1
|
Das P, Maresch M, Dey N, Sulaiman N, Ashour AG, Ammar HM, Basem M, Al Muharraqi MA, McGrath M, Jacob MV, O'Brien FJ, Keogh MB. Programmed wound healing in aged skin may be enhanced by mesenchymal cell loaded gene-activated scaffolds. APL Bioeng 2025; 9:026112. [PMID: 40290726 PMCID: PMC12033048 DOI: 10.1063/5.0240504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Aging can prolong the wound healing and is associated with decline in stem cells, delays in cellular migration, and lower vascularization. Tissue engineering has largely evolved to incorporate advanced three-dimensional wound dressings, scaffolds, and hydrogels that may be seeded with mesenchymal stromal cells (MSCs) to foster an environment conducive to regeneration and enhance the healing process. The effectiveness of stem cell-seeded scaffolds can be improved by incorporating activating agents such as nucleic acids resulting in gene-activated scaffolds (GAS), thereby facilitating targeted wound healing in aged patients. In this study, we assess the in vivo wound healing potential of a promising MSC seeded gene-activated collagen scaffold, containing the anti-fibrotic agent β-klotho and pro-angiogenic stromal derived factor (SDF-1α) in aged male Sprague Dawley rats (20-24 month old). A MSC cell loaded split skin model compared MSC only with the clinical standard dressing +Jelonet, MSCs +gene-free collagen scaffold, and MSCs +SDF-1α/β-klotho dual gene-activated collagen scaffold up to 21 days. Our results showed wound healing in all groups except in MSC +Jelonet which showed scab formation with exudate. MSC only group healed primarily via fibrotic contraction. In contrast, the scaffold groups showed host tissue integration and a redistribution of extracellular matrix proteins, less contraction, and complete re-epithelized wounds at day 21. The dual GAS displayed programmed wound healing with the greatest neo-vascularization CD31 expression. In conclusion, wound healing in aged rats can be effectively modulated when MSCs are loaded on biocompatible collagen scaffolds, particularly when these scaffolds are loaded with anti-fibrotic and pro-angiogenic factors. This approach enhances blood vessel formation while reducing fibrosis, suggesting a promising potential for programmed wound healing strategies in aged chronic wounds.
Collapse
Affiliation(s)
- Priya Das
- TERG Bahrain, School of Postgraduate Studies and Research, Royal College of Surgeons in Ireland, Manama, Kingdom of Bahrain
| | | | | | - Noof Sulaiman
- TERG Bahrain, School of Postgraduate Studies and Research, Royal College of Surgeons in Ireland, Manama, Kingdom of Bahrain
| | | | | | | | | | - Matthew McGrath
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | - Michael B. Keogh
- Present address: Human Biology & Director of Research Laboratories, School of Medicine, RCSI P.O. Box 15503, Adliya, Kingdom of Bahrain. Author to whom correspondence should be addressed:. Tel.: +973 17351450
| |
Collapse
|
2
|
Pipis N, James BD, Allen JB. Multifunctional DNA-Collagen Biomaterials: Developmental Advances and Biomedical Applications. ACS Biomater Sci Eng 2025; 11:1253-1268. [PMID: 39869382 PMCID: PMC11897955 DOI: 10.1021/acsbiomaterials.4c01475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
The complexation of nucleic acids and collagen forms a platform biomaterial greater than the sum of its parts. This union of biomacromolecules merges the extracellular matrix functionality of collagen with the designable bioactivity of nucleic acids, enabling advances in regenerative medicine, tissue engineering, gene delivery, and targeted therapy. This review traces the historical foundations and critical applications of DNA-collagen complexes and highlights their capabilities, demonstrating them as biocompatible, bioactive, and tunable platform materials. These complexes form structures across length scales, including nanoparticles, microfibers, and hydrogels, a process controlled by the relative amount of each component and the type of nucleic acid and collagen. The broad distribution of different types of collagen within the body contributes to the extensive biological relevance of DNA-collagen complexes. Functional nucleic acids can form these complexes, such as siRNA, antisense oligonucleotides, DNA origami nanostructures, and, in particular, single-stranded DNA aptamers, often distinguished by their rapid self-assembly at room temperature and formation without external stimuli and modifications. The simple and seamless integration of nucleic acids within collagenous matrices enhances biomimicry and targeted bioactivity, and provides stability against enzymatic degradation, positioning DNA-collagen complexes as an advanced biomaterial system for many applications including angiogenesis, bone tissue regeneration, wound healing, and more.
Collapse
Affiliation(s)
- Nikolaos Pipis
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Bryan D. James
- Department
of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Josephine B. Allen
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department
of Materials Science & Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
3
|
Song H, Hao D, Zhou J, Farmer D, Wang A. Development of pro-angiogenic skin substitutes for wound healing. Wound Repair Regen 2024; 32:208-216. [PMID: 38308588 DOI: 10.1111/wrr.13154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/13/2023] [Accepted: 12/12/2023] [Indexed: 02/05/2024]
Abstract
Wounds pose significant challenges to public health, primarily due to the loss of the mechanical integrity and barrier function of the skin and impaired angiogenesis, causing physical morbidities and psychological trauma to affect patients. Reconstructing the vasculature of the wound bed is crucial for promoting wound healing, reducing scar formation and enhancing the quality of life for patients. The development of pro-angiogenic skin substitutes has emerged as a promising strategy to facilitate vascularization and expedite the healing process of burn wounds. This review provides an overview of the various types of skin substitutes employed in wound healing, explicitly emphasising those designed to enhance angiogenesis. Synthetic scaffolds, biological matrices and tissue-engineered constructs incorporating stem cells and primary cells, cell-derived extracellular vesicles (EVs), pro-angiogenic growth factors and peptides, as well as gene therapy-based skin substitutes are thoroughly examined. The review summarises the existing challenges, future directions and potential innovations in pro-angiogenic dressing for skin substitutes. It highlights the need for continued research to develop new technologies and combine multiple strategies and factors, and to overcome obstacles and advance the field, ultimately leading to improved outcomes for wound patients.
Collapse
Affiliation(s)
- Hengyue Song
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Dake Hao
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Diana Farmer
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
- Department of Biomedical Engineering, UC Davis, Davis, California, USA
| |
Collapse
|
4
|
Sharma P, Kumar A, Agarwal T, Dey AD, Moghaddam FD, Rahimmanesh I, Ghovvati M, Yousefiasl S, Borzacchiello A, Mohammadi A, Yella VR, Moradi O, Sharifi E. Nucleic acid-based therapeutics for dermal wound healing. Int J Biol Macromol 2022; 220:920-933. [PMID: 35987365 DOI: 10.1016/j.ijbiomac.2022.08.099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 02/06/2023]
Abstract
Non-healing wounds have long been the subject of scientific and clinical investigations. Despite breakthroughs in understanding the biology of delayed wound healing, only limited advances have been made in properly treating wounds. Recently, research into nucleic acids (NAs) such as small-interfering RNA (siRNA), microRNA (miRNA), plasmid DNA (pDNA), aptamers, and antisense oligonucleotides (ASOs) has resulted in the development of a latest therapeutic strategy for wound healing. In this regard, dendrimers, scaffolds, lipid nanoparticles, polymeric nanoparticles, hydrogels, and metal nanoparticles have all been explored as NA delivery techniques. However, the translational possibility of NA remains a substantial barrier. As a result, different NAs must be identified, and their distribution method must be optimized. This review explores the role of NA-based therapeutics in various stages of wound healing and provides an update on the most recent findings in the development of NA-based nanomedicine and biomaterials, which may offer the potential for the invention of novel therapies for this long-term condition. Further, the challenges and potential for miRNA-based techniques to be translated into clinical applications are also highlighted.
