, Fus-Kujawa A, Teper P, Botor M, Kubacki J, Sieroń AL, Kowalczuk A. Star polymer-based nanolayers with immobilized complexes of polycationic stars and DNA for deposition gene delivery and recovery of intact transfected cells. Int J Pharm 2020; 589:119823. [PMID: 32861771 DOI: 10.1016/j.ijpharm.2020.119823] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/31/2022]
Abstract
We designed a novel thermoresponsive system of nanolayers composed of star poly[oligo(ethylene glycol) methacrylate]s (S-POEGMA) covalently bonded to a solid support and covered with polyplexes of cationic star polymers and plasmid DNA (pDNA). S-POEGMA stars were attached to the solid support via a UV-mediated "grafting to" method. To the best of our knowledge, for the first time, the conformational changes of obtained star nanolayers, occurring with changes in temperature, were studied using a quartz crystal microbalance technique. Next, the polyplexes of star poly[N,N'-dimethylaminoethyl methacrylate-ran-di(ethylene glycol) methacrylate] (S-P(DMAEMA-DEGMA)) with pDNA, exhibiting a phase transition temperature (TCP) in culture medium DMEM, were deposited on S-POEGMA layers when the temperature increased above the TCP of polyplex. The thermoresponsivity of the system was then the main mechanism for controlling the adhesion, proliferation, transfection and detachment of HT-1080 cells. The nanolayers promoted the effective cell culture and delivered nucleic acids into cells, with a transfection efficiency several times higher than that of the control. The detachment of the transfected cells was regulated only by the change of temperature. The studies demonstrated that we obtained a novel and effective system, based on a star polymer architecture, useful for gene delivery and tissue engineering applications.
Collapse
Affiliation(s)
- Barbara Mendrek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland
| | - Agnieszka Fus-Kujawa
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland
| | - Paulina Teper
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland
| | - Malwina Botor
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland
| | - Jerzy Kubacki
- A. Chelkowski Institute of Physics, University of Silesia, Uniwersytecka 4, 40-007 Katowice, Poland; Silesian Center for Education and Interdisciplinary Research, 75 Pulku Piechoty 1A, 41-500 Chorzów, Poland
| | - Aleksander L Sieroń
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland
| | - Agnieszka Kowalczuk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland.
| |
Collapse
|
12
|
Husteden C, Doberenz F, Goergen N, Pinnapireddy SR, Janich C, Langner A, Syrowatka F, Repanas A, Erdmann F, Jedelská J, Bakowsky U, Groth T, Wölk C. Contact-Triggered Lipofection from Multilayer Films Designed as Surfaces for in Situ Transfection Strategies in Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2020; 12:8963-8977. [PMID: 32003972 DOI: 10.1021/acsami.9b18968] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biomaterials, which release active compounds after implantation, are an essential tool for targeted regenerative medicine. In this study, thin multilayer films loaded with lipid/DNA complexes (lipoplexes) were designed as surface coatings for in situ transfection applicable in tissue engineering and regenerative medicine. The film production and embedding of lipoplexes were based on the layer-by-layer (LbL) deposition technique. Hyaluronic acid (HA) and chitosan (CHI) were used as the polyelectrolyte components. The embedded plasmid DNA was complexed using a new designed cationic lipid formulation, namely, OH4/DOPE 1/1, the advantageous characteristics of which have been proven already. Three different methods were tested regarding its efficiency of lipid and DNA deposition. Therefore, several surface specific analytics were used to characterize the LbL formation, the lipid DNA embedding, and the surface characteristics of the multilayer films, such as fluorescence microscopy, surface plasmon resonance spectroscopy, ellipsometry, zeta potential measurements, atomic force microscopy, and scanning electron microscopy. Interaction studies were conducted for optimized lipoplex-loaded polyelectrolyte multilayers (PEMs) that showed an efficient attachment of C2C12 cells on the surface. Furthermore, no acute toxic effects were found in cell culture studies, demonstrating biocompatibility. Cell culture experiments with C2C12 cells, a cell line which is hard to transfect, demonstrated efficient transfection of the reporter gene encoding for green fluorescent protein. In vivo experiments using the chicken embryo chorion allantois membrane animal replacement model showed efficient gene-transferring rates in living complex tissues, although the DNA-loaded films were stored over 6 days under wet and dried conditions. Based on these findings, it can be concluded that OH4/DOPE 1/1 lipoplex-loaded PEMs composed of HA and CHI can be an efficient tool for in situ transfection in regenerative medicine.
Collapse
Affiliation(s)
- Catharina Husteden
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Falko Doberenz
- Institute of Pharmacy, Department Biomedical Materials , Martin Luther University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
| | - Nathalie Goergen
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Shashank Reddy Pinnapireddy
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Christopher Janich
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Andreas Langner
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Frank Syrowatka
- Interdisciplinary Center of Materials Science , Martin-Luther-University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
| | - Alexandros Repanas
- Institute of Pharmacy, Department Biomedical Materials , Martin Luther University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
| | - Frank Erdmann
- Institute of Pharmacy, Department of Pharmacology , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
| | - Jarmila Jedelská
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics , University of Marburg , Robert-Koch-Str. 4 , 35037 Marburg , Germany
| | - Thomas Groth
- Institute of Pharmacy, Department Biomedical Materials , Martin Luther University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
- Interdisciplinary Center of Materials Science , Martin-Luther-University Halle-Wittenberg , Heinrich-Damerow-Str. 4 , 06120 Halle (Saale) , Germany
- Laboratory of Biomedical Nanotechnologies, Institute of Bionic Technologies and Engineering , I.M. Sechenov First Moscow State University , Trubetskaya Street 8 , 119991 Moscow , Russian Federation
| | - Christian Wölk
- Institute of Pharmacy, Department of Medicinal Chemistry , Martin Luther University Halle-Wittenberg , Wolfgang-Langenbeck-Str. 4 , 06120 Halle (Saale) , Germany
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine , Leipzig University , 04317 Leipzig , Germany
| |
Collapse
|
13
|
Kelly DC, Raftery RM, Curtin CM, O'Driscoll CM, O'Brien FJ. Scaffold-Based Delivery of Nucleic Acid Therapeutics for Enhanced Bone and Cartilage Repair. J Orthop Res 2019; 37:1671-1680. [PMID: 31042304 DOI: 10.1002/jor.24321] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/09/2019] [Indexed: 02/04/2023]
Abstract
Recent advances in tissue engineering have made progress toward the development of biomaterials capable of the delivery of growth factors, such as bone morphogenetic proteins, in order to promote enhanced tissue repair. However, controlling the release of these growth factors on demand and within the desired localized area is a significant challenge and the associated high costs and side effects of uncontrolled delivery have proven increasingly problematic in clinical orthopedics. Gene therapy may be a valuable tool to avoid the limitations of local delivery of growth factors. Following a series of setbacks in the 1990s, the field of gene therapy is now seeing improvements in safety and efficacy resulting in substantial clinical progress and a resurgence in confidence. Biomaterial scaffold-mediated gene therapy provides a template for cell infiltration and tissue formation while promoting transfection of cells to engineer therapeutic proteins in a sustained but ultimately transient fashion. Additionally, scaffold-mediated delivery of RNA-based therapeutics can silence specific genes associated with orthopedic pathological states. This review will provide an overview of the current state-of-the-art in the field of gene-activated scaffolds and their use within orthopedic tissue engineering applications. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1671-1680, 2019.
Collapse
Affiliation(s)
- Domhnall C Kelly
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caitriona M O'Driscoll
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland.,Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| |
Collapse
|
14
|
Biomaterial-based delivery systems of nucleic acid for regenerative research and regenerative therapy. Regen Ther 2019; 11:123-130. [PMID: 31338391 PMCID: PMC6626072 DOI: 10.1016/j.reth.2019.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/10/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Regenerative medicine is a new and promising medical method aiming at treating patients with defective or dysfunctional tissues by maintaining or enhancing the biological activity of cells. The development of biomaterial-based technologies, such as cell scaffolds and carriers for drug delivery system, are highly required to promote the regenerative research and regenerative therapy. Nucleic acids are one of the most feasible factors to efficiently modify the biological activity of cells. The effective and stable delivery of nucleic acids into cells is highly required to succeed in the modification. Biomaterials-based non-viral carriers or biological carriers, like exosomes, play an important role in the efficient delivery of nucleic acids. This review introduces the examples of regenerative research and regenerative therapy based on the delivery of nucleic acids with biomaterials technologies and emphasizes their importance to accomplish regenerative medicine. Modifying the activity of cells is important for regenerative medicine. Various nucleic acids regulate gene expression to modify the activity of cells. Intracellular delivery system is vital to the nucleic acids-based modification. Biomaterials are useful for the intracellular delivery of nucleic acids.
