1
|
Naaz A, Turnquist HR, Gorantla VS, Little SR. Drug delivery strategies for local immunomodulation in transplantation: Bridging the translational gap. Adv Drug Deliv Rev 2024; 213:115429. [PMID: 39142608 DOI: 10.1016/j.addr.2024.115429] [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: 02/15/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Drug delivery strategies for local immunomodulation hold tremendous promise compared to current clinical gold-standard systemic immunosuppression as they could improve the benefit to risk ratio of life-saving or life-enhancing transplants. Such strategies have facilitated prolonged graft survival in animal models at lower drug doses while minimizing off-target effects. Despite the promising outcomes in preclinical animal studies, progression of these strategies to clinical trials has faced challenges. A comprehensive understanding of the translational barriers is a critical first step towards clinical validation of effective immunomodulatory drug delivery protocols proven for safety and tolerability in pre-clinical animal models. This review overviews the current state-of-the-art in local immunomodulatory strategies for transplantation and outlines the key challenges hindering their clinical translation.
Collapse
Affiliation(s)
- Afsana Naaz
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, 15213, United States.
| | - Heth R Turnquist
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States.
| | - Vijay S Gorantla
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States; Departments of Surgery, Ophthalmology and Bioengineering, Wake Forest School of Medicine, Wake Forest Institute of Regenerative Medicine, Winston Salem, NC, 27101, United States.
| | - Steven R Little
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
2
|
Schwager JM, Di Maggio N, Grosso A, Rasadurai A, Minder N, Hubbell JA, Kappos EA, Schaefer DJ, Briquez PS, Banfi A, Burger MG. Semaphorin 3A promotes the long-term persistence of human SVF-derived microvascular networks in engineered grafts. Front Bioeng Biotechnol 2024; 12:1396450. [PMID: 39234267 PMCID: PMC11371724 DOI: 10.3389/fbioe.2024.1396450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction The stromal vascular fraction (SVF) of human adipose tissue is an attractive cell source for engineering grafts with intrinsic vascularization potential, as it is rich in vasculogenic progenitors. However, in order to maintain their functional perfusion it is important to promote the in vivo stabilization of newly assembled microvascular networks. We previously found that Semaphorin 3A (Sema3A) promotes the rapid stabilization of new blood vessels induced by VEGF overexpression in skeletal muscle. Here we investigated whether Sema3A could promote the assembly, connection to circulation and persistence of human SVF-derived microvascular networks in engineered grafts. Methods Recombinant Sema3A was engineered with a transglutaminase substrate sequence (TG-Sema3A) to allow cross-linking into fibrin hydrogels. Grafts were prepared with freshly isolated human SVF cells in fibrin hydrogels decorated with 0, 0.1 or 100 μg/ml TG-Sema3A and implanted subcutaneously in immune-deficient mice. Results After 1 week in vivo, the assembly of human-derived networks was similar in all conditions. The outer part of the grafts was populated by blood vessels of both human and mouse origin, which formed abundant hybrid structures within a common basal lamina. About 90% of human-derived blood vessels were functionally connected to the host circulation in all conditions. However, in the control samples human vessels were unstable. In fact, they significantly regressed by 6 weeks and could no longer be found by 12 weeks. In contrast, a low Sema3A dose (0.1 μg/ml) promoted further human vascular expansion by about 2-fold at 6 weeks and protected them from regression until 12 weeks. From a mechanistic point of view, the stabilization of SVF-derived vessels by 0.1 μg/ml of Sema3A correlated with the recruitment of a specific population of monocytes expressing its receptor Neuropilin-1. Discussion In conclusion, Sema3A is a potent stimulator of in vivo long-term persistence of microvascular networks derived from human SVF. Therefore, decoration of matrices with Sema3a can be envisioned to promote the functional support of tissue engineered grafts.