Collapse
Affiliation(s)
- Preety Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; Government Pharmacy College Kangra, Nagrota Bhagwan, Himachal Pradesh, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Tarun Agarwal
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, 00133 Rome, Italy
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Mahsa Ghovvati
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Satar Yousefiasl
- School of Dentistry, Hamadan University of Medical Sciences, Hamadan 6517838736, Iran
| | - Assunta Borzacchiello
- Institute for Polymers, Composites, and Biomaterials (IPCB), National Research Council (CNR), Naples 80125, Italy
| | - Abbas Mohammadi
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Venkata Rajesh Yella
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Omid Moradi
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University, 374-37515 Tehran, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan 6517838736, Iran.
| |
Collapse
|
5
|
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: 196] [Impact Index Per Article: 49.0] [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
|
6
|
Effect of PDGF-B Gene-Activated Acellular Matrix and Mesenchymal Stem Cell Transplantation on Full Thickness Skin Burn Wound in Rat Model. Tissue Eng Regen Med 2020; 18:235-251. [PMID: 33145744 DOI: 10.1007/s13770-020-00302-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/10/2020] [Accepted: 09/16/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Full thickness burn wounds are lack of angiogenesis, cell migration, epithelialisation and finally scar tissue formation. Tissue engineered composite graft can provide sustained release of growth factor and promote the wound healing by cell migration, early angiogenesis and proliferation of extracellular matrix and wound remodeling. The objective of this study was to evaluate the gene embedded (pDNA-platelet-derived growth factor, PDGF-B) porcine acellular urinary bladder matrix with transfected mesenchymal stem cells (rBMSC) on healing of full thickness burn wound in rat model. METHODS Full thickness burn wound of 2 × 2 cm size was created in dorsum of rat model under general anesthesia. Burn wounds were treated with silver sulfadiazine; porcine acellular urinary bladder matrix (PAUBM); PAUBM transfected with pDNA-PDGF-B; PAUBM seeded with rBMSC; PAUBM seeded with rBMSC transfected with pDNA-PDGF-B in groups A, B, C, D and E respectively. The wound healing was assessed based on clinical, macroscopically, immunologically, histopathological and RT-qPCR parameters. RESULTS Wound was significantly healed in group E and group D with early extracellular matrix deposition, enhanced granulation tissue formation and early angiogenesis compared to all other groups. The immunologic response against porcine acellular matrix showed that PDGF-B gene activated matrix along with stem cell group showed less antibody titer against acellular matrix than other groups in all intervals. PDGF gene activated matrix releasing the PDGF-B and promote the healing of full thickness burn wound with neovascularization and neo tissue formation. PDGF gene also enhances secretion of other growth factors results in PDGF mediated regenerative activities. This was confirmed in RT-qPCR at various time intervals. CONCLUSION Gene activated matrix encoded for PDGF-B protein transfected stem cells have been clinically proven for early acceleration of angiogenesis and tissue regeneration in burn wounds in rat models. Evaluation of PDGF-B gene-activated acellular matrix and mesenchymal stem cell in full thickness skin burn wound in rat.
Collapse
|
7
|
Sarkar T, Sarkar S, Gangopadhyay DN. Gene Therapy and its Application in Dermatology. Indian J Dermatol 2020; 65:341-350. [PMID: 33165431 PMCID: PMC7640808 DOI: 10.4103/ijd.ijd_323_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Gene therapy is an experimental technique to treat genetic diseases. It is based on the introduction of nucleic acid with the help of a vector, into a diseased cell or tissue, to correct the gene expression and thus prevent, halt, or reverse a pathological process. It is a promising treatment approach for genetic diseases, inherited diseases, vaccination, cancer, immunomodulation, as well as healing of some refractory ulcers. Both viral and nonviral vectors can be used to deliver the correct gene. An ideal vector should have the ability for sustained gene expression, acceptable coding capacity, high transduction efficiency, and devoid of mutagenicity. There are different techniques of vector delivery, but these techniques are still under research for assessment of their safety and effectiveness. The major challenges of gene therapy are immunogenicity, mutagenicity, and lack of sustainable therapeutic benefit. Despite these constraints, therapeutic success was obtained in a few genetic and inherited skin diseases. Skin being the largest, superficial, easily accessible and assessable organ of the body, may be a promising target for gene therapy research in the recent future.
Collapse
Affiliation(s)
- Tanusree Sarkar
- From Department of Dermatology, Burdwan Medical College, West Bengal, India
| | - Somenath Sarkar
- Department of Dermatology, B. S Medical College, West Bengal, India
| | | |
Collapse
|
8
|
Zhang N, Chin JS, Chew SY. Localised non-viral delivery of nucleic acids for nerve regeneration in injured nervous systems. Exp Neurol 2018; 319:112820. [PMID: 30195695 DOI: 10.1016/j.expneurol.2018.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023]
Abstract
Axons damaged by traumatic injuries are often unable to spontaneously regenerate in the adult central nervous system (CNS). Although the peripheral nervous system (PNS) has some regenerative capacity, its ability to regrow remains limited across large lesion gaps due to scar tissue formation. Nucleic acid therapy holds the potential of improving regeneration by enhancing the intrinsic growth ability of neurons and overcoming the inhibitory environment that prevents neurite outgrowth. Nucleic acids modulate gene expression by over-expression of neuronal growth factor or silencing growth-inhibitory molecules. Although in vitro outcomes appear promising, the lack of efficient non-viral nucleic acid delivery methods to the nervous system has limited the application of nucleic acid therapeutics to patients. Here, we review the recent development of efficient non-viral nucleic acid delivery platforms, as applied to the nervous system, including the transfection vectors and carriers used, as well as matrices and scaffolds that are currently used. Additionally, we will discuss possible improvements for localised nucleic acid delivery.
Collapse
Affiliation(s)
- Na Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Jiah Shin Chin
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; NTU Institute of Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, 639798, Singapore
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore.
| |
Collapse
|
9
|
Fontana G, Delgado LM, Cigognini D. Biologically Inspired Materials in Tissue Engineering. EXTRACELLULAR MATRIX FOR TISSUE ENGINEERING AND BIOMATERIALS 2018. [DOI: 10.1007/978-3-319-77023-9_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
10
|
Gene-activated matrix/bone marrow-derived mesenchymal stem cells constructs regenerate sweat glands-like structure in vivo. Sci Rep 2017; 7:17630. [PMID: 29247230 PMCID: PMC5732266 DOI: 10.1038/s41598-017-17967-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/04/2017] [Indexed: 11/08/2022] Open
Abstract
It is a significant challenge to regenerate full-thickness skin defects with sweat glands. Various skin substitutes have been developed to resolve this issue with minimal success. In this study, to yield a novel construct for in situ regeneration of sweat glands, the collagen-chitosan porous scaffold was combined with Lipofectamine 2000/pDNA-EGF complexes to obtain the gene-activated scaffold (GAS), which was then seeded with bone marrow-derived mesenchymal stem cells (BM-MSCs). The porous scaffold functionalized as a reservoir for the incorporated gene complexes which were released in a sustained manner. The seeded BM-MSCs were transfected in situ by the released complexes and specially differentiated into sweat gland cells in vitro under the induction of the expressed epidermal growth factor (EGF). Application in vivo of the GAS/BM-MSCs constructs on the full-thickness skin defects of SD rats confirmed that GAS/BM-MSCs could accelerate the wound healing process and induce the in situ regeneration of the full-thickness skin with sweat gland-like structures. Analyzed by immunohistochemical staining, RT-qPCR and Western-blotting, the levels of the major sweat gland markers such as carcino-embryonic antigen (CEA), cytokeratin 8 (CK8) and cytokeratin 14 (CK14) were all up-regulated, indicating that GAS/BM-MSCs can facilitate the regeneration of sweat glands-like structure in vivo.