Collapse
Key Words
- Biomaterials
- CRISPR, clustered regularly interspaced short palindromic repeats
- Cas, CRISPR-associated systems
- Cell scaffold
- DDS, drug delivery system
- Drug delivery system
- ECM, extracellular matrix
- MSC, mesenchymal stem cells
- Nucleic acids
- PEG, polyethylene glycol
- PLGA, poly(d,l-lactic acid-co-glycolic acid)
- RISC, RNA-induced silencing complex
- RNAi, RNA interferince
- Regenerative research
- Regenerative therapy
- TALEN, transcription activator-like effector nuclease
- ZFN, zinc finger nucleases
- lncRNA, long non-coding RNA
- mRNA, messenger RNA
- miRNA, microRNA
- siRNA, small interfering RNA
Collapse
|
15
|
Tamay DG, Dursun Usal T, Alagoz AS, Yucel D, Hasirci N, Hasirci V. 3D and 4D Printing of Polymers for Tissue Engineering Applications. Front Bioeng Biotechnol 2019; 7:164. [PMID: 31338366 PMCID: PMC6629835 DOI: 10.3389/fbioe.2019.00164] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022] Open
Abstract
Three-dimensional (3D) and Four-dimensional (4D) printing emerged as the next generation of fabrication techniques, spanning across various research areas, such as engineering, chemistry, biology, computer science, and materials science. Three-dimensional printing enables the fabrication of complex forms with high precision, through a layer-by-layer addition of different materials. Use of intelligent materials which change shape or color, produce an electrical current, become bioactive, or perform an intended function in response to an external stimulus, paves the way for the production of dynamic 3D structures, which is now called 4D printing. 3D and 4D printing techniques have great potential in the production of scaffolds to be applied in tissue engineering, especially in constructing patient specific scaffolds. Furthermore, physical and chemical guidance cues can be printed with these methods to improve the extent and rate of targeted tissue regeneration. This review presents a comprehensive survey of 3D and 4D printing methods, and the advantage of their use in tissue regeneration over other scaffold production approaches.
Collapse
Affiliation(s)
- Dilara Goksu Tamay
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | - Tugba Dursun Usal
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Ayse Selcen Alagoz
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey
| | - Deniz Yucel
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey
- Department of Histology and Embryology, School of Medicine, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Nesrin Hasirci
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
- Department of Biomedical Engineering, Middle East Technical University, Ankara, Turkey
- Department of Chemistry, Middle East Technical University, Ankara, Turkey
| | - Vasif Hasirci
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
- Department of Medical Engineering, School of Engineering, Acıbadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
16
|
Uludag H, Ubeda A, Ansari A. At the Intersection of Biomaterials and Gene Therapy: Progress in Non-viral Delivery of Nucleic Acids. Front Bioeng Biotechnol 2019; 7:131. [PMID: 31214586 PMCID: PMC6558074 DOI: 10.3389/fbioe.2019.00131] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022] Open
Abstract
Biomaterials play a critical role in technologies intended to deliver therapeutic agents in clinical settings. Recent explosion of our understanding of how cells utilize nucleic acids has garnered excitement to develop a range of older (e.g., antisense oligonucleotides, plasmid DNA and transposons) and emerging (e.g., short interfering RNA, messenger RNA and non-coding RNAs) nucleic acid agents for therapy of a wide range of diseases. This review will summarize biomaterials-centered advances to undertake effective utilization of nucleic acids for therapeutic purposes. We first review various types of nucleic acids and their unique abilities to deliver a range of clinical outcomes. Using recent advances in T-cell based therapy as a case in point, we summarize various possibilities for utilizing biomaterials to make an impact in this exciting therapeutic intervention technology, with the belief that this modality will serve as a therapeutic paradigm for other types of cellular therapies in the near future. We subsequently focus on contributions of biomaterials in emerging nucleic acid technologies, specifically focusing on the design of intelligent nanoparticles, deployment of mRNA as an alternative to plasmid DNA, long-acting (integrating) expression systems, and in vitro/in vivo expansion of engineered T-cells. We articulate the role of biomaterials in these emerging nucleic acid technologies in order to enhance the clinical impact of nucleic acids in the near future.
Collapse
Affiliation(s)
- Hasan Uludag
- Department of Chemical and Materinals Engineering, University of Alberta, Edmonton, AB, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Anyeld Ubeda
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Aysha Ansari
- Department of Chemical and Materinals Engineering, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
17
|
Patel S, Athirasala A, Menezes PP, Ashwanikumar N, Zou T, Sahay G, Bertassoni LE. Messenger RNA Delivery for Tissue Engineering and Regenerative Medicine Applications. Tissue Eng Part A 2019; 25:91-112. [PMID: 29661055 PMCID: PMC6352544 DOI: 10.1089/ten.tea.2017.0444] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/09/2018] [Indexed: 12/25/2022] Open
Abstract
The ability to control cellular processes and precisely direct cellular reprogramming has revolutionized regenerative medicine. Recent advances in in vitro transcribed (IVT) mRNA technology with chemical modifications have led to development of methods that control spatiotemporal gene expression. Additionally, there is a current thrust toward the development of safe, integration-free approaches to gene therapy for translational purposes. In this review, we describe strategies of synthetic IVT mRNA modifications and nonviral technologies for intracellular delivery. We provide insights into the current tissue engineering approaches that use a hydrogel scaffold with genetic material. Furthermore, we discuss the transformative potential of novel mRNA formulations that when embedded in hydrogels can trigger controlled genetic manipulation to regenerate tissues and organs in vitro and in vivo. The role of mRNA delivery in vascularization, cytoprotection, and Cas9-mediated xenotransplantation is additionally highlighted. Harmonizing mRNA delivery vehicle interactions with polymeric scaffolds can be used to present genetic cues that lead to precise command over cellular reprogramming, differentiation, and secretome activity of stem cells-an ultimate goal for tissue engineering.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Avathamsa Athirasala
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
| | - Paula P. Menezes
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Postgraduate Program in Health Sciences, Department of Pharmacy, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - N. Ashwanikumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Ting Zou
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
| | - Luiz E. Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
- Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
18
|
Biomaterial-guided delivery of gene vectors for targeted articular cartilage repair. Nat Rev Rheumatol 2018; 15:18-29. [DOI: 10.1038/s41584-018-0125-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
19
|
Controlled Non-Viral Gene Delivery in Cartilage and Bone Repair: Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Nguyen MK, Jeon O, Dang PN, Huynh CT, Varghai D, Riazi H, McMillan A, Herberg S, Alsberg E. RNA interfering molecule delivery from in situ forming biodegradable hydrogels for enhancement of bone formation in rat calvarial bone defects. Acta Biomater 2018; 75:105-114. [PMID: 29885529 PMCID: PMC6119505 DOI: 10.1016/j.actbio.2018.06.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 11/22/2022]
Abstract
RNA interference (RNAi) may be an effective and valuable tool for promoting the growth of functional tissue, as short interfering RNA (siRNA) and microRNA (miRNA) can block the expression of genes that have negative effects on tissue regeneration. Our group has recently reported that the localized and sustained presentation of siRNA against noggin (siNoggin) and miRNA-20a from in situ forming poly(ethylene glycol) (PEG) hydrogels enhanced osteogenic differentiation of encapsulated human bone marrow-derived mesenchymal stem cells (hMSCs). Here, the capacity of the hydrogel system to accelerate bone formation in a rat calvarial bone defect model is presented. After 12 weeks post-implantation, the hydrogels containing encapsulated hMSCs and miRNA-20a resulted in more bone formation in the defects than the hydrogels containing hMSCs without siRNA or with negative control siRNA. This localized and sustained RNA interfering molecule delivery system may provide an excellent platform for healing bony defects and other tissues. STATEMENT OF SIGNIFICANCE Delivery of RNAi molecules may be a valuable strategy to guide cell behavior for tissue engineering applications, but to date there have been no reports of a biomaterial system capable of both encapsulation of cells and controlled delivery of incorporated RNA. Here, we present PEG hydrogels that form in situ via Michael type reaction, and that permit encapsulation of hMSCs and the concomitant controlled delivery of siNoggin and/or miRNA-20a. These RNAs were chosen to suppress noggin, a BMP-2 antagonist, and/or PPAR-γ, a negative regulator of BMP-2-mediated osteogenesis, and therefore promote osteogenic differentiation of hMSCs and subsequent bone repair in critical-sized rat calvarial defects. Simultaneous delivery of hMSCs and miRNA-20a enhanced repair of these defects compared to hydrogels containing hMSCs without siRNA or with negative control siRNA. This in situ forming PEG hydrogel system offers an exciting platform for healing critical-sized bone defects by localized, controlled delivery of RNAi molecules to encapsulated hMSCs and surrounding cells.