Collapse
Affiliation(s)
- Juan M Schwager
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Nunzia Di Maggio
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Andrea Grosso
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Abeelan Rasadurai
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Nadja Minder
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Elisabeth A Kappos
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
- Department of Clinical Research, Medical Faculty, University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Priscilla S Briquez
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Andrea Banfi
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Maximilian G Burger
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
3
|
Che Z, Sun Q, Zhao Z, Wu Y, Xing H, Song K, Chen A, Wang B, Cai M. Growth factor-functionalized titanium implants for enhanced bone regeneration: A review. Int J Biol Macromol 2024; 274:133153. [PMID: 38897500 DOI: 10.1016/j.ijbiomac.2024.133153] [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: 02/26/2024] [Revised: 06/02/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
Titanium and titanium alloys are widely favored materials for orthopedic implants due to their exceptional mechanical properties and biological inertness. The additional benefit of sustained local release of bioactive substances further promotes bone tissue formation, thereby augmenting the osseointegration capacity of titanium implants and attracting increasing attention in bone tissue engineering. Among these bioactive substances, growth factors have shown remarkable osteogenic and angiogenic induction capabilities. Consequently, researchers have developed various physical, chemical, and biological loading techniques to incorporate growth factors into titanium implants, ensuring controlled release kinetics. In contrast to conventional treatment modalities, the localized release of growth factors from functionalized titanium implants not only enhances osseointegration but also reduces the risk of complications. This review provides a comprehensive examination of the types and mechanisms of growth factors, along with a detailed exploration of the methodologies used to load growth factors onto the surface of titanium implants. Moreover, it highlights recent advancements in the application of growth factors to the surface of titanium implants (Scheme 1). Finally, the review discusses current limitations and future prospects for growth factor-functionalized titanium implants. In summary, this paper presents cutting-edge design strategies aimed at enhancing the bone regenerative capacity of growth factor-functionalized titanium implants-a significant advancement in the field of enhanced bone regeneration.
Collapse
Affiliation(s)
- Zhenjia Che
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Yanglin Wu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Hu Xing
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Kaihang Song
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Aopan Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| |
Collapse
|
4
|
Morey M, Larrañaga A, Abbah SA, Bohara R, Aljaabary A, Pandit A. Glucose-Responsive Fibrin Hydrogel-Based Multimodal Nucleic Acid Delivery System. Adv Biol (Weinh) 2023; 7:e2300161. [PMID: 37401646 DOI: 10.1002/adbi.202300161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Indexed: 07/05/2023]
Abstract
Nucleic acid therapy has emerged as a potential alternative for promoting wound healing by gene expression modification. On the other hand, protecting the nucleic acid payload from degradation, efficient bioresponsive delivery and effective transfection into cells remain challenging. A glucose-responsive gene delivery system for treating diabetic wounds would be advantageous as it would be responsive to the underlying pathology giving a regulated payload delivery with fewer side effects. Herein a GOx-based glucose-responsive delivery system is designed based on fibrin-coated polymeric microcapsules (FCPMC) using the layer-by-layer (LbL) approach that simultaneously delivers two nucleic acids in diabetic wounds. The designed FCPMC displays an ability to effectively load many nucleic acids in polyplexes and release it over a prolonged period with no cytotoxic effects seen in in vitro studies. Furthermore, the developed system does not show any undesired effects in vivo. When applied to wounds in genetically diabetic db/db mice, the fabricated system on its own improves reepithelialization and angiogenesis while decreasing inflammation. Also, key proteins involved in the wound healing process, i.e., Actn2, MYBPC1, and desmin, are upregulated in the glucose-responsive fibrin hydrogel (GRFHG) treated group of animals. In conclusion, the fabricated hydrogel promotes wound healing. Furthermore, the system may be encapsulated with various therapeutic nucleic acids that aid wound healing.