Collapse
|
11
|
Effect of sustained PDGF nonviral gene delivery on repair of tooth-supporting bone defects. Gene Ther 2016; 24:31-39. [PMID: 27824330 PMCID: PMC5269540 DOI: 10.1038/gt.2016.73] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/13/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022]
Abstract
Recombinant human platelet-derived growth factor-BB (rhPDGF-BB) promotes soft tissue and bone healing, and is Food and Drug Administration-approved for treatment of diabetic ulcers and periodontal defects. The short half-life of topical rhPDGF-BB protein application necessitates bolus, high-dose delivery. Gene therapy enables sustained local growth factor production. A novel gene activated matrix delivering polyplexes of polyethylenimine (PEI)-plasmid DNA encoding PDGF was evaluated for promotion of periodontal wound repair in vivo. PEI-pPDGF-B polyplexes were tested in human periodontal ligament fibroblasts and human gingival fibroblasts for cell viability and transfection efficiency. Collagen scaffolds containing PEI-pPDGF-B polyplexes at two doses, rhPDGF-BB, PEI vector or collagen alone were randomly delivered to experimentally induced tooth-supporting periodontal defects in a rodent model. Mandibulae were collected at 21 days for histologic observation and histomorphometry. PEI-pPDGF-B polyplexes were biocompatible to cells tested and enzyme-linked immunosorbent assay confirmed the functionality of transfection. Significantly greater osteogenesis was observed for collagen alone and rhPDGF-BB versus the PEI-containing groups. Defects treated with sustained PDGF gene delivery demonstrated delayed healing coupled with sustained inflammatory cell infiltrates lateral to the osseous defects. Continuous PDGF-BB production by nonviral gene therapy could have delayed bone healing. This nonviral gene delivery system in this model appeared to prolong inflammatory response, slowing alveolar bone regeneration in vivo.
Collapse
|
12
|
Das S, Baker AB. Biomaterials and Nanotherapeutics for Enhancing Skin Wound Healing. Front Bioeng Biotechnol 2016; 4:82. [PMID: 27843895 PMCID: PMC5087310 DOI: 10.3389/fbioe.2016.00082] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023] Open
Abstract
Wound healing is an intricate process that requires complex coordination between many cell types and an appropriate extracellular microenvironment. Chronic wounds often suffer from high protease activity, persistent infection, excess inflammation, and hypoxia. While there has been intense investigation to find new methods to improve cutaneous wound care, the management of chronic wounds, burns, and skin wound infection remain challenging clinical problems. Ideally, advanced wound dressings can provide enhanced healing and bridge the gaps in the healing processes that prevent chronic wounds from healing. These technologies have great potential for improving outcomes in patients with poorly healing wounds but face significant barriers in addressing the heterogeneity and clinical complexity of chronic or severe wounds. Active wound dressings aim to enhance the natural healing process and work to counter many aspects that plague poorly healing wounds, including excessive inflammation, ischemia, scarring, and wound infection. This review paper discusses recent advances in the development of biomaterials and nanoparticle therapeutics to enhance wound healing. In particular, this review focuses on the novel cutaneous wound treatments that have undergone significant preclinical development or are currently used in clinical practice.
Collapse
Affiliation(s)
- Subhamoy Das
- Department of Biomedical Engineering, University of Texas at Austin , Austin, TX , USA
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
13
|
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: 54] [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
|
14
|
Urello MA, Kiick KL, Sullivan MO. Integration of growth factor gene delivery with collagen-triggered wound repair cascades using collagen-mimetic peptides. Bioeng Transl Med 2016; 1:207-219. [PMID: 27981245 PMCID: PMC5125401 DOI: 10.1002/btm2.10037] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022] Open
Abstract
Growth factors (GFs) play vital roles in wound repair. Many GF therapies have reached clinical trials, but success has been hindered by safety concerns and a lack of efficacy. Previously, we presented an approach to produce protein factors in wound beds through localized gene delivery mediated by biomimetic peptides. Modification of polyethylenimine (PEI) DNA polyplexes with collagen-mimetic peptides (CMPs) enabled tailoring of polyplex release/retention and improved gene transfer activity in a cell-responsive manner. In this work, CMP-mediated delivery from collagen was shown to improve expression of platelet-derived growth factor-BB (PDGF-BB) and promote a diverse range of cellular processes associated with wound healing, including proliferation, extracellular matrix production, and chemotaxis. Collagens were pre-exposed to physiologically-simulating conditions (complete media, 37°C) for days to weeks prior to cell seeding to simulate the environment within typical wound dressings. In cell proliferation studies, significant increases in cell counts were demonstrated in collagen gels containing CMP-modified polyplex versus unmodified polyplex, and these effects became most pronounced following prolonged preincubation periods of greater than a week. Collagen containing CMP-modified polyplexes also induced a twofold increase in gel contraction as well as enhanced directionality and migratory activity in response to cell-secreted PDGF-BB gradients. While these PDGF-BB-triggered behaviors were observed in collagens containing unmodified polyplexes, the responses withstood much longer preincubation periods in CMP-modified polyplex samples (10 days vs. <5 days). Furthermore, enhanced closure rates in an in vitro wound model suggested that CMP-based PDGF-BB delivery may have utility in actual wound repair and other regenerative medicine applications.
Collapse
Affiliation(s)
- Morgan A. Urello
- Dept. of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDE19716
| | - Kristi L. Kiick
- Dept. of Material Science and EngineeringUniversity of DelawareNewarkDE19716
| | | |
Collapse
|
15
|
Morgado PI, Aguiar-Ricardo A, Correia IJ. Asymmetric membranes as ideal wound dressings: An overview on production methods, structure, properties and performance relationship. J Memb Sci 2015. [DOI: 10.1016/j.memsci.2015.04.064] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
16
|
Barker JC, Barker AD, Bills J, Huang J, Wight-Carter M, Delgado I, Noble DL, Huang LJ, Porteus MH, Davis KE. Genome Editing of Mouse Fibroblasts by Homologous Recombination for Sustained Secretion of PDGF-B and Augmentation of Wound Healing. Plast Reconstr Surg 2014; 134:389e-401e. [DOI: 10.1097/prs.0000000000000427] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
17
|
Jang JH, Houchin TL, Shea LD. Gene delivery from polymer scaffolds for tissue engineering. Expert Rev Med Devices 2014; 1:127-38. [PMID: 16293016 DOI: 10.1586/17434440.1.1.127] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The combination of gene therapy with tissue engineering offers the potential to direct progenitor cell proliferation and differentiation into functional tissue replacements. Many approaches to engineering tissue replacements feature a polymer scaffold to create and maintain a space, support cell adhesion, and organize tissue formation. Polymer scaffolds, either natural, synthetic, or a combination of the two, have also been adapted to serve as delivery vehicles for viral and nonviral vectors, which can induce the expression of tissue inductive factors. Gene delivery is a versatile approach, capable of targeting any cellular process through localized expression of tissue inductive factors. The design and application of tissue engineering scaffolds for localized gene transfer are reviewed. Scaffolds are designed either to release the vector into the local tissue environment or maintain the vector at the polymer surface, which is regulated by the effective affinity of the vector for the polymer. Polymeric delivery can enhance gene transfer locally, promote and extend transgene expression, avoid vector distribution to distant tissues, and reduce the immune response to the vector. Scaffolds capable of controlled DNA delivery can provide a fundamental tool for directing progenitor cell function, which has applications with the engineering of numerous types of tissue. The utility of this approach will increase with the development of design parameters that correlate release and transgene expression, and with continued research into the biology of tissue formation.