Collapse
Affiliation(s)
- Minh K Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Oju Jeon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Phuong N Dang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Cong T Huynh
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Davood Varghai
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Hooman Riazi
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Alexandra McMillan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Samuel Herberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH 44106, United States.
| |
Collapse
|
21
|
Betz VM, Kochanek S, Rammelt S, Müller PE, Betz OB, Messmer C. Recent advances in gene-enhanced bone tissue engineering. J Gene Med 2018; 20:e3018. [PMID: 29601661 DOI: 10.1002/jgm.3018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/18/2018] [Accepted: 03/18/2018] [Indexed: 12/13/2022] Open
Abstract
The loss of bone tissue represents a critical clinical condition that is frequently faced by surgeons. Substantial progress has been made in the area of bone research, providing insight into the biology of bone under physiological and pathological conditions, as well as tools for the stimulation of bone regeneration. The present review discusses recent advances in the field of gene-enhanced bone tissue engineering. Gene transfer strategies have emerged as highly effective tissue engineering approaches for supporting the repair of the musculoskeletal system. By contrast to treatment with recombinant proteins, genetically engineered cells can release growth factors at the site of injury over extended periods of time. Of particular interest are the expedited technologies that can be applied during a single surgical procedure in a cost-effective manner, allowing translation from bench to bedside. Several promising methods based on the intra-operative genetic manipulation of autologous cells or tissue fragments have been developed in preclinical studies. Moreover, gene therapy for bone regeneration has entered the clinical stage with clinical trials for the repair of alveolar bone. Current trends in gene-enhanced bone engineering are also discussed with respect to the movement of the field towards expedited, translational approaches. It is possible that gene-enhanced bone tissue engineering will become a clinical reality within the next few years.
Collapse
Affiliation(s)
- Volker M Betz
- Department of Gene Therapy, University of Ulm, Ulm, Germany.,Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| | | | - Stefan Rammelt
- University Center of Orthopedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany
| | - Peter E Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver B Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carolin Messmer
- Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| |
Collapse
|
22
|
Munsell EV, Kurpad DS, Freeman TA, Sullivan MO. Histone-targeted gene transfer of bone morphogenetic protein-2 enhances mesenchymal stem cell chondrogenic differentiation. Acta Biomater 2018; 71:156-167. [PMID: 29481871 PMCID: PMC5899933 DOI: 10.1016/j.actbio.2018.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/13/2018] [Accepted: 02/20/2018] [Indexed: 01/27/2023]
Abstract
Skeletal tissue regeneration following traumatic injury involves a complex cascade of growth factor signals that direct the differentiation of mesenchymal stem cells (MSCs) within the fracture. The necessity for controlled and localized expression of these factors has highlighted the role gene therapy may play as a promising treatment option for bone repair. However, the design of nanocarrier systems that negotiate efficient intracellular trafficking and nuclear delivery represents a significant challenge. Recent investigations have highlighted the roles histone tail sequences play in directing nuclear delivery and activating DNA transcription. We previously established the ability to recapitulate these natural histone tail activities within non-viral nanocarriers, improving gene transfer and expression by enabling effective navigation to the nucleus via retrograde vesicular trafficking. Herein, we demonstrate that histone-targeting leads to ∼4-fold enhancements in osteogenic bone morphogenetic protein-2 (BMP-2) expression by MSCs over 6 days, as compared with standard polymeric transfection reagents. This improved expression augmented chondrogenesis, an essential first step in fracture healing. Importantly, significant enhancements of cartilage-specific protein expression were triggered by histone-targeted gene transfer, as compared with the response to treatment with equivalent amounts of recombinant BMP-2 protein. In fact, an ∼100-fold increase in recombinant BMP-2 was required to achieve similar levels of chondrogenic gene and protein expression. The enhancements in differentiation achieved using histone-targeting were in part enabled by an increase in transcription factor expression, which functioned to drive MSC chondrogenesis. These novel findings demonstrate the utility of histone-targeted gene transfer strategies to enable substantial reductions in BMP-2 dosing for bone regenerative applications. STATEMENT OF SIGNIFICANCE This contribution addresses significant limitations in non-viral gene transfer for bone regenerative applications by exploiting a novel histone-targeting approach for cell-triggered delivery that induces osteogenic BMP-2 expression coincident with the initiation of bone repair. During repair, proliferating MSCs respond to a complex series of growth factor signals that direct their differentiation along cellular lineages essential to mature bone formation. Although these MSCs are ideal targets for enhanced transfection during cellular mitosis, few non-viral delivery approaches exist to enable maximization of this effect. Accordingly, this contribution seeks to utilize our histone-targeted nanocarrier design strategy to stimulate BMP-2 gene transfer in dividing MSCs. This gene-based approach leads to significantly augmented MSC chondrogenesis, an essential first step in bone tissue repair.
Collapse
Affiliation(s)
- Erik V Munsell
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States.
| | - Deepa S Kurpad
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States.
| | - Theresa A Freeman
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States.
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States.
| |
Collapse
|
23
|
Dobos A, Grandhi TSP, Godeshala S, Meldrum DR, Rege K. Parallel fabrication of macroporous scaffolds. Biotechnol Bioeng 2018; 115:1729-1742. [DOI: 10.1002/bit.26593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Andrew Dobos
- Biomedical Engineering; Arizona State University; Tempe Arizona
| | | | | | - Deirdre R. Meldrum
- Center for Biosignatures Discovery Automation, Biodesign Institute; Arizona State University; Tempe Arizona
| | - Kaushal Rege
- Chemical Engineering; Arizona State University; Tempe Arizona
| |
Collapse
|
24
|
Namini MS, Bayat N, Tajerian R, Ebrahimi-Barough S, Azami M, Irani S, Jangjoo S, Shirian S, Ai J. A comparison study on the behavior of human endometrial stem cell-derived osteoblast cells on PLGA/HA nanocomposite scaffolds fabricated by electrospinning and freeze-drying methods. J Orthop Surg Res 2018; 13:63. [PMID: 29587806 PMCID: PMC5870175 DOI: 10.1186/s13018-018-0754-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/28/2018] [Indexed: 01/08/2023] Open
Abstract
Background An engineered tissue structure is an artificial scaffold combined with cells and signaling factors. Among various polymers, the polylactide-co-glycolide/hydroxyapatite (PLGA/HA) has attracted much attention due to their optimal properties. The aim of this study was to study the behavior of human endometrial stem cell (hEnSC)-derived osteoblast cells cultured on PLGA/HA nanocomposite scaffolds. Methods hEnSCs were isolated and exposed to osteogenic media for 21 days. Differentiated cells were cultured on PLGA/HA synthetic scaffolds. The PLGA/HA-based nanocomposite scaffolds were fabricated using either electrospinning or freeze-drying methods. Behavior of the cells was evaluated a week after seeding hEnSC-derived osteoblast-like cells on these scaffolds. Osteogenesis was investigated in terms of alkaline phosphatase activity, gene expression, immunocytochemistry (ICC), proliferation, and scanning electron microscopy (SEM). Moreover, scaffold properties, such as pore size and morphology of the cells, onto the scaffolds were evaluated using SEM. Furthermore, biocompatibility of these scaffolds was confirmed by 3-(4,5-dimethylthiazoyl-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Results The matrix mineralization was proved by alizarin red staining, and the osteogenic media-treated cultures positively expressed osteocalcin and osteopontin markers. Moreover, qRT-PCR results confirmed the positive gene expression of osteopontin and osteonectin in the differentiated osteoblast-like cells. The results of behavior assessment of the cultured cells on electrospinning and freeze-dried scaffolds showed that the behavior of the cultured cells on the freeze-dried PLGA/HA scaffolds was significantly better than the electrospinning PLGA/HA scaffolds. Conclusion It has been shown that the freeze-dried PLGA/HA nanocomposite scaffolds can appropriately support the attachment and proliferation of the differentiated osteoblast cells and are a suitable candidate for bone tissue engineering.