Collapse
Affiliation(s)
- Mangesh Morey
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Aitor Larrañaga
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Sunny Akogwu Abbah
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Amal Aljaabary
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
5
|
Kim S, Hwangbo H, Chae S, Lee H. Biopolymers and Their Application in Bioprinting Processes for Dental Tissue Engineering. Pharmaceutics 2023; 15:2118. [PMID: 37631331 PMCID: PMC10457894 DOI: 10.3390/pharmaceutics15082118] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Dental tissues are composed of multiple tissues with complex organization, such as dentin, gingiva, periodontal ligament, and alveolar bone. These tissues have different mechanical and biological properties that are essential for their functions. Therefore, dental diseases and injuries pose significant challenges for restorative dentistry, as they require innovative strategies to regenerate damaged or missing dental tissues. Biomimetic bioconstructs that can effectively integrate with native tissues and restore their functionalities are desirable for dental tissue regeneration. However, fabricating such bioconstructs is challenging due to the diversity and complexity of dental tissues. This review provides a comprehensive overview of the recent developments in polymer-based tissue engineering and three-dimensional (3D) printing technologies for dental tissue regeneration. It also discusses the current state-of-the-art, focusing on key techniques, such as polymeric biomaterials and 3D printing with or without cells, used in tissue engineering for dental tissues. Moreover, the final section of this paper identifies the challenges and future directions of this promising research field.
Collapse
Affiliation(s)
- Suhon Kim
- Barun Plant Orthodontics and Dental Clinic, Seongnam 13312, Republic of Korea;
| | - Hanjun Hwangbo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea; (H.H.); (S.C.)
| | - SooJung Chae
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea; (H.H.); (S.C.)
| | - Hyeongjin Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea; (H.H.); (S.C.)
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea
| |
Collapse
|
6
|
Hofmann AT, Slezak P, Neumann S, Ferguson J, Redl H, Mittermayr R. Ischemia Impaired Wound Healing Model in the Rat—Demonstrating Its Ability to Test Proangiogenic Factors. Biomedicines 2023; 11:biomedicines11041043. [PMID: 37189661 DOI: 10.3390/biomedicines11041043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Chronic wounds remain a serious clinical problem with insufficient therapeutic approaches. In this study we investigated the dose dependency of rhVEGF165 in fibrin sealant in both ischemic and non-ischemic excision wounds using our recently developed impaired-wound healing model. An abdominal flap was harvested from the rat with unilateral ligation of the epigastric bundle and consequent unilateral flap ischemia. Two excisional wounds were set in the ischemic and non-ischemic area. Wounds were treated with three different rhVEGF165 doses (10, 50 and 100 ng) mixed with fibrin or fibrin alone. Control animals received no therapy. Laser Doppler imaging (LDI) and immunohistochemistry were performed to verify ischemia and angiogenesis. Wound size was monitored with computed planimetric analysis. LDI revealed insufficient tissue perfusion in all groups. Planimetric analysis showed slower wound healing in the ischemic area in all groups. Wound healing was fastest with fibrin treatment—irrespective of tissue vitality. Lower dose VEGF (10 and 50 ng) led to faster wound healing compared to high-dose VEGF. Immunohistochemistry showed the highest vessel numbers in low-dose VEGF groups. In our previously established model, different rhVEGF165 treatments led to dose-dependent differences in angiogenesis and wound healing, but the fastest wound closure was achieved with fibrin matrix alone.