Collapse
Affiliation(s)
- Jae-Hyung Jang
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Rd E156 Evanston, IL 60208-3120, USA
| | | | | |
Collapse
|
18
|
|
19
|
Horch RE, Kneser U, Polykandriotis E, Schmidt VJ, Sun J, Arkudas A. Tissue engineering and regenerative medicine -where do we stand? J Cell Mol Med 2012; 16:1157-65. [PMID: 22436120 PMCID: PMC3823070 DOI: 10.1111/j.1582-4934.2012.01564.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Tissue Engineering (TE) in the context of Regenerative Medicine (RM) has been hailed for many years as one of the most important topics in medicine in the twenty-first century. While the first clinically relevant TE efforts were mainly concerned with the generation of bioengineered skin substitutes, subsequently TE applications have been continuously extended to a wide variety of tissues and organs. The advent of either embryonic or mesenchymal adult stem-cell technology has fostered many of the efforts to combine this promising tool with TE approaches and has merged the field into the term Regenerative Medicine. As a typical example in translational medicine, the discovery of a new type of cells called Telocytes that have been described in many organs and have been detected by electron microscopy opens another gate to RM. Besides cell-therapy strategies, the application of gene therapy combined with TE has been investigated to generate tissues and organs. The vascularization of constructs plays a crucial role besides the matrix and cell substitutes. Therefore, novel in vivo models of vascularization have evolved allowing axial vascularization with subsequent transplantation of constructs. This article is intended to give an overview over some of the most recent developments and possible applications in RM through the perspective of TE achievements and cellular research. The synthesis of TE with innovative methods of molecular biology and stem-cell technology appears to be very promising.
Collapse
Affiliation(s)
- Raymund E Horch
- Department of Plastic and Hand Surgery And Laboratory for Tissue Engineering and Regenerative Medicine, Friedrich Alexander University Erlangen-Nuernberg, Erlangen, Germany.
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
INTRODUCTION Tissue defects, sustained through disease or trauma, present enormous challenges in regenerative medicine. Modern tissue engineering (TE) aims at replacing or repairing these defects through a combined approach of biodegradable scaffolds, suitable cell sources and appropriate environmental cues, such as biomolecules presented on scaffold surfaces or sustainably released from within. AREAS COVERED This review provides a brief overview of the various drugs and bioactive molecules of interest to TE, as well as a selection of materials that have been proposed for TE scaffolds and matrices in the past. It then proceeds to discuss encapsulation, immobilization and controlled release strategies for bioactive proteins, before discussing recent advances in this area with a special focus on soft TE. EXPERT OPINION Overall, minimal clinical success has been achieved so far in using growth factor, morphogen, or adhesion factor modified scaffolds and matrices; only one growth factor delivery system (Regranex Gel), has been approved by the FDA for clinical use, with only a handful of other growth factors being approved for human use so far. However, many more growth factors are currently in clinical Phase I - II or preclinical trials and many delivery systems utilize materials already approved by the FDA for other purposes. With respect to drug delivery in soft TE, a combination of increased research efforts in hydrogel and support material development as well as growth factor development is needed before clinical success is realized.
Collapse
Affiliation(s)
- Katharina Ladewig
- The University of Melbourne, Department of Chemical and Biomolecular Engineering , Parkville VIC, Australia.
| |
Collapse
|
21
|
Mahdipour E, Mace KA. Hox transcription factor regulation of adult bone-marrow-derived cell behaviour during tissue repair and regeneration. Expert Opin Biol Ther 2011; 11:1079-90. [PMID: 21513461 DOI: 10.1517/14712598.2011.579096] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Bone marrow offers a valuable source of stem/progenitor cells that contribute to the repair of injured tissues. Failure in the function of these cells results in delayed or reduced tissue repair. Identification of factors that can correct these defects is critical to treating the underlying dysfunction. Notably, homeobox (Hox) transcription factors have been identified as having significant effects on BMDC behaviour, including differentiation, migration and adhesion in injured tissue, and may provide a basis for future therapies. AREAS COVERED Hox protein regulation of bone-marrow-derived cell (BMDC) differentiation, factors that influence BMDC behaviour in response to injury, the effects of the diabetic environment on BMDCs, methods that can be used to reprogramme BMDCs, and the use of Hox transcription factors to correct BMDC behaviour. EXPERT OPINION Hox gene therapy has been successfully employed to change cell behaviour using ex vivo 'reprogramming' strategies overexpressing selected Hox genes in BMDCs to direct the fate of these cells to the desired cell type, promoting tissue repair.
Collapse
Affiliation(s)
- Elahe Mahdipour
- University of Manchester, Healing Foundation Centre, Faculty of Life Sciences, Oxford Road, Manchester M13 9PT, UK
| | | |
Collapse
|
22
|
Priya T, Sabu M, Jolly C. Role ofMangifera indicabark polyphenols on rat gastric mucosa against ethanol and cold-restraint stress. Nat Prod Res 2011; 25:815-26. [DOI: 10.1080/14786410903003822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
23
|
|
24
|
Lei Y, Rahim M, Ng Q, Segura T. Hyaluronic acid and fibrin hydrogels with concentrated DNA/PEI polyplexes for local gene delivery. J Control Release 2011; 153:255-61. [PMID: 21295089 DOI: 10.1016/j.jconrel.2011.01.028] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 01/10/2011] [Accepted: 01/25/2011] [Indexed: 01/08/2023]
Abstract
Local delivery of DNA through a hydrogel scaffold would increase the applicability of gene therapy in tissue regeneration and cancer therapy. However, the delivery of DNA/cationic polymer nanoparticles (polyplexes) using hydrogels is challenging due to the aggregation and inactivation of polyplexes during their incorporation into hydrogel scaffolds. We developed a novel process (termed caged nanoparticle encapsulation or CnE) to load concentrated and unaggregated non-viral gene delivery nanoparticles into various hydrogels. Previously, we showed that PEG hydrogels loaded with DNA/PEI polyplexes through this process were able to deliver genes both in vitro and in vivo. In this study, we found that hyaluronic acid and fibrin hydrogels with concentrated and unaggregated polyplexes loaded through CnE were able to deliver genes in vivo as well, demonstrating the universality of the process.
Collapse
Affiliation(s)
- Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, 420 Westwood Plaza, BH5531, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
25
|
Waddington SN, Crossley R, Sheard V, Howe SJ, Buckley SMK, Coughlan L, Gilham DE, Hawkins RE, McKay TR. Gene delivery of a mutant TGFβ3 reduces markers of scar tissue formation after cutaneous wounding. Mol Ther 2010; 18:2104-11. [PMID: 20736928 DOI: 10.1038/mt.2010.174] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The transforming growth factor-β (TGFβ) family plays a critical regulatory role in repair and coordination of remodeling after cutaneous wounding. TGFβ1-mediated chemotaxis promotes the recruitment of fibroblasts to the wound site and their resultant myofibroblastic transdifferentiation that is responsible for elastic fiber deposition and wound closure. TGFβ3 has been implicated in an antagonistic role regulating overt wound closure and promoting ordered dermal remodeling. We generated a mutant form of TGFβ3 (mutTGFβ3) by ablating its binding site for the latency-associated TGFβ binding protein (LTBP-1) in order to improve bioavailability and activity. The mutated cytokine is secreted as the stable latency-associated peptide (LAP)-associated form and is activated by normal intracellular and extracellular mechanisms including integrin-mediated activation but is not sequestered. We show localized intradermal transduction using a lentiviral vector expressing the mutTGFβ3 in a mouse skin wounding model reduced re-epithelialization density and fibroblast/myofibroblast transdifferentiation within the wound area, both indicative of reduced scar tissue formation.
Collapse
|
26
|
Guo R, Xu S, Ma L, Huang A, Gao C. Enhanced angiogenesis of gene-activated dermal equivalent for treatment of full thickness incisional wounds in a porcine model. Biomaterials 2010; 31:7308-20. [PMID: 20598366 DOI: 10.1016/j.biomaterials.2010.06.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 06/04/2010] [Indexed: 11/26/2022]
Abstract
Angiogenesis of dermal equivalent is one of the key issues for treatment of full thickness skin defects. To develop a gene-activated bilayer dermal equivalent (BDE), N,N,N-trimethyl chitosan chloride (TMC), a cationic gene delivery vector, was used to form complexes with the plasmid DNA encoding vascular endothelial growth factor-165 (VEGF-165), which was then incorporated into a collagen-chitosan/silicone membrane scaffold. To evaluate the angiogenesis property in vivo, full thickness skin defects were made on the back of pigs, into which the TMC/pDNA-VEGF complexes loaded BDE and other three control BDEs, i.e. the blank BDE, and the BDEs loaded with pDNA-VEGF and TMC/pDNA-eGFP complexes, respectively, were transplanted. Biopsy specimens were harvested at day 7, 10 and 14 after surgery for histology, immunohistochemistry, immunofluorescence, real-time quantitative PCR (RT-qPCR) and western blotting analyses. The results showed that the TMC/pDNA-VEGF group had the strongest VEGF expression in mRNA and protein levels, resulting in the highest densities of newly-formed and mature vessels. The ultra-thin skin graft was further transplanted onto the dermis regenerated by the TMC/pDNA-VEGF complexes loaded BDE at day 10 and well survived. At 112 days grafting, the healing skin had a similar structure and approximately 80% tensile strength of the normal skin.