Collapse
Affiliation(s)
- Mojdeh Salehi Namini
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Neda Bayat
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Roxana Tajerian
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Azami
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Saranaz Jangjoo
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahr-e Kord, Iran.,Shiraz Molecular Pathology Research Center, Dr Daneshbod Lab Pathology, Shiraz, Iran
| | - Jafar Ai
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
The development of an alginate/polycaprolactone composite scaffold for in situ transfection application. Carbohydr Polym 2018; 183:29-36. [DOI: 10.1016/j.carbpol.2017.11.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/05/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022]
|
26
|
Walsh DP, Heise A, O’Brien FJ, Cryan SA. An efficient, non-viral dendritic vector for gene delivery in tissue engineering. Gene Ther 2017; 24:681-691. [DOI: 10.1038/gt.2017.58] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 05/25/2017] [Accepted: 06/16/2017] [Indexed: 12/19/2022]
|
27
|
García JR, García AJ. Biomaterial-mediated strategies targeting vascularization for bone repair. Drug Deliv Transl Res 2016; 6:77-95. [PMID: 26014967 DOI: 10.1007/s13346-015-0236-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Repair of non-healing bone defects through tissue engineering strategies remains a challenging feat in the clinic due to the aversive microenvironment surrounding the injured tissue. The vascular damage that occurs following a bone injury causes extreme ischemia and a loss of circulating cells that contribute to regeneration. Tissue-engineered constructs aimed at regenerating the injured bone suffer from complications based on the slow progression of endogenous vascular repair and often fail at bridging the bone defect. To that end, various strategies have been explored to increase blood vessel regeneration within defects to facilitate both tissue-engineered and natural repair processes. Developments that induce robust vascularization will need to consolidate various parameters including optimization of embedded therapeutics, scaffold characteristics, and successful integration between the construct and the biological tissue. This review provides an overview of current strategies as well as new developments in engineering biomaterials to induce reparation of a functional vascular supply in the context of bone repair.
Collapse
Affiliation(s)
- José R García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA. .,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
28
|
Lind T, Gustafson AM, Calounova G, Hu L, Rasmusson A, Jonsson KB, Wernersson S, Åbrink M, Andersson G, Larsson S, Melhus H, Pejler G. Increased Bone Mass in Female Mice Lacking Mast Cell Chymase. PLoS One 2016; 11:e0167964. [PMID: 27936149 PMCID: PMC5148084 DOI: 10.1371/journal.pone.0167964] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/24/2016] [Indexed: 12/22/2022] Open
Abstract
Here we addressed the potential impact of chymase, a mast-cell restricted protease, on mouse bone phenotype. We show that female mice lacking the chymase Mcpt4 acquired a persistent expansion of diaphyseal bone in comparison with wild type controls, reaching a 15% larger diaphyseal cross sectional area at 12 months of age. Mcpt4-/- mice also showed increased levels of a bone anabolic serum marker and higher periosteal bone formation rate. However, they were not protected from experimental osteoporosis, suggesting that chymase regulates normal bone homeostasis rather than the course of osteoporosis. Further, the absence of Mcpt4 resulted in age-dependent upregulation of numerous genes important for bone formation but no effects on osteoclast activity. In spite of the latter, Mcpt4-/- bones had increased cortical porosity and reduced endocortical mineralization. Mast cells were found periosteally and, notably, bone-proximal mast cells in Mcpt4-/- mice were degranulated to a larger extent than in wild type mice. Hence, chymase regulates degranulation of bone mast cells, which could affect the release of mast cell-derived factors influencing bone remodelling. Together, these findings reveal a functional impact of mast cell chymase on bone. Further studies exploring the possibility of using chymase inhibitors as a strategy to increase bone volume may be warranted.
Collapse
Affiliation(s)
- Thomas Lind
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden
- * E-mail:
| | - Ann-Marie Gustafson
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden
- Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| | - Gabriela Calounova
- Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| | - Lijuan Hu
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden
| | - Annica Rasmusson
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden
| | - Kenneth B. Jonsson
- Uppsala University Hospital, Department of Surgical Sciences, Uppsala, Sweden
| | - Sara Wernersson
- Swedish University of Agricultural Sciences, Department of Anatomy, Physiology and Biochemistry, Uppsala, Sweden
| | - Magnus Åbrink
- Swedish University of Agricultural Sciences, Department of Biomedical Science and Veterinary Public Health, Uppsala, Sweden
| | - Göran Andersson
- Karolinska Institute, Division of Pathology, Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sune Larsson
- Uppsala University Hospital, Department of Surgical Sciences, Uppsala, Sweden
| | - Håkan Melhus
- Uppsala University Hospital, Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala, Sweden
| | - Gunnar Pejler
- Uppsala University, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
- Swedish University of Agricultural Sciences, Department of Anatomy, Physiology and Biochemistry, Uppsala, Sweden
| |
Collapse
|
29
|
Ting M, Jefferies SR, Xia W, Engqvist H, Suzuki JB. Classification and Effects of Implant Surface Modification on the Bone: Human Cell-Based In Vitro Studies. J ORAL IMPLANTOL 2016; 43:58-83. [PMID: 27897464 DOI: 10.1563/aaid-joi-d-16-00079] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Implant surfaces are continuously being improved to achieve faster osseointegration and a stronger bone to implant interface. This review will present the various implant surfaces, the parameters for implant surface characterization, and the corresponding in vitro human cell-based studies determining the strength and quality of the bone-implant contact. These in vitro cell-based studies are the basis for animal and clinical studies and are the prelude to further reviews on how these surfaces would perform when subjected to the oral environment and functional loading.
Collapse
Affiliation(s)
- Miriam Ting
- 1 Temple University Kornberg School of Dentistry, Philadelphia, Pa
| | - Steven R Jefferies
- 2 Department of Restorative Dentistry, Temple University Kornberg School of Dentistry, Philadelphia, Pa
| | - Wei Xia
- 3 Department of Engineering Science, Uppsala University, Uppsala, Sweden
| | - Håkan Engqvist
- 3 Department of Engineering Science, Uppsala University, Uppsala, Sweden
| | - Jon B Suzuki
- 4 Department of Periodontology and Oral Implantology, Temple University Kornberg School of Dentistry, Philadelphia, Pa
| |
Collapse
|
30
|
Bagherifard S. Mediating bone regeneration by means of drug eluting implants: From passive to smart strategies. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 71:1241-1252. [PMID: 27987680 DOI: 10.1016/j.msec.2016.11.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/06/2016] [Accepted: 11/02/2016] [Indexed: 02/03/2023]
Abstract
In addition to excellent biocompatibility and mechanical performance, the new generation of bone and craniofacial implants are expected to proactively contribute to the regeneration process and dynamically interact with the host tissue. To this end, integration and sustained delivery of therapeutic agents has become a rapidly expanding area. The incorporated active molecules can offer supplementary features including promoting oteoconduction and angiogenesis, impeding bacterial infection and modulating host body reaction. Major limitations of the current practices consist of low drug stability overtime, poor control of release profile and kinetics as well as complexity of finding clinically appropriate drug dosage. In consideration of the multifaceted cascade of bone regeneration process, this research is moving towards dual/multiple drug delivery, where precise control on simultaneous or sequential delivery, considering the possible synergetic interaction of the incorporated bioactive factors is of utmost importance. Herein, recent advancements in fabrication of synthetic load bearing implants equipped with various drug delivery systems are reviewed. Smart drug delivery solutions, newly developed to provide higher tempo-spatial control on the delivery of the pharmaceutical agents for targeted and stimuli responsive delivery are highlighted. The future trend of implants with bone drug delivery mechanisms and the most common challenges hindering commercialization and the bench to bedside progress of the developed technologies are covered.