Collapse
|
7
|
Shaik R, Xu J, Wang Y, Hong Y, Zhang G. Fibrin-Enriched Cardiac Extracellular Matrix Hydrogel Promotes In Vitro Angiogenesis. ACS Biomater Sci Eng 2023; 9:877-888. [PMID: 36630688 PMCID: PMC10064974 DOI: 10.1021/acsbiomaterials.2c01148] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiogenesis is essential for cardiac repair after myocardial infarction. Promoting angiogenesis has been demonstrated as an effective approach for myocardial infarction treatment. Several different strategies for inducing myocardial angiogenesis have been explored, including exogenous delivery of angiogenic genes, proteins, microRNAs, cells, and extracellular vesicles. Various types of injectable hydrogels have been investigated for cardiac tissue repair. One of the most promising injectable hydrogels in cardiac regeneration is a cardiac extracellular matrix hydrogel that is derived from decellularized porcine myocardium. It can be delivered minimally invasively via transendocardial delivery. The safety and efficacy of cardiac extracellular matrix hydrogels have been shown in small and large animal myocardial infarction models as well as clinical trials. The main mechanisms underlying the therapeutic benefits of cardiac extracellular matrix hydrogels have been elucidated and involved in the modulation of the immune response, downregulation of pathways related to heart failure progression and fibrosis, upregulation of genes important for cardiac muscle contraction, and enhancing cardiomyocyte differentiation and maturation from stem cells. However, no potent capillary network formation induced by cardiac extracellular matrix hydrogels has been reported. In this study, we tested the feasibility of incorporating a fibrin matrix into cardiac extracellular matrix hydrogels to improve the angiogenic properties of the hydrogel. Our in vitro results demonstrate that fibrin-enriched cardiac extracellular matrix hydrogels can induce robust endothelial cell tube formation from human umbilical vein endothelial cells and promote the sprouting of human mesenchymal stem cell spheroids. The obtained information from this study is very critical toward the future in vivo evaluation of fibrin-enriched cardiac extracellular matrix hydrogels in promoting myocardial angiogenesis.
Collapse
Affiliation(s)
- Rubia Shaik
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, State College, University Park, Pennsylvania 16801, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
8
|
Soares DG, Rosa V. Regenerating the Dental Pulp-Scaffold Materials and Approaches. Dent Clin North Am 2022; 66:643-657. [PMID: 36216451 DOI: 10.1016/j.cden.2022.05.010] [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] [Indexed: 06/16/2023]
Abstract
Novel technologies and platforms have allowed significant breakthroughs in dental pulp tissue engineering. The development of injectable scaffolds that can be combined with stem cells, growth factors, or other bioactive compounds has enabled the regeneration of functional dental pulps able to secrete dentin in preclinical and clinical studies. Similarly, cell-homing technologies and scaffold-free strategies aim to modulate dental pulp self-regeneration mediated by resident stem cells and can evade some of the technical challenges related to cell-based tissue engineering strategies. This article will discuss emerging technologies and platforms for the clinical applications of dental pulp tissue engineering.
Collapse
Affiliation(s)
- Diana Gabriela Soares
- Department of Operative Dentistry, Endodontics and Dental Materials, São Paulo University - USP, Bauru School of Dentistry, Dr. Octavio Pinheiro Brizola, 9-75, Bauru, Sao Paulo 17012-901, Brazil.
| | - Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, 9 Lower Kent Ridge Road, Level 10, Singapore 119085, Singapore.
| |
Collapse
|
9
|
Deering J, Lin DSY, D'Elia A, Zhang B, Grandfield K. Fabrication of succinate-alginate xerogel films for in vitro coupling of osteogenesis and neovascularization. BIOMATERIALS ADVANCES 2022; 141:213122. [PMID: 36162345 DOI: 10.1016/j.bioadv.2022.213122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/30/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
The osseointegration of metallic implants is reliant on a cascade of molecular interactions and the delivery of macromolecules to the implant environment that occurs before substantial bone formation. Early blood vessel formation is a requisite first step in the healing timeline for osteoid formation, where vascular development can be accelerated as a result of controlled hypoxic conditioning. In this study, alginate-derived xerogel films containing varied concentrations of disodium succinate salt which has been shown to induce pseudohypoxia (short-term hypoxic effects while maintaining an oxygenated environment) were developed. Xerogels were characterized for their morphology, succinate release over time and cellular response with osteoblast-mimicking Saos-2 and human umbilical vein endothelial cells (HUVEC). Scanning electron microscopy revealed a multiscale topography that may favour osseointegration and alamarBlue assays indicated no cytotoxic effects during in vitro proliferation of Saos-2 cells. pH measurements of eluted succinate reach 95 % of peak value after 7 h of immersion for all gels containing 10 mM of succinate or less, and 60 % within the first 40 min. In vitro exposure of HUVECs to succinate-conditioned media increased the net concentration of total proteins measured by bicinchoninic acid (BCA) assay and maintains stable vascular endothelial growth factor (VEGF) and extracellular platelet-derived growth factor (PDGF) for vessel formation through comparison of enzyme-linked immunosorbent assays (ELISAs) of the culture media and cell lysate. Tube formation assays also showed a sustained increase in tube diameter across the first 48 h of HUVEC culture when succinate concentrations of 1 and 10 μM in the xerogel. Overall, the succinate-alginate films serve as a prospective organic coating for bone-interfacing implant materials which may induce temporary pseudohypoxic conditions favourable for early angiogenesis and bone regeneration in vivo at succinate concentrations of 1 or 10 μM.