Collapse
Affiliation(s)
- Rui Guo
- Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | | | | | | | | |
Collapse
|
27
|
Sun W, Lin H, Chen B, Zhao W, Zhao Y, Xiao Z, Dai J. Collagen scaffolds loaded with collagen-binding NGF-beta accelerate ulcer healing. J Biomed Mater Res A 2010; 92:887-95. [PMID: 19283824 DOI: 10.1002/jbm.a.32445] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Studies have shown that exogenous nerve growth factor (NGF) accelerates ulcer healing, but the inefficient growth factor delivery system limits its clinical application. In this report, we found that the native human NGF-beta fused with a collagen-binding domain (CBD) could form a collagen-based NGF targeting delivery system, and the CBD-fused NGF-beta could bind to collagen membranes efficiently. Using the rabbit dermal ischemic ulcer model, we have found that this targeting delivery system maintains a higher concentration and stronger bioactivity of NGF-beta on the collagen membranes by promoting peripheral nerve growth. Furthermore, it enhances the rate of ulcer healing through accelerating the re-epithelialization of dermal ulcer wounds and the formation of capillary lumens within the newly formed tissue area. Thus, collagen membranes loaded with collagen-targeting human NGF-beta accelerate ulcer healing efficiently.
Collapse
Affiliation(s)
- Wenjie Sun
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Barbato JE, Kibbe MR, Tzeng E. The Emerging Role of Gene Therapy in the Treatment of Cardiovascular Diseases. Crit Rev Clin Lab Sci 2010. [DOI: 10.1080/10408360390250621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
Ono I, Yamashita T, Hida T, Jin HY, Ito Y, Hamada H, Akasaka Y, Ishii T, Jimbow K. Combined administration of basic fibroblast growth factor protein and the hepatocyte growth factor gene enhances the regeneration of dermis in acute incisional wounds. Wound Repair Regen 2009. [DOI: 10.1111/j.1067-1927.2004.012113.x-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
30
|
Mogford JE, Tawil B, Jia S, Mustoe TA. Fibrin sealant combined with fibroblasts and platelet-derived growth factor enhance wound healing in excisional wounds. Wound Repair Regen 2009; 17:405-10. [DOI: 10.1111/j.1524-475x.2009.00481.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Giordano C, Causa F, Bianco F, Perale G, Netti PA, Ambrosio L, Cigada A. Gene delivery systems for gene therapy in tissue engineering and central nervous system applications. Int J Artif Organs 2009; 31:1017-26. [PMID: 19115193 DOI: 10.1177/039139880803101205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The present review aims to describe the potential applications of gene delivery systems to tissue engineering and central nervous system diseases. Some key experimental work has been done with interesting results, but the subject is far from being fully explored. The combined approach of gene therapy and material science has a huge potential to improve the therapeutic approaches now available for a wide range of medical applications. Focus is given to this multidisciplinary strategy in neurodegenerative pathologies, where the use of polymeric matrices as gene carriers might make a crucial difference.
Collapse
Affiliation(s)
- C Giordano
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy.
| | | | | | | | | | | | | |
Collapse
|
32
|
Evaluation of electrical stimulation for ischemic wound therapy: a feasibility study using the lapine wound model. Arch Dermatol Res 2008; 301:323-7. [DOI: 10.1007/s00403-008-0918-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 11/10/2008] [Indexed: 10/21/2022]
|
33
|
Lin Z, Sugai JV, Jin Q, Chandler LA, Giannobile WV. Platelet-derived growth factor-B gene delivery sustains gingival fibroblast signal transduction. J Periodontal Res 2008; 43:440-9. [PMID: 18823454 DOI: 10.1111/j.1600-0765.2008.01089.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Platelet-derived growth factor-BB is a potent mediator of tooth-supporting periodontal tissue repair and regeneration. A limitation of the effects of topical platelet-derived growth factor-BB application is its short half-life in vivo. Gene therapy has shown strong promise for the long-term delivery of platelet-derived growth factor in both skin ulcer healing and periodontal tissue engineering. However, little is known regarding the extended effects of platelet-derived growth factor-B on cell signaling via gene delivery, especially at the level of phosphorylation of intracellular kinases. This study sought to evaluate the effect of gene transfer by Ad-PDGF-B on human gingival fibroblasts (HGFs) and the subsequent regulation of genes and cell-surface proteins associated with cellular signaling. MATERIAL AND METHODS HGFs from human subjects were treated by adenoviral PDGF-B, PDGF-1308 (a dominant negative mutant of PDGF) and recombinant human platelet-derived growth factor-BB, and then incubated in serum-free conditions for various time points and harvested at 1, 6, 12, 24, 48, 72 and 96 h. Exogenous PDGF-B was measured by RT-PCR and Western blot. Cell proliferation was evaluated by [methyl-3H]thymidine incorporation assay. We used proteomic arrays to explore phosphorylation patterns of 23 different intracellular kinases after PDGF-B gene transfer. The expression of alpha and beta PDGFR and Akt were measured by Western blot analysis. RESULTS Sustained in vitro expression of PDGF-B in HGFs by Ad-PDGF-B transduction was seen at both the mRNA and protein levels. Compared to rhPDGF-BB and Ad-PDGF-1308, Ad-PDGF-B maintained cell growth in serum-free conditions, with robust increases in DNA synthesis. Gene delivery of PDGF-B also prolonged downregulation of the growth arrest specific gene (gas) PDGF alpha R. Of the 23 intracellular kinases that we tested in proteomic arrays, Akt revealed the most notable long-term cell signaling effect as a result of the over-expression of Ad-PDGF-B, compared with pulse recombinant human platelet-derived growth factor BB. Prolonged Akt phosphorylation was induced by treatment with Ad-PDGF-B, for at least up to 96 h. CONCLUSION These findings further demonstrate that gene delivery of PDGF-B displays sustained signal transduction effects in human gingival fibroblasts that are higher than those conveyed by treatment with recombinant human platelet-derived growth factor-BB protein. These data on platelet-derived growth factor gene delivery contribute to an improved understanding of these pathways that are likely to play a role in the control of clinical outcomes of periodontal regenerative therapy.
Collapse
Affiliation(s)
- Z Lin
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | |
Collapse
|
34
|
Biondi M, Ungaro F, Quaglia F, Netti PA. Controlled drug delivery in tissue engineering. Adv Drug Deliv Rev 2008; 60:229-42. [PMID: 18031864 DOI: 10.1016/j.addr.2007.08.038] [Citation(s) in RCA: 280] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 08/09/2007] [Indexed: 11/15/2022]
Abstract
The concept of tissue and cell guidance is rapidly evolving as more information regarding the effect of the microenvironment on cellular function and tissue morphogenesis become available. These disclosures have lead to a tremendous advancement in the design of a new generation of multifunctional biomaterials able to mimic the molecular regulatory characteristics and the three-dimensional architecture of the native extracellular matrix. Micro- and nano-structured scaffolds able to sequester and deliver in a highly specific manner biomolecular moieties have already been proved to be effective in bone repairing, in guiding functional angiogenesis and in controlling stem cell differentiation. Although these platforms represent a first attempt to mimic the complex temporal and spatial microenvironment presented in vivo, an increased symbiosis of material engineering, drug delivery technology and cell and molecular biology may ultimately lead to biomaterials that encode the necessary signals to guide and control developmental process in tissue- and organ-specific differentiation and morphogenesis.