Collapse
Affiliation(s)
- Sara Bagherifard
- Politecnico di Milano, Department of Mechanical Engineering, Milan, Italy.
| |
Collapse
|
31
|
Raisin S, Belamie E, Morille M. Non-viral gene activated matrices for mesenchymal stem cells based tissue engineering of bone and cartilage. Biomaterials 2016; 104:223-37. [DOI: 10.1016/j.biomaterials.2016.07.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/14/2016] [Accepted: 07/16/2016] [Indexed: 12/22/2022]
|
32
|
D'Mello S, Atluri K, Geary SM, Hong L, Elangovan S, Salem AK. Bone Regeneration Using Gene-Activated Matrices. AAPS JOURNAL 2016; 19:43-53. [PMID: 27655418 DOI: 10.1208/s12248-016-9982-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/26/2016] [Indexed: 01/04/2023]
Abstract
Gene delivery to bone is a potential therapeutic strategy for directed, sustained, and regulated protein expression. Tissue engineering strategies for bone regeneration include delivery of proteins, genes (viral and non-viral-mediated delivery), and/or cells to the bone defect site. In addition, biomimetic scaffolds and scaffolds incorporating bone anabolic agents greatly enhance the bone repair process. Regional gene therapy has the potential of enhancing bone defect healing and bone regeneration by delivering osteogenic genes locally to the osseous lesions, thereby reducing systemic toxicity and the need for using supraphysiological dosages of therapeutic proteins. By implanting gene-activated matrices (GAMs), sustained gene expression and continuous osteogenic protein production in situ can be achieved in a way that stimulates osteogenesis and bone repair within osseous defects. Critical parameters substantially affecting the therapeutic efficacy of gene therapy include the choice of osteogenic transgene(s), selection of non-viral or viral vectors, the wound environment, and the selection of ex vivo and in vivo gene delivery strategies, such as GAMs. It is critical for gene therapy applications that clinically beneficial amounts of proteins are synthesized endogenously within and around the lesion in a sustained manner. It is therefore necessary that reliable and reproducible methods of gene delivery be developed and tested for their efficacy and safety before translating into clinical practice. Practical considerations such as the age, gender, and systemic health of patients and the nature of the disease process also need to be taken into account in order to personalize the treatments and progress towards developing a clinically applicable gene therapy for healing bone defects. This review discusses tissue engineering strategies to regenerate bone with specific focus on non-viral gene delivery systems.
Collapse
Affiliation(s)
- Sheetal D'Mello
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, 115 S. Grand Avenue, S228 PHAR, Iowa City, Iowa, 52242, USA
| | - Keerthi Atluri
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, 115 S. Grand Avenue, S228 PHAR, Iowa City, Iowa, 52242, USA
| | - Sean M Geary
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, 115 S. Grand Avenue, S228 PHAR, Iowa City, Iowa, 52242, USA
| | - Liu Hong
- Department of Prosthodontics, College of Dentistry, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Satheesh Elangovan
- Department of Periodontics, College of Dentistry, University of Iowa, 801 Newton Road, S464, Iowa City, Iowa, 52242, USA.
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, 115 S. Grand Avenue, S228 PHAR, Iowa City, Iowa, 52242, USA. .,Department of Periodontics, College of Dentistry, University of Iowa, 801 Newton Road, S464, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
33
|
Li J, Xu Q, Teng B, Yu C, Li J, Song L, Lai YX, Zhang J, Zheng W, Ren PG. Investigation of angiogenesis in bioactive 3-dimensional poly(d,l-lactide-co-glycolide)/nano-hydroxyapatite scaffolds by in vivo multiphoton microscopy in murine calvarial critical bone defect. Acta Biomater 2016; 42:389-399. [PMID: 27326916 DOI: 10.1016/j.actbio.2016.06.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 12/19/2022]
Abstract
UNLABELLED Reconstruction of critical size bone defects remains a major clinical challenge because of poor bone regeneration, which is usually due to poor angiogenesis during repair. Satisfactory vascularization is a prerequisite for the survival of grafts and the integration of new tissue with existing tissue. In this work, we investigated angiogenesis in 3D scaffolds by in vivo multiphoton microscopy during bone formation in a murine calvarial critical bone defect model and evaluated bone regeneration 8weeks post-implantation. The continuous release of bioactive lentiviral vectors (LV-pdgfb) from the scaffolds could be detected for 5days in vitro. In vivo, the released LV-pdgfb transfected adjacent cells and expressed PDGF-BB, facilitating angiogenesis and enhancing bone regeneration. The expression of both pdgfb and the angiogenesis-related genes vWF and VEGFR2 was significantly increased in the pdgfb gene-carrying scaffold (PHp) group. In addition, microCT scanning and histomorphology results proved that there was more new bone ingrowth in the PHp group than in the PLGA/nHA (PH) and control groups. MicroCT parameters, including BMD, BV/TV, Tb.Sp, and Tb.N indicated that there was significantly more new bone formation in the PHp group than in the other groups. With regard to neovascularization, 8weeks post-implantation, blood vessel areas (BVAs) were 9428±944μm(2), 4090±680.3μm(2), and none in the PHp, PH, and control groups, respectively. At each time point, BVAs in the PHp scaffolds were significantly higher than in the PH scaffolds. To our knowledge, this is the first use of multiphoton microscopy in bone tissue-engineering to investigate angiogenesis in scaffolds in vivo. This method represents a valuable tool for investigating neovascularization in bone scaffolds to determine if a certain scaffold is beneficial to neovascularization. We also proved that delivery of the pdgfb gene alone can improve both angiogenesis and bone regeneration Acronyms. STATEMENT OF SIGNIFICANCE Reconstruction of critical size bone defects remains a major clinical challenge because of poor bone regeneration, which is usually due to poor angiogenesis during repair. Satisfactory vascularization is a prerequisite for the survival of grafts and the integration of new tissue with existing tissue. In this work, we investigated angiogenesis in 3D scaffolds by in vivo multiphoton microscopy during bone formation in a murine calvarial critical bone defect model and evaluated bone regeneration 8weeks post-implantation. To verify that pdgfb-expressing vectors carried by the scaffolds can promote angiogenesis in 3D-printed scaffolds in vivo, we monitored angiogenesis within the implants by multiphoton microscopy. To our knowledge, this is the first study to dynamically investigate angiogenesis in bone tissue engineering scaffolds in vivo.
Collapse
Affiliation(s)
- Jian Li
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Qiang Xu
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Bin Teng
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Chen Yu
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Orthopedics Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Jian Li
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Orthopedics Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Liang Song
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yu-Xiao Lai
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jian Zhang
- Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Wei Zheng
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| | - Pei-Gen Ren
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
34
|
Keeney M, Chung MT, Zielins ER, Paik KJ, McArdle A, Morrison SD, Ransom RC, Barbhaiya N, Atashroo D, Jacobson G, Zare RN, Longaker MT, Wan DC, Yang F. Scaffold-mediated BMP-2 minicircle DNA delivery accelerated bone repair in a mouse critical-size calvarial defect model. J Biomed Mater Res A 2016; 104:2099-107. [PMID: 27059085 DOI: 10.1002/jbm.a.35735] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 12/31/2022]
Abstract
Scaffold-mediated gene delivery holds great promise for tissue regeneration. However, previous attempts to induce bone regeneration using scaffold-mediated non-viral gene delivery rarely resulted in satisfactory healing. We report a novel platform with sustained release of minicircle DNA (MC) from PLGA scaffolds to accelerate bone repair. MC was encapsulated inside PLGA scaffolds using supercritical CO2 , which showed prolonged release of MC. Skull-derived osteoblasts transfected with BMP-2 MC in vitro result in higher osteocalcin gene expression and mineralized bone formation. When implanted in a critical-size mouse calvarial defect, scaffolds containing luciferase MC lead to robust in situ protein production up to at least 60 days. Scaffold-mediated BMP-2 MC delivery leads to substantially accelerated bone repair as early as two weeks, which continues to progress over 12 weeks. This platform represents an efficient, long-term nonviral gene delivery system, and may be applicable for enhancing repair of a broad range of tissues types. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2099-2107, 2016.
Collapse
Affiliation(s)
- Michael Keeney
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305.,Department of Bioengineering, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305
| | - Michael T Chung
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Elizabeth R Zielins
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Kevin J Paik
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Adrian McArdle
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Shane D Morrison
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Ryan C Ransom
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Namrata Barbhaiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305.,Department of Bioengineering, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305
| | - David Atashroo
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Gunilla Jacobson
- Department of Chemistry, Stanford University, 333 Campus Drive Mudd Building, Room 121 Stanford, Stanford, California, 94305-4401
| | - Richard N Zare
- Department of Chemistry, Stanford University, 333 Campus Drive Mudd Building, Room 121 Stanford, Stanford, California, 94305-4401
| | - Michael T Longaker
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Derrick C Wan
- Department of Surgery, Plastic and Reconstructive Surgery Division, Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, 257 Campus Drive, Stanford University, Stanford, California, 94305-5148
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305.,Department of Bioengineering, Stanford University School of Medicine, Clark Center E-150, 300 Pasteur Drive, Edwards R105, MC5341, Stanford, California, 94305
| |
Collapse
|
35
|
Kim YD, Pofali P, Park TE, Singh B, Cho K, Maharjan S, Dandekar P, Jain R, Choi YJ, Arote R, Cho CS. Gene therapy for bone tissue engineering. Tissue Eng Regen Med 2016; 13:111-125. [PMID: 30603391 PMCID: PMC6170855 DOI: 10.1007/s13770-016-9063-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023] Open
Abstract
Gene therapy holds a great promise and has been extensively investigated to improve bone formation and regeneration therapies in bone tissue engineering. A variety of osteogenic genes can be delivered by combining different vectors (viral or non-viral), scaffolds and delivery methodologies. Ex vivo & in vivo gene enhanced tissue engineering approaches have led to successful osteogenic differentiation and bone formation. In this article, we review recent advances of gene therapy-based bone tissue engineering discussing strengths and weaknesses of various strategies as well as general overview of gene therapy.