Collapse
Affiliation(s)
- Joseph Deering
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada
| | - Dawn S Y Lin
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Andrew D'Elia
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada; School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Kathryn Grandfield
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada; School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada; Brockhouse Institute for Materials Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
10
|
Legrand JMD, Martino MM. Growth Factor and Cytokine Delivery Systems for Wound Healing. Cold Spring Harb Perspect Biol 2022; 14:a041234. [PMID: 35667794 PMCID: PMC9341469 DOI: 10.1101/cshperspect.a041234] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Skin wound healing is a highly coordinated process involving multiple tissue-resident and recruited cell types. Cells within the wound microenvironment respond to key secreted factors such as pro-proliferative growth factors and immunomodulatory cytokines to repair the skin and promptly restore its essential barrier role. Therefore, recombinant growth factors and cytokines are promising therapeutics for skin wounds, in particular for large acute wounds such as burns, or wounds associated with underlying pathologies such as nonhealing chronic and diabetic wounds. However, translation of growth factors and cytokines into clinically effective treatments has been limited. Short half-life, poor stability, rapid diffusion, uncontrolled signaling, and systemic side effects are currently the key challenges to developing efficient growth factor- and cytokine-based therapies. To overcome these limitations, novel delivery systems have been developed to improve the regenerative potential of recombinant growth factors and cytokines. In this review, we discuss biomaterial and protein engineering strategies used to optimize the delivery of growth factor and cytokine therapeutics for skin wound treatment.
Collapse
Affiliation(s)
- Julien M D Legrand
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
11
|
Burger MG, Grosso A, Briquez PS, Born GME, Lunger A, Schrenk F, Todorov A, Sacchi V, Hubbell JA, Schaefer DJ, Banfi A, Di Maggio N. Robust coupling of angiogenesis and osteogenesis by VEGF-decorated matrices for bone regeneration. Acta Biomater 2022; 149:111-125. [PMID: 35835287 DOI: 10.1016/j.actbio.2022.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022]
Abstract
Rapid vascularization of clinical-size bone grafts is an unsolved challenge in regenerative medicine. Vascular endothelial growth factor-A (VEGF) is the master regulator of angiogenesis. Its over-expression by genetically modified human osteoprogenitors has been previously evaluated to drive vascularization in osteogenic grafts, but has been observed to cause paradoxical bone loss through excessive osteoclast recruitment. However, during bone development angiogenesis and osteogenesis are physiologically coupled by VEGF expression. Here we investigated whether the mode of VEGF delivery may be a key to recapitulate its physiological function. VEGF activity requires binding to the extracellular matrix, and heterogeneous levels of expression lead to localized microenvironments of excessive dose. Therefore we hypothesized that a homogeneous distribution of matrix-associated factor in the microenvironment may enable efficient coupling of angiogenesis and bone formation. This was achieved by decorating fibrin matrices with a cross-linkable engineered version of VEGF (TG-VEGF) that is released only by enzymatic cleavage by invading cells. In ectopic grafts, both TG-VEGF and VEGF-expressing progenitors similarly improved vascularization within the first week, but efficient bone formation was possible only in the factor-decorated matrices, whereas heterogenous, cell-based VEGF expression caused significant bone loss. In critical-size orthotopic calvaria defects, TG-VEGF effectively improved early vascular invasion, osteoprogenitor survival and differentiation, as