Collapse
Affiliation(s)
- Marco Biondi
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | | | | | | |
Collapse
|
35
|
Sun W, Lin H, Xie H, Chen B, Zhao W, Han Q, Zhao Y, Xiao Z, Dai J. Collagen membranes loaded with collagen-binding human PDGF-BB accelerate wound healing in a rabbit dermal ischemic ulcer model. Growth Factors 2007; 25:309-18. [PMID: 18236209 DOI: 10.1080/08977190701803885] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Studies have shown that exogenous platelet-derived growth factor-BB (PDGF-BB) could accelerate the ulcer healing, but the lack of efficient growth factor delivery system limits its clinical application. Our previous work has demonstrated that the native human PDGF-BB was added a collagen-binding domain (CBD), TKKTLRT, to develop a collagen-based PDGF targeting delivery system. Here, we showed that this CBD-fused PDGF-BB (CBD-PDGF) could bind to collagen membrane efficiently. We used the rabbit dermal ischemic ulcer model to study the effects of CBD-PDGF loaded on collagen membranes. Results revealed that this system maintained a higher concentration and stronger bioactivity of PDGF-BB on the collagen membranes and promoted the re-epithelialization of dermal ulcer wounds, the collagen deposition, and the formation of capillary lumens within the newly formed tissue area. It demonstrated that collagen membranes loaded with collagen-targeting human PDGF-BB could effectively promote ulcer healing.
Collapse
Affiliation(s)
- Wenjie Sun
- Key laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Aiming for regeneration of severed or lost parts of the body, the combined application of gene therapy and tissue engineering has received much attention by regenerative medicine. Techniques of molecular biology can enhance the regenerative potential of a biomaterial by co-delivery of therapeutic genes, and several different strategies have been used to achieve that goal. Possibilities for application are many-fold and have been investigated to regenerate tissues such as skin, cartilage, bone, nerve, liver, pancreas and blood vessels. This review discusses advantages and problems encountered with the different gene delivery strategies as far as they relate to tissue engineering, analyses the positive aspects of polymeric gene delivery from matrices and discusses advances and future challenges of gene transfer strategies in selected tissues.
Collapse
Affiliation(s)
- Oliver Bleiziffer
- Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Erlangen, Germany
- *Correspondence to: Ulrich KNESER, M.D. Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Krankenhausstr. 12, 91054 Erlangen, Germany. Tel.: +49-9131-85-33277; Fax: +49-9131-85-39327 E-mail:
| | - Elof Eriksson
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Feng Yao
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Erlangen, Germany
| | - Ulrich Kneser
- Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Erlangen, Germany
- *Correspondence to: Ulrich KNESER, M.D. Department of Plastic and Hand Surgery, University of Erlangen Medical Center, Krankenhausstr. 12, 91054 Erlangen, Germany. Tel.: +49-9131-85-33277; Fax: +49-9131-85-39327 E-mail:
| |
Collapse
|
37
|
De Laporte L, Shea LD. Matrices and scaffolds for DNA delivery in tissue engineering. Adv Drug Deliv Rev 2007; 59:292-307. [PMID: 17512630 PMCID: PMC1949490 DOI: 10.1016/j.addr.2007.03.017] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2006] [Accepted: 03/28/2007] [Indexed: 12/13/2022]
Abstract
Regenerative medicine aims to create functional tissue replacements, typically through creating a controlled environment that promotes and directs the differentiation of stem or progenitor cells, either endogenous or transplanted. Scaffolds serve a central role in many strategies by providing the means to control the local environment. Gene delivery from the scaffold represents a versatile approach to manipulating the local environment for directing cell function. Research at the interface of biomaterials, gene therapy, and drug delivery has identified several design parameters for the vector and the biomaterial scaffold that must be satisfied. Progress has been made towards achieving gene delivery within a tissue engineering scaffold, though the design principles for the materials and vectors that produce efficient delivery require further development. Nevertheless, these advances in obtaining transgene expression with the scaffold have created opportunities to develop greater control of either delivery or expression and to identify the best practices for promoting tissue formation. Strategies to achieve controlled, localized expression within the tissue engineering scaffold will have broad application to the regeneration of many tissues, with great promise for clinical therapies.
Collapse
Affiliation(s)
- Laura De Laporte
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208
| | - Lonnie D. Shea
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208
- The Robert H. Lurie Comprehensive Cancer Center of Northwestern University Chicago, IL 60611
| |
Collapse
|
38
|
Andreadis ST. Gene-modified tissue-engineered skin: the next generation of skin substitutes. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2006; 103:241-74. [PMID: 17195466 DOI: 10.1007/10_023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tissue engineering combines the principles of cell biology, engineering and materials science to develop three-dimensional tissues to replace or restore tissue function. Tissue engineered skin is one of most advanced tissue constructs, yet it lacks several important functions including those provided by hair follicles, sebaceous glands, sweat glands and dendritic cells. Although the complexity of skin may be difficult to recapitulate entirely, new or improved functions can be provided by genetic modification of the cells that make up the tissues. Gene therapy can also be used in wound healing to promote tissue regeneration or prevent healing abnormalities such as formation of scars and keloids. Finally, gene-enhanced skin substitutes have great potential as cell-based devices to deliver therapeutics locally or systemically. Although significant progress has been made in the development of gene transfer technologies, several challenges have to be met before clinical application of genetically modified skin tissue. Engineering challenges include methods for improved efficiency and targeted gene delivery; efficient gene transfer to the stem cells that constantly regenerate the dynamic epidermal tissue; and development of novel biomaterials for controlled gene delivery. In addition, advances in regulatable vectors to achieve spatially and temporally controlled gene expression by physiological or exogenous signals may facilitate pharmacological administration of therapeutics through genetically engineered skin. Gene modified skin substitutes are also employed as biological models to understand tissue development or disease progression in a realistic three-dimensional context. In summary, gene therapy has the potential to generate the next generation of skin substitutes with enhanced capacity for treatment of burns, chronic wounds and even systemic diseases.
Collapse
Affiliation(s)
- Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical & Biological Engineering, University at Buffalo, The State University of New York (SUNY), Amherst, NY 14260, USA.
| |
Collapse
|
39
|
Abstract
Modern molecular and genetic technologies enable the modification of a cellular genome through transfer of specific genes. The various procedures alter specific cell functions, which allow the transfected cell to produce any encoded transgene information. The transfected cell then synthesizes proteins that are normally not produced or only in very small amounts. Numerous animal studies have demonstrated that gene therapy may support and accelerate the healing and regeneration of specific tissues such as skin, tendons, cartilage, and bones. Currently, further animal studies are evaluating new vectors with reduced immunogenicity in the continuous effort to improve the efficacy and safety of gene transfer. In the forthcoming decade we expect gene therapy to have an important influence on the treatment of fractures, cartilage lesions, and infection.
Collapse
Affiliation(s)
- A Oberholzer
- Zentrum für Spezielle Chirurgie des Bewegungsapparates, Klinik für Unfall- und Wiederherstellungschirurgie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin.
| | | | | | | |
Collapse
|
40
|
Andreadis ST, Geer DJ. Biomimetic approaches to protein and gene delivery for tissue regeneration. Trends Biotechnol 2006; 24:331-7. [PMID: 16716420 DOI: 10.1016/j.tibtech.2006.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Revised: 03/07/2006] [Accepted: 05/09/2006] [Indexed: 12/31/2022]
Abstract
Novel therapeutic strategies that promote wound healing seek to mimic the response of the body to wounding, to regenerate rather than repair injured tissues. Many synthetic or natural biomaterials have been developed for this purpose and are used to deliver wound therapeutics in a controlled manner that prevents unwanted and potentially harmful side-effects. Here, we review the natural and synthetic biomaterials that have been developed for protein and gene delivery to enhance tissue regeneration. Particular emphasis is placed on novel biomimetic materials that respond to environmental stimuli or release their cargo according to cellular demand. Engineering biomaterials to release therapeutic agents in response to physiologic signals mimics the natural healing process and can promote faster tissue regeneration and reduce scarring in severe acute or chronic wounds.