Collapse
Affiliation(s)
- Young-Dong Kim
- Department of Molecular Genetics, School of Dentistry, Seoul National University, Seoul, Korea
| | - Prasad Pofali
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Tae-Eun Park
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Kihyun Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Sushila Maharjan
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Rohidas Arote
- Department of Molecular Genetics, School of Dentistry, Seoul National University, Seoul, Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
36
|
Morand DN, Davideau JL, Clauss F, Jessel N, Tenenbaum H, Huck O. Cytokines during periodontal wound healing: potential application for new therapeutic approach. Oral Dis 2016; 23:300-311. [PMID: 26945691 DOI: 10.1111/odi.12469] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/12/2016] [Accepted: 03/01/2016] [Indexed: 12/15/2022]
Abstract
Regeneration of periodontal tissues is one of the main goals of periodontal therapy. However, current treatment, including surgical approach, use of membrane to allow maturation of all periodontal tissues, or use of enamel matrix derivatives, presents limitations in their indications and outcomes leading to the development of new tissue engineering strategies. Several cytokines are considered as key molecules during periodontal destruction process. However, their role during each phase of periodontal wound healing remains unclear. Control and modulation of the inflammatory response and especially, release of cytokines or activation/inhibition in a time- and spatial-controlled manner may be a potential perspective for periodontal tissue engineering. The aim of this review was to summarize the specific role of several cytokines during periodontal wound healing and the potential therapeutic interest of inflammatory modulation for periodontal regeneration especially related to the expression sequence of cytokines.
Collapse
Affiliation(s)
- D N Morand
- INSERM (French National Institute of Health and Medical Research), UMR 1109, Osteoarticular and Dental Regenerative Nanomedicine Laboratory, Faculté de Médecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Department of periodontology, Dental Faculty, University of Strasbourg, Strasbourg, France
| | - J-L Davideau
- INSERM (French National Institute of Health and Medical Research), UMR 1109, Osteoarticular and Dental Regenerative Nanomedicine Laboratory, Faculté de Médecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Department of periodontology, Dental Faculty, University of Strasbourg, Strasbourg, France
| | - F Clauss
- INSERM (French National Institute of Health and Medical Research), UMR 1109, Osteoarticular and Dental Regenerative Nanomedicine Laboratory, Faculté de Médecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Department of periodontology, Dental Faculty, University of Strasbourg, Strasbourg, France
| | - N Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1109, Osteoarticular and Dental Regenerative Nanomedicine Laboratory, Faculté de Médecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - H Tenenbaum
- INSERM (French National Institute of Health and Medical Research), UMR 1109, Osteoarticular and Dental Regenerative Nanomedicine Laboratory, Faculté de Médecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Department of periodontology, Dental Faculty, University of Strasbourg, Strasbourg, France
| | - O Huck
- INSERM (French National Institute of Health and Medical Research), UMR 1109, Osteoarticular and Dental Regenerative Nanomedicine Laboratory, Faculté de Médecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Department of periodontology, Dental Faculty, University of Strasbourg, Strasbourg, France
| |
Collapse
|
37
|
Komatsu K, Shibata T, Shimada A, Ideno H, Nakashima K, Tabata Y, Nifuji A. Cationized gelatin hydrogels mixed with plasmid DNA induce stronger and more sustained gene expression than atelocollagen at calvarial bone defects in vivo. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2016; 27:419-30. [PMID: 26848778 DOI: 10.1080/09205063.2016.1139486] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gene transduction of exogenous factors at local sites in vivo is a promising approach to promote regeneration of tissue defects owing to its simplicity and capacity for expression of a variety of genes. Gene transduction by viral vectors is highly efficient; however, there are safety concerns associated with viruses. As a method for nonviral gene transduction, plasmid DNA delivery is safer and simpler, but requires an efficient carrier substance. Here, we aimed to develop a simple, efficient method for bone regeneration by gene transduction and to identify optimal conditions for plasmid DNA delivery at bone defect sites. We focused on carrier substances and compared the efficiencies of two collagen derivatives, atelocollagen, and gelatin hydrogel, as substrates for plasmid DNA delivery in vivo. To assess the efficiencies of these substrates, we examined exogenous expression of green fluorescence protein (GFP) by fluorescence microscopy, polymerase chain reaction, and immunohistochemistry. GFP expression at the bone defect site was higher when gelatin hydrogel was used as a substrate to deliver plasmids than when atelocollagen was used. Moreover, the gelatin hydrogel was almost completely absorbed at the defect site, whereas some atelocollagen remained. When a plasmid harboring bone morphogenic protein 2 was delivered with the substrate to bony defect sites, more new bone formation was observed in the gelatin group than in the atelocollagen group. These results suggested that the gelatin hydrogel was more efficient than atelocollagen as a substrate for local gene delivery and may be a superior material for induction of bone regeneration.
Collapse
Affiliation(s)
- K Komatsu
- a Department of Pharmacology , Tsurumi University School of Dental Medicine , Yokohama , Japan
| | - T Shibata
- a Department of Pharmacology , Tsurumi University School of Dental Medicine , Yokohama , Japan
| | - A Shimada
- a Department of Pharmacology , Tsurumi University School of Dental Medicine , Yokohama , Japan
| | - H Ideno
- a Department of Pharmacology , Tsurumi University School of Dental Medicine , Yokohama , Japan
| | - K Nakashima
- a Department of Pharmacology , Tsurumi University School of Dental Medicine , Yokohama , Japan
| | - Y Tabata
- b Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences , Kyoto University , Kyoto , Japan
| | - A Nifuji
- a Department of Pharmacology , Tsurumi University School of Dental Medicine , Yokohama , Japan
| |
Collapse
|
38
|
D'Mello S, Elangovan S, Salem AK. FGF2 gene activated matrices promote proliferation of bone marrow stromal cells. Arch Oral Biol 2015; 60:1742-9. [PMID: 26433191 DOI: 10.1016/j.archoralbio.2015.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 07/29/2015] [Accepted: 09/07/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND In this study, we report on the results from the development and early in vitro testing of a gene activated matrix encoding basic human fibroblast growth factor 2 (FGF2) in bone marrow stromal cells (BMSCs). METHODS Polyethylenimine (PEI), a cationic polymer, was utilized as a gene delivery vector and collagen scaffolds were used as the carrier to deliver the PEI-pDNA nano-sized complexes (nanoplexes) encoding the FGF2 protein. Initially, the BMSCs were transfected in vitro with the PEI-pFGF2 nanoplexes, prepared at a N/P ratio of 10, with cells alone and naked DNA as controls. This was followed by transfection experiments using collagen scaffold containing complexes, with the scaffold alone as a control. The transfection efficacy of the nanoplexes was assessed using ELISA for the determination of FGF2 protein expressed by the transfected cells. The functionality of transfection was assessed by evaluating cellular recruitment, attachment, and proliferation of BMSCs on the scaffold using imaging techniques. RESULTS BMSCs transfected with the PEI-pFGF2 nanoplexes (either alone or within the scaffold) led to higher expression of FGF2, compared to controls. Scanning electron microscopy and confocal imaging confirmed the recruitment and attachment of BMSCs to scaffolds containing the PEI-pFGF2 nanoplexes. Confocal microscopy showed a significantly higher number of proliferating cells within PEI-pFGF2 nanoplex-loaded scaffolds than with empty scaffolds. CONCLUSIONS This first in vitro evaluation in BMSCs provides evidence that gene activated matrices (GAMs) encoding the FGF2 protein may have strong translational potential for clinical applications that require enhanced osseous and periodontal tissue regeneration.
Collapse
Affiliation(s)
- Sheetal D'Mello
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, IA, USA
| | - Satheesh Elangovan
- Department of Periodontics, College of Dentistry, University of Iowa, IA, USA.