well as bone repair compared to both controls and VEGF-expressing progenitors. In conclusion, homogenous distribution of matrix-associated VEGF protein preserves the physiological coupling of angiogenesis and osteogenesis, providing an attractive and clinically applicable strategy to engineer vascularized bone. STATEMENT OF SIGNIFICANCE: The therapeutic regeneration of vascularized bone is an unsolved challenge in regenerative medicine. Stimulation of blood vessel growth by over-expression of VEGF has been associated with paradoxical bone loss, whereas angiogenesis and osteogenesis are physiologically coupled by VEGF during development. Here we found that controlling the distribution of VEGF dose in an osteogenic graft is key to recapitulate its physiological function. In fact, homogeneous decoration of fibrin matrices with engineered VEGF could improve both vascularization and bone formation in orthotopic critical-size defects, dispensing with the need for combined osteogenic factor delivery. VEGF-decorated fibrin matrices provide a readily translatable platform for engineering a controlled microenvironment for bone regeneration.
Collapse
Affiliation(s)
- Maximilian G Burger
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland; Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland
| | - Andrea Grosso
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Priscilla S Briquez
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S Ellis Ave, Chicago, IL 60637, USA
| | - Gordian M E Born
- Tissue Engineering, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Alexander Lunger
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland; Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland
| | - Flavio Schrenk
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Atanas Todorov
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland; Tissue Engineering, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Veronica Sacchi
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, 5640 S Ellis Ave, Chicago, IL 60637, USA
| | - Dirk J Schaefer
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland; Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland
| | - Andrea Banfi
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland; Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Petersgraben 4, 4031, Basel, Switzerland.
| | - Nunzia Di Maggio
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| |
Collapse
|
12
|
Zhang W, Wu Y, Chen Q, Zhang H, Zhou M, Chen K, Cao C, Guo H, Xu J, Liu H, Lin H, Liu C, Liu R. Statistic Copolymers Working as Growth Factor-Binding Mimics of Fibronectin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200775. [PMID: 35570405 PMCID: PMC9313494 DOI: 10.1002/advs.202200775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/06/2022] [Indexed: 05/07/2023]
Abstract
Growth factors (GFs) play important roles in biological system and are widely used in tissue regeneration. However, their application is greatly hindered by short in vivo lifetime of GFs. GFs are bound to fibronectin dynamically in the extracellular matrix, which inspired the authors to mimic the GF binding domain of fibronectin and design GF-binding amphiphilic copolymers bearing positive charges. The optimal amino acid polymer can bind to a variety of representative GFs, such as bone morphogenetic protein-2 (BMP-2) and TGF-β1 from the transforming growth factor-β superfamily, PDGF-AA and PDGF-BB from the platelet-derived growth factor family, FGF-10 and FGF-21 from the fibroblast growth factor family, epidermal growth factor from the EGF family and hepatocyte growth factor from the plasminogen-related growth factor family, with binding affinities up to the nanomolar level. 3D scaffolds immobilized with the optimal copolymer enable sustained release of loaded BMP-2 without burst release and significantly enhances the in vivo function of BMP-2 for bone formation. This strategy opens new avenues in designing GF-binding copolymers as synthetic mimics of fibronectin for diverse applications.