Collapse
Affiliation(s)
- Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260, USA.
| | | |
Collapse
|
41
|
Salvay DM, Shea LD. Inductive tissue engineering with protein and DNA-releasing scaffolds. MOLECULAR BIOSYSTEMS 2005; 2:36-48. [PMID: 16880921 PMCID: PMC2657198 DOI: 10.1039/b514174p] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cellular differentiation, organization, proliferation and apoptosis are determined by a combination of an intrinsic genetic program, matrix/substrate interactions, and extracellular cues received from the local microenvironment. These molecular cues come in the form of soluble (e.g. cytokines) and insoluble (e.g. ECM proteins) factors, as well as signals from surrounding cells that can promote specific cellular processes leading to tissue formation or regeneration. Recent developments in the field of tissue engineering have employed biomaterials to present these cues, providing powerful tools to investigate the cellular processes involved in tissue development, or to devise therapeutic strategies based on cell replacement or tissue regeneration. These inductive scaffolds utilize natural and/or synthetic biomaterials fabricated into three-dimensional structures. This review summarizes the use of scaffolds in the dual role of structural support for cell growth and vehicle for controlled release of tissue inductive factors, or DNA encoding for these factors. The confluence of molecular and cell biology, materials science and engineering provides the tools to create controllable microenvironments that mimic natural developmental processes and direct tissue formation for experimental and therapeutic applications.
Collapse
Affiliation(s)
- David M. Salvay
- Department of Chemical and Biological Engineering, 2145 Sheridan Rd E156 Evanston, IL 60208-3120. E-mail: ; Fax: 847-491-3728; Tel: 847-491-7043
| | - Lonnie D. Shea
- Department of Chemical and Biological Engineering, 2145 Sheridan Rd E156 Evanston, IL 60208-3120. E-mail: ; Fax: 847-491-3728; Tel: 847-491-7043
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Rd E156 Evanston, IL 60208-3120
| |
Collapse
|
42
|
Jang JH, Rives CB, Shea LD. Plasmid delivery in vivo from porous tissue-engineering scaffolds: transgene expression and cellular transfection. Mol Ther 2005; 12:475-83. [PMID: 15950542 PMCID: PMC2648405 DOI: 10.1016/j.ymthe.2005.03.036] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Revised: 03/22/2005] [Accepted: 03/24/2005] [Indexed: 01/27/2023] Open
Abstract
Tissue engineering scaffolds capable of sustained plasmid release can promote gene transfer locally and stimulate new tissue formation. We have investigated the scaffold design parameters that influence the extent and duration of transgene expression and have characterized the distribution of transfected cells. Porous scaffolds with encapsulated plasmid were fabricated from poly(lactide-co-glycolide) with a gas foaming procedure, with wet granulation employed to mix the components homogeneously prior to foaming. Wet granulation enhanced plasmid incorporation relative to standard procedures and also enhanced in vivo transgene expression, possibly through the increased loading and maintenance of the scaffold pore structure. The plasmid loading regulated the quantity and duration of transgene expression, with expression for 105 days achieved at the highest dosage. Expression was localized to the implantation site, though the distribution of transfected cells varied with time. Transfected cells were initially observed at the scaffold periphery (day 3), then within the pores and adjacent to the polymer (day 17), and finally throughout the scaffold interior (day 126). Delivery of a plasmid encoding VEGF increased the blood vessel density relative to control. Correlating scaffold design with gene transfer efficiency and tissue formation will facilitate application of plasmid-releasing scaffolds to multiple tissues.
Collapse
Affiliation(s)
- Jae-Hyung Jang
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road E156, Evanston, IL 60208-3120, USA
| | - Christopher B. Rives
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road E156, Evanston, IL 60208-3120, USA
| | - Lonnie D. Shea
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road E156, Evanston, IL 60208-3120, USA
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road E156, Evanston, IL 60208-3120, USA
- To whom correspondence and reprint requests should be addressed. Fax: +1 847 491 3728. E-mail:
| |
Collapse
|
43
|
Petrie NC, Vranckx JJ, Hoeller D, Yao F, Eriksson E. Gene delivery of PDGF for wound healing therapy. J Tissue Viability 2005; 15:16-21. [PMID: 16302501 DOI: 10.1016/s0965-206x(05)54002-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nicola C Petrie
- Laboratory of Wound Repair and Gene Transfer, Division of Plastic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | |
Collapse
|
44
|
Abstract
The skin is an attractive target for gene therapy because it is easily accessible and shows great potential as an ectopic site for protein delivery in vivo. Genetically modified epidermal cells can be used to engineer three-dimensional skin substitutes, which when transplanted can act as in vivo 'bioreactors' for delivery of therapeutic proteins locally or systemically. Although some gene transfer technologies have the potential to afford permanent genetic modification, differentiation and eventual loss of genetically modified cells from the epidermis results in temporary transgene expression. Therefore, to achieve stable long-term gene expression, it is critical to deliver genes to epidermal stem cells, which possess unlimited growth potential and self-renewal capacity. This review discusses the recent advances in epidermal stem cell isolation, gene transfer and engineering of skin substitutes. Recent efforts that employ gene therapy and tissue engineering for the treatment of genetic diseases, chronic wounds and systemic disorders, such as leptin deficiency or diabetes, are reviewed. Finally, the use of gene-modified tissue-engineered skin as a biological model for understanding tissue development, wound healing and epithelial carcinogenesis is also discussed.
Collapse
Affiliation(s)
- Stelios T Andreadis
- University at Buffalo, Bioengineering Laboratory, Department of Chemical and Biological Engineering, State University of New York, Amherst, NY 14260, USA.
| |
Collapse
|
45
|
Lee PY, Chesnoy S, Huang L. Electroporatic delivery of TGF-beta1 gene works synergistically with electric therapy to enhance diabetic wound healing in db/db mice. J Invest Dermatol 2004; 123:791-8. [PMID: 15373787 DOI: 10.1111/j.0022-202x.2004.23309.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Electrical stimulation (ES) is a therapeutic treatment for wound healing. Electroporation, a type of ES, is a well-established method for gene delivery. We hypothesize that proper conditions can be found with which both electrical and gene therapies can be additively applied to treat diabetic wound healing. For the studies of transforming growth factor-beta1 (TGF-beta1) local expression and therapeutic effects, full thickness excisional wound model of db/db mice was used, we measured TGF-beta1 cytokine level at 24 h postwounding and examined wounds histologically. Furthermore, wound closure was evaluated by wound-area measurements at each day for 14 d. We found that syringe electrodes are more effective than the conventional caliper electrodes. Furthermore, diabetic skin was more sensitive to the electroporative damage than the normal skin. The optimal condition for diabetic skin was six pulses of 100 V per cm for 20 ms. Under such condition, the healing rate of electrically treated wound was significantly accelerated. Furthermore, when TGF-beta1 gene was delivered by electric pulses, the healing rate was further enhanced. Five to seven days postapplication of intradermal injection of plasmid TGF-beta1 followed by electroporation, the wound bed showed an increased reepithelialization rate, collagen synthesis, and angiogenesis. The data indicates that indeed the electric effect and gene effect work synergistic in the genetically diabetic model.