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, IA, USA; Department of Periodontics, College of Dentistry, University of Iowa, IA, USA.
| |
Collapse
|
39
|
Herrmann M, Verrier S, Alini M. Strategies to Stimulate Mobilization and Homing of Endogenous Stem and Progenitor Cells for Bone Tissue Repair. Front Bioeng Biotechnol 2015; 3:79. [PMID: 26082926 PMCID: PMC4451737 DOI: 10.3389/fbioe.2015.00079] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/16/2015] [Indexed: 12/17/2022] Open
Abstract
The gold standard for the treatment of critical-size bone defects is autologous or allogenic bone graft. This has several limitations including donor site morbidity and the restricted supply of graft material. Cell-based tissue engineering strategies represent an alternative approach. Mesenchymal stem cells (MSCs) have been considered as a source of osteoprogenitor cells. More recently, focus has been placed on the use of endothelial progenitor cells (EPCs), since vascularization is a critical step in bone healing. Although many of these approaches have demonstrated effectiveness for bone regeneration, cell-based therapies require time consuming and cost-expensive in vitro cell expansion procedures. Accordingly, research is becoming increasingly focused on the homing and stimulation of native cells. The stromal cell-derived factor-1 (SDF-1) - CXCR4 axis has been shown to be critical for the recruitment of MSCs and EPCs. Vascular endothelial growth factor (VEGF) is a key factor in angiogenesis and has been targeted in many studies. Here, we present an overview of the different approaches for delivering homing factors to the defect site by absorption or incorporation to biomaterials, gene therapy, or via genetically manipulated cells. We further review strategies focusing on the stimulation of endogenous cells to support bone repair. Finally, we discuss the major challenges in the treatment of critical-size bone defects and fracture non-unions.
Collapse
Affiliation(s)
| | | | - Mauro Alini
- AO Research Institute Davos , Davos , Switzerland
| |
Collapse
|
40
|
Gupta K, Singh S, Garg KN. Gene therapy in dentistry: Tool of genetic engineering. Revisited. Arch Oral Biol 2015; 60:439-46. [DOI: 10.1016/j.archoralbio.2014.11.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 11/26/2014] [Accepted: 11/27/2014] [Indexed: 01/17/2023]
|
41
|
Subramanian G, Bialorucki C, Yildirim-Ayan E. Nanofibrous yet injectable polycaprolactone-collagen bone tissue scaffold with osteoprogenitor cells and controlled release of bone morphogenetic protein-2. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 51:16-27. [PMID: 25842103 DOI: 10.1016/j.msec.2015.02.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/21/2015] [Accepted: 02/23/2015] [Indexed: 01/01/2023]
Abstract
In this work, we developed a nanofibrous, yet injectable orthobiologic tissue scaffold that is capable of hosting osteoprogenitor cells and controlling kinetic release profile of the encapsulated pro-osteogenic factor without diminishing its bioactivity over 21days. This innovative injectable scaffold was synthesized by incorporating electrospun and subsequently O2 plasma-functionalized polycaprolactone (PCL) nanofibers within the collagen type-I solution along with MC3T3-E1 cells (pre-osteoblasts) and bone morphogenetic protein-2 (BMP2). Through changing the PCL nanofiber concentration within the injectable scaffolds, we were able to tailor the mechanical strength, protein retention capacity, bioactivity preservation, and osteoinductive potential of the scaffolds. The nanofibrous internal structure of the scaffold allowed us to use a low dose of BMP2 (200ng/ml) to achieve osteoblastic differentiation in in vitro culture. The osteogenesis capacity of the injectable scaffolds were evaluated though measuring MC3T3-E1 cell proliferation, ALP activity, matrix mineralization, and early- and late-osteoblast specific gene expression profiles over 21days. The results demonstrated that the nanofibrous injectable scaffold provides not only an osteoinductive environment for osteoprogenitor cells to differentiate, but also a suitable biomechanical and biochemical environment to act as a reservoir for osteogenic factors with controlled release profile.
Collapse
Affiliation(s)
- Gayathri Subramanian
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA
| | - Callan Bialorucki
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, OH 43614, USA.
| |
Collapse
|
42
|
Pilipchuk SP, Plonka AB, Monje A, Taut AD, Lanis A, Kang B, Giannobile WV. Tissue engineering for bone regeneration and osseointegration in the oral cavity. Dent Mater 2015; 31:317-38. [PMID: 25701146 DOI: 10.1016/j.dental.2015.01.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 12/19/2014] [Accepted: 01/11/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The focus of this review is to summarize recent advances on regenerative technologies (scaffolding matrices, cell/gene therapy and biologic drug delivery) to promote reconstruction of tooth and dental implant-associated bone defects. METHODS An overview of scaffolds developed for application in bone regeneration is presented with an emphasis on identifying the primary criteria required for optimized scaffold design for the purpose of regenerating physiologically functional osseous tissues. Growth factors and other biologics with clinical potential for osteogenesis are examined, with a comprehensive assessment of pre-clinical and clinical studies. Potential novel improvements to current matrix-based delivery platforms for increased control of growth factor spatiotemporal release kinetics are highlighting including recent advancements in stem cell and gene therapy. RESULTS An analysis of existing scaffold materials, their strategic design for tissue regeneration, and use of growth factors for improved bone formation in oral regenerative therapies results in the identification of current limitations and required improvements to continue moving the field of bone tissue engineering forward into the clinical arena. SIGNIFICANCE Development of optimized scaffolding matrices for the predictable regeneration of structurally and physiologically functional osseous tissues is still an elusive goal. The introduction of growth factor biologics and cells has the potential to improve the biomimetic properties and regenerative potential of scaffold-based delivery platforms for next-generation patient-specific treatments with greater clinical outcome predictability.
Collapse
Affiliation(s)
- Sophia P Pilipchuk
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, 1101 Beal Avenue, Ann Arbor, MI 48109, USA.
| | - Alexandra B Plonka
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, 1011 N. University Avenue, Ann Arbor, MI 48109, USA.
| | - Alberto Monje
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, 1011 N. University Avenue, Ann Arbor, MI 48109, USA.
| | - Andrei D Taut
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, 1011 N. University Avenue, Ann Arbor, MI 48109, USA.
| | - Alejandro Lanis
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, 1011 N. University Avenue, Ann Arbor, MI 48109, USA.
| | - Benjamin Kang
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, 1011 N. University Avenue, Ann Arbor, MI 48109, USA.
| | - William V Giannobile
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, 1011 N. University Avenue, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, 1101 Beal Avenue, Ann Arbor, MI 48109, USA.
| |
Collapse
|
43
|
Umebayashi M, Sumita Y, Kawai Y, Watanabe S, Asahina I. Gene-Activated Matrix Comprised of Atelocollagen and Plasmid DNA Encoding BMP4 or Runx2 Promotes Rat Cranial Bone Augmentation. Biores Open Access 2015; 4:164-74. [PMID: 26309793 PMCID: PMC4497668 DOI: 10.1089/biores.2014.0057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
To date, therapeutic method for in vivo gene delivery has not been established on bone engineering though its potential usefulness has been suggested. For clinical applications, an effective condition should be developed to transfer the genes in vivo without any transfection reagents or virus vectors. In this study, to facilitate the clinical setting of this strategy, particularly aimed at atrophic bone repair, we simply investigated whether manufactured gene-activated matrix (GAM) with atelocollagen containing a certain amount of plasmid (p) DNA encoding osteogenic proteins could augment the cranial bone in rat. GAMs were manufactured by mixing 0.02, 0.1, or 1 mg of AcGFP plasmid vectors harboring cDNA of BMP4 (pBMP4) or Runx2 (pRunx2) with 2% bovine atelocollagen and β-tricalcium phosphate granules. Before manufacturing GAMs, to determine the biological activity of generated pDNAs, we confirmed GFP expression and increased level of alkaline phosphatase activities in MC3T3-E1 cells transfected with pBMP4 or pRunx2 during culture. Then, GAMs were lyophilized and transplanted to onlay placement on the cranium. At 2 weeks of transplantation, GFP-expressing cells could be detectable in only GAMs containing 1 mg of AcGFP plasmid vectors. Then, at 4 weeks, significant bone formation was recognized in GAMs containing 1 mg of pDNAs encoding BMP4 or Runx2 but not in 0.02 or 0.1 mg of GAMs. These newly formed bone tissues surrounded by osteocalcin-stained area were augmented markedly until 8 weeks after transplantation. In contrast, minimal bone formation was observed in GAMs without harboring cDNA of osteogenic proteins. Meanwhile, when GAMs were transplanted to the cranial bone defect, bone formation was detectable in specimens containing 1 mg of pBMP4 or pRunx2 at 8 weeks as well. Thus, atelocollagen-based GAM reliably could form the engineered bone even for the vertical augmentation when containing a certain amount of plasmid vectors encoding osteogenic proteins. This study supports facilitating the clinical application of GAM for bone engineering.