Collapse
Affiliation(s)
- Wenjing Zhang
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Yueming Wu
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Qi Chen
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Haodong Zhang
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Min Zhou
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Kang Chen
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Chuntao Cao
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Han Guo
- Shanghai Synchrotron Radiation Facility (SSRF)Shanghai Advanced Research InstituteChinese Academy of SciencesShanghai201204China
| | - Jianrong Xu
- Academy of Integrative MedicineShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Honglai Liu
- School of Chemistry and Molecular EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Haodong Lin
- Department of Orthopedic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Runhui Liu
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| |
Collapse
|
13
|
Huang J, Han Q, Cai M, Zhu J, Li L, Yu L, Wang Z, Fan G, Zhu Y, Lu J, Zhou G. Effect of Angiogenesis in Bone Tissue Engineering. Ann Biomed Eng 2022; 50:898-913. [PMID: 35525871 DOI: 10.1007/s10439-022-02970-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 04/17/2022] [Indexed: 12/20/2022]
Abstract
The reconstruction of large skeletal defects is still a tricky challenge in orthopedics. The newly formed bone tissue migrates sluggishly from the periphery to the center of the scaffold due to the restrictions of exchange of oxygen and nutrition impotent cells osteogenic differentiation. Angiogenesis plays an important role in bone reconstruction and more and more studies on angiogenesis in bone tissue engineering had been published. Promising advances of angiogenesis in bone tissue engineering by scaffold designs, angiogenic factor delivery, in vivo prevascularization and in vitro prevascularization are discussed in detail. Among all the angiogenesis mode, angiogenic factor delivery is the common methods of angiogenesis in bone tissue engineering and possible research directions in the future.
Collapse
Affiliation(s)
- Jianhao Huang
- Department of Orthopedics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, People's Republic of China
| | - Qixiu Han
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China
| | - Meng Cai
- Department of Orthopedics, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210002, People's Republic of China
| | - Jie Zhu
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Lingfeng Yu
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China
| | - Zhen Wang
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China
| | - Gentao Fan
- Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China
| | - Yan Zhu
- Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China
| | - Jingwei Lu
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China. .,Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China.
| | - Guangxin Zhou
- Department of Orthopedics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, People's Republic of China. .,Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China. .,Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China. .,The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
14
|
Palanisamy P, Alam M, Li S, Chow SKH, Zheng Y. Low-Intensity Pulsed Ultrasound Stimulation for Bone Fractures Healing: A Review. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:547-563. [PMID: 33949710 PMCID: PMC9290611 DOI: 10.1002/jum.15738] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/04/2021] [Accepted: 04/18/2021] [Indexed: 05/17/2023]
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a developing technology, which has been proven to improve fracture healing process with minimal thermal effects. This noninvasive treatment accelerates bone formation through various molecular, biological, and biomechanical interactions with tissues and cells. Although LIPUS treatment has shown beneficial effects on different bone fracture locations, only very few studies have examined its effects on deeper bones. This study provides an overview on therapeutic ultrasound for fractured bones, possible mechanisms of action, clinical evidences, current limitations, and its future prospects.
Collapse
Affiliation(s)
- Poornima Palanisamy
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong KongS.A.RChina
| | - Monzurul Alam
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong KongS.A.RChina
| | - Shuai Li
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong KongS.A.RChina
| | - Simon K. H. Chow
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong KongS.A.RChina
| | - Yong‐Ping Zheng
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong KongS.A.RChina
| |
Collapse
|
15
|
Fibrin-based factor delivery for therapeutic angiogenesis: friend or foe? Cell Tissue Res 2022; 387:451-460. [PMID: 35175429 PMCID: PMC8975770 DOI: 10.1007/s00441-022-03598-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 12/28/2022]
Abstract
Therapeutic angiogenesis aims at promoting the growth of blood vessels to restore perfusion in ischemic tissues or aid tissue regeneration. Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis in development, repair, and disease. However, exploiting VEGF for therapeutic purposes has been challenging and needs to take into account some key aspects of VEGF biology. In particular, the spatial localization of angiogenic signals within the extracellular matrix is crucial for physiological assembly and function of new blood vessels. Fibrin is the provisional matrix that is universally deposited immediately after injury and supports the initial steps of tissue regeneration. It provides therefore several ideal features as a substrate to promote therapeutic vascularization, especially through its ability to present growth factors in their physiological matrix-bound state and to modulate their availability for signaling. Here, we provide an overview of fibrin uses as a tissue-engineering scaffold material and as a tunable platform to finely control dose and duration of delivery of recombinant factors in therapeutic angiogenesis. However, in some cases, fibrin has also been associated with undesirable outcomes, namely the promotion of fibrosis and scar formation that actually prevent physiological tissue regeneration. Understanding the mechanisms that tip the balance between the pro- and anti-regenerative functions of fibrin will be the key to fully exploit its therapeutic potential.