Collapse
Affiliation(s)
- Pui-Yan Lee
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
46
|
Bellocq NC, Kang DW, Wang X, Jensen GS, Pun SH, Schluep T, Zepeda ML, Davis ME. Synthetic Biocompatible Cyclodextrin-Based Constructs for Local Gene Delivery to Improve Cutaneous Wound Healing. Bioconjug Chem 2004; 15:1201-11. [PMID: 15546185 DOI: 10.1021/bc0498119] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The localized, sustained delivery of growth factors for wound healing therapy is actively being explored by gene transfer to the wound site. Biocompatible matrices such as bovine collagen have demonstrated usefulness in sustaining gene therapy vectors that express growth factors in local sites for tissue repair. Here, new synthetic biocompatible materials are prepared and shown to deliver a protein to cultured cells via the use of an adenoviral delivery vector. The synthetic construct consists of a linear, beta-cyclodextrin-containing polymer and an adamantane-based cross-linking polymer. When the two polymers are combined, they create an extended network by the formation of inclusion complexes between the cyclodextrins and adamantanes. The properties of the network are altered by controlling the polymer molecular weights and the number of adamantanes on the cross-linking polymer, and these modifications and others such as replacement of the beta-cyclodextrin (host) and adamantane (guest) with other cyclodextrins (hosts such as alpha, gamma, and substituted members) and inclusion complex forming molecules (guests) provide the ability to rationally design network characteristics. Fibroblasts exposed to these synthetic constructs show proliferation rates and migration patterns similar to those obtained with collagen. Gene delivery (green fluorescent protein) to fibroblasts via the inclusion of adenoviral vectors in the synthetic construct is equivalent to levels observed with collagen. These in vitro results suggest that the synthetic constructs are suitable for in vivo tissue repair applications.
Collapse
Affiliation(s)
- Nathalie C Bellocq
- Insert Therapeutics, Inc., 2585 Nina Street, Pasadena, California 91107, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Ono I, Yamashita T, Hida T, Jin HY, Ito Y, Hamada H, Akasaka Y, Ishii T, Jimbow K. Local administration of hepatocyte growth factor gene enhances the regeneration of dermis in acute incisional wounds. J Surg Res 2004; 120:47-55. [PMID: 15172189 DOI: 10.1016/j.jss.2003.08.242] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2003] [Indexed: 02/02/2023]
Abstract
Hepatocyte growth factor (HGF) has a number of biological activities, e.g., mitogenic, motogenic, antiapoptotic, antifibrous, and morphogenic. It also has angiogenic and angioprotective activities for endothelial cells. The aim of this study was to characterize the role of HGF in wound healing by administering the HGF gene locally to acute incisional skin wounds created on the backs of rats. To create wounds, the backs of Wistar rats were clipped and three 2-cm-long incisional wounds were made deep to the fascia. The wounds contained pannicrus carnosum and were created at intervals of 2 cm. After suturing, the HGF gene was then administered intradermally. Apoptotic cells in wound lesions were identified by TUNEL method as well as by immunological detection of active caspase-3. In the HGF-treated animals, we found almost complete suppression of apoptosis and well-organized wound healing. Histopathological examination revealed that the proliferation of fibroblasts was suppressed and that scar formation was less apparent in the HGF-treated animals compared to the controls. It is thought that administration of the HGF gene immediately after surgery may enhance the healing process through suppressing apoptosis, which occurred in the controls 1 week after suturing the incisional wound. In addition, locally increased HGF expression due to the introduction of the HGF gene to cells around wounds enhances dermal regeneration, possibly by promoting regeneration of dermal tissue, which results in less scarring due to its antifibrotic effect. Thus, HGF supplementation through gene therapy may be an effective strategy for treating wounds, as it increases the regeneration of the dermis to allow for "scarless wound healing."
Collapse
Affiliation(s)
- Ichiro Ono
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Pannier AK, Shea LD. Controlled release systems for DNA delivery. Mol Ther 2004; 10:19-26. [PMID: 15233938 DOI: 10.1016/j.ymthe.2004.03.020] [Citation(s) in RCA: 190] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2004] [Accepted: 03/24/2004] [Indexed: 12/01/2022] Open
Abstract
Adapting controlled release technologies to the delivery of DNA has the potential to overcome extracellular barriers that limit gene therapy. Controlled release systems can enhance gene delivery and increase the extent and duration of transgene expression relative to more traditional delivery methods (e.g., injection). These systems typically deliver vectors locally, which can avoid distribution to distant tissues, decrease toxicity to nontarget cells, and reduce the immune response to the vector. Delivery vehicles for controlled release are fabricated from natural and synthetic polymers, which function either by releasing the vector into the local tissue environment or by maintaining the vector at the polymer surface. Vector release or binding is regulated by the effective affinity of the vector for the polymer, which depends upon the strength of molecular interactions. These interactions occur through nonspecific binding based on vector and polymer composition or through the incorporation of complementary binding sites (e.g., biotin-avidin). This review examines the delivery of nonviral and viral vectors from natural and synthetic polymers and presents opportunities for continuing developments to increase their applicability.
Collapse
Affiliation(s)
- Angela K Pannier
- Department of Interdepartmental Biological Sciences, Northwestern University, 2145 Sheridan Road, E156, Evanston, IL 60208-3120, USA
| | | |
Collapse
|
49
|
Efron PA, Moldawer LL. Cytokines and wound healing: the role of cytokine and anticytokine therapy in the repair response. ACTA ACUST UNITED AC 2004; 25:149-60. [PMID: 15091141 DOI: 10.1097/01.bcr.0000111766.97335.34] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Wound healing is an integrated and complex process involving a large number of regulatory molecules, including proinflammatory cytokines and growth factors, and an orchestrated tissue response. Dysregulation in cytokine or growth factor expression dramatically alters the normal wound healing process, and blocking the inappropriate production of specific proinflammatory cytokines or supplementing the milieu with increased quantities of growth factors has demonstrated the central role played by these mediators. Both protein-based and DNA-based (gene transfer) therapies are currently under clinical development as tools to improve the healing process. Although there has been some success with these approaches in both experimental models and in patients, only through a better understanding of the complexity and diversity of the wound healing process, as well as an improved comprehension of the time-dependent and concentration-dependent responses to individual proinflammatory cytokines or growth factors, will further development in the therapeutic treatment of healing wounds be attained.
Collapse
Affiliation(s)
- Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, USA
| | | |
Collapse
|
50
|
Anusaksathien O, Webb SA, Jin QM, Giannobile WV. Platelet-derived growth factor gene delivery stimulates ex vivo gingival repair. ACTA ACUST UNITED AC 2004; 9:745-56. [PMID: 13678451 PMCID: PMC2586961 DOI: 10.1089/107632703768247421] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Destruction of tooth support due to the chronic inflammatory disease periodontitis is a major cause of tooth loss. There are limitations with available treatment options to tissue engineer soft tissue periodontal defects. The exogenous application of growth factors (GFs) such as platelet-derived growth factor (PDGF) has shown promise to enhance oral and periodontal tissue regeneration. However, the topical administration of GFs has not led to clinically significant improvements in tissue regeneration because of problems in maintaining therapeutic protein levels at the defect site. The utilization of PDGF gene transfer may circumvent many of the limitations with protein delivery to soft tissue wounds. The objective of this study was to test the effect of PDGF-A and PDGF-B gene transfer to human gingival fibroblasts (HGFs) on ex vivo repair in three-dimensional collagen lattices. HGFs were transduced with adenovirus encoding PDGF-A and PDGF-B genes. Defect fill of bilayer collagen gels was measured by image analysis of cell repopulation into the gingival defects. The modulation of gene expression at the defect site and periphery was measured by RT-PCR during a 10-day time course after gene delivery. The results demonstrated that PDGF-B gene transfer stimulated potent (>4-fold) increases in cell repopulation and defect fill above that of PDGF-A and corresponding controls. PDGF-A and PDGF-B gene expression was maintained for at least 10 days. PDGF gene transfer upregulated the expression of phosphatidylinosital 3-kinase and integrin alpha5 subunit at 5 days after adenovirus transduction. These results suggest that PDGF gene transfer has potential for periodontal soft tissue-engineering applications.
Collapse
Affiliation(s)
- Orasa Anusaksathien
- Center for Craniofacial Regeneration and Department of Periodontics, Prevention, and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
| | | | | | | |
Collapse
|