Collapse
Affiliation(s)
- Mayumi Umebayashi
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University , Nagasaki, Japan
| | - Yoshinori Sumita
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University , Nagasaki, Japan
| | - Yousuke Kawai
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University , Nagasaki, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, Institute of Medical Science, The University of Tokyo , Tokyo, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University , Nagasaki, Japan
| |
Collapse
|
44
|
Makhdom AM, Nayef L, Tabrizian M, Hamdy RC. The potential roles of nanobiomaterials in distraction osteogenesis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1-18. [DOI: 10.1016/j.nano.2014.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/25/2014] [Accepted: 05/16/2014] [Indexed: 10/25/2022]
|
45
|
Yue J, Wu J, Liu D, Zhao X, Lu WW. BMP2 gene delivery to bone mesenchymal stem cell by chitosan-g-PEI nonviral vector. NANOSCALE RESEARCH LETTERS 2015; 10:203. [PMID: 25977673 PMCID: PMC4420764 DOI: 10.1186/s11671-015-0906-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/18/2015] [Indexed: 05/21/2023]
Abstract
Nanotechnology has made a significant impact on the development of nanomedicine. Nonviral vectors have been attracting more attention for the advantage of biosafety in gene delivery. Polyethylenimine (PEI)-conjugated chitosan (chitosan-g-PEI) emerged as a promising nonviral vector and has been demonstrated in many tumor cells. However, there is a lack of study focused on the behavior of this vector in stem cells which hold great potential in regenerative medicine. Therefore, in this study, in vitro gene delivering effect of chitosan-g-PEI was investigated in bone marrow stem cells. pIRES2-ZsGreen1-hBMP2 dual expression plasmid containing both the ZsGreen1 GFP reporter gene and the BMP2 functional gene was constructed for monitoring the transgene expression level. Chitosan-g-PEI-mediated gene transfer showed 17.2% of transfection efficiency and more than 80% of cell viability in stem cells. These values were higher than that of PEI. The expression of the delivered BMP2 gene in stem cells enhanced the osteogenic differentiation. These results demonstrated that chitosan-g-PEI is capable of applying in delivering gene to stem cells and providing potential applications in stem cell-based gene therapy.
Collapse
Affiliation(s)
- Jianhui Yue
- />Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
- />Shenzhen Key Laboratory of Marine Biomedical Materials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
| | - Jun Wu
- />Department of Orthopaedic and Traumatology, The University of Hong Kong, 21 Sassoon Rd., Pokfulam, Hong Kong, 999077 People’s Republic of China
| | - Di Liu
- />Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
- />Department of Pharmacology, Harbin Medical University, 157 Baojian Rd., Harbin, 150081 People’s Republic of China
| | - Xiaoli Zhao
- />Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
- />Shenzhen Key Laboratory of Marine Biomedical Materials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
| | - William W Lu
- />Department of Orthopaedic and Traumatology, The University of Hong Kong, 21 Sassoon Rd., Pokfulam, Hong Kong, 999077 People’s Republic of China
| |
Collapse
|
46
|
Fang YL, Chen XG, W T G. Gene delivery in tissue engineering and regenerative medicine. J Biomed Mater Res B Appl Biomater 2014; 103:1679-99. [PMID: 25557560 DOI: 10.1002/jbm.b.33354] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/07/2014] [Accepted: 11/18/2014] [Indexed: 12/13/2022]
Abstract
As a promising strategy to aid or replace tissue/organ transplantation, gene delivery has been used for regenerative medicine applications to create or restore normal function at the cell and tissue levels. Gene delivery has been successfully performed ex vivo and in vivo in these applications. Excellent proliferation capabilities and differentiation potentials render certain cells as excellent candidates for ex vivo gene delivery for regenerative medicine applications, which is why multipotent and pluripotent cells have been intensely studied in this vein. In this review, gene delivery is discussed in detail, along with its applications to tissue engineering and regenerative medicine. A definition of a stem cell is compared to a definition of a stem property, and both provide the foundation for an in-depth look at gene delivery investigations from a germ lineage angle.
Collapse
Affiliation(s)
- Y L Fang
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| | - X G Chen
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| | - Godbey W T
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| |
Collapse
|
47
|
Mitragotri S, Burke PA, Langer R. Overcoming the challenges in administering biopharmaceuticals: formulation and delivery strategies. Nat Rev Drug Discov 2014; 13:655-72. [PMID: 25103255 PMCID: PMC4455970 DOI: 10.1038/nrd4363] [Citation(s) in RCA: 1086] [Impact Index Per Article: 108.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The formulation and delivery of biopharmaceutical drugs, such as monoclonal antibodies and recombinant proteins, poses substantial challenges owing to their large size and susceptibility to degradation. In this Review we highlight recent advances in formulation and delivery strategies--such as the use of microsphere-based controlled-release technologies, protein modification methods that make use of polyethylene glycol and other polymers, and genetic manipulation of biopharmaceutical drugs--and discuss their advantages and limitations. We also highlight current and emerging delivery routes that provide an alternative to injection, including transdermal, oral and pulmonary delivery routes. In addition, the potential of targeted and intracellular protein delivery is discussed.
Collapse
Affiliation(s)
- Samir Mitragotri
- Department of Chemical Engineering, Center for Bioengineering, University of California, Santa Barbara, California 92106, USA
| | - Paul A Burke
- Burke Bioventures LLC, 277 Broadway, Cambridge, Massachusetts 02139, USA
| | - Robert Langer
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
48
|
Nguyen MK, Alsberg E. Bioactive factor delivery strategies from engineered polymer hydrogels for therapeutic medicine. Prog Polym Sci 2014; 39:1236-1265. [PMID: 25242831 PMCID: PMC4167348 DOI: 10.1016/j.progpolymsci.2013.12.001] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Polymer hydrogels have been widely explored as therapeutic delivery matrices because of their ability to present sustained, localized and controlled release of bioactive factors. Bioactive factor delivery from injectable biopolymer hydrogels provides a versatile approach to treat a wide variety of diseases, to direct cell function and to enhance tissue regeneration. The innovative development and modification of both natural-(e.g., alginate (ALG), chitosan, hyaluronic acid (HA), gelatin, heparin (HEP), etc.) and synthetic-(e.g., polyesters, polyethyleneimine (PEI), etc.) based polymers has resulted in a variety of approaches to design drug delivery hydrogel systems from which loaded therapeutics are released. This review presents the state-of-the-art in a wide range of hydrogels that are formed though self-assembly of polymers and peptides, chemical crosslinking, ionic crosslinking and biomolecule recognition. Hydrogel design for bioactive factor delivery is the focus of the first section. The second section then thoroughly discusses release strategies of payloads from hydrogels for therapeutic medicine, such as physical incorporation, covalent tethering, affinity interactions, on demand release and/or use of hybrid polymer scaffolds, with an emphasis on the last 5 years.
Collapse
Affiliation(s)
- Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 204 Wickenden, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 204 Wickenden, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Orthopaedic Surgery, Case Western Reserve University, 204 Wickenden, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
49
|
Sustained localized presentation of RNA interfering molecules from in situ forming hydrogels to guide stem cell osteogenic differentiation. Biomaterials 2014; 35:6278-6286. [PMID: 24831973 DOI: 10.1016/j.biomaterials.2014.04.048] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/13/2014] [Indexed: 02/07/2023]
Abstract
To date, RNA interfering molecules have been used to differentiate stem cells on two-dimensional (2D) substrates that do not mimic three-dimensional (3D) microenvironments in the body. Here, in situ forming poly(ethylene glycol) (PEG) hydrogels were engineered for controlled, localized and sustained delivery of RNA interfering molecules to differentiate stem cells encapsulated within the 3D polymer network. RNA interfering molecules were released from the hydrogels in a sustained and controlled manner over the course of 3-6 weeks, and exhibited high bioactivity. Importantly, it was demonstrated that the delivery of siRNA and/or miRNA from the hydrogel constructs enhanced the osteogenic differentiation of encapsulated stem cells. Prolonged delivery of siRNA and/or miRNA from this polymeric scaffold permitted extended regulation of cell behavior, unlike traditional siRNA experiments performed in vitro. This approach presents a powerful new methodology for controlling cell fate, and is promising for multiple applications in tissue engineering and regenerative medicine.
Collapse
|
50
|
Power SM, Matic DB, Holdsworth DW. Accounting for cranial vault growth in experimental design. Oral Surg Oral Med Oral Pathol Oral Radiol 2014; 117:e325-e329. [DOI: 10.1016/j.oooo.2012.08.460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 08/28/2012] [Indexed: 11/16/2022]
|