Collapse
|
16
|
Plasminogen-Loaded Fibrin Scaffold as Drug Delivery System for Wound Healing Applications. Pharmaceutics 2022; 14:pharmaceutics14020251. [PMID: 35213982 PMCID: PMC8879571 DOI: 10.3390/pharmaceutics14020251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 01/18/2022] [Indexed: 01/13/2023] Open
Abstract
Plasminogen is a protein involved in intravascular and extravascular fibrinolysis, as well as in wound healing, cell migration, tissue formation and angiogenesis. In recent years its role in healing of tympanic perforations has been demonstrated in plasminogen deficient mice. The aim of this work was to fabricate a fibrin-based drug delivery system able to provide a local and sustained release of plasminogen at the wound site. Initially, the biological activity of plasminogen was evaluated by in vitro experiments on cell cultures. A metabolic assay (MTT) was carried out on L929 mouse fibroblast to determine the concentration that does not affect cell viability, which turned out to be 64 nM. The effect of plasminogen on cell migration was evaluated through a scratch test on human keratinocytes: cells treated with 64 nM plasminogen showed faster scratch closure than in complete medium. Fibrin scaffold loaded with plasminogen was fabricated by a spray process. SEM analysis showed the typical nano-fibrillar structure of a fibrin scaffold. Tensile tests highlighted significantly higher value of the ultimate stress and strain of fibrin scaffold with respect to fibrin clot. The in-vitro release kinetic showed an initial plasminogen burst, after that the release slowed, reaching a plateau at 7 days. Plasminogen-loaded fibrin scaffold applied in full-thickness diabetic mouse lesions showed a significantly higher closure rate at 14 days than scaffold used as a reference material. Histological analysis demonstrated an improved reepithelization and collagen deposition in granulation tissue in mouse treated with plasminogen-loaded fibrin scaffold in comparison to unloaded fibrin scaffold. The obtained results demonstrated the suitability of the fibrin scaffold loaded with plasminogen as drug delivery system and suggest its use in wound healing applications, such as for the treatment of chronic diabeticulcers.
Collapse
|
17
|
Linsley CS, Sung K, White C, Abecunas CA, Tawil BJ, Wu BM. Functionalizing Fibrin Hydrogels with Thermally Responsive Oligonucleotide Tethers for On-Demand Delivery. Bioengineering (Basel) 2022; 9:bioengineering9010025. [PMID: 35049734 PMCID: PMC8773154 DOI: 10.3390/bioengineering9010025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
There are a limited number of stimuli-responsive biomaterials that are capable of delivering customizable dosages of a therapeutic at a specific location and time. This is especially true in tissue engineering and regenerative medicine applications, where it may be desirable for the stimuli-responsive biomaterial to also serve as a scaffolding material. Therefore, the purpose of this study was to engineer a traditionally non-stimuli responsive scaffold biomaterial to be thermally responsive so it could be used for on-demand drug delivery applications. Fibrin hydrogels are frequently used for tissue engineering and regenerative medicine applications, and they were functionalized with thermally labile oligonucleotide tethers using peptides from substrates for factor XIII (FXIII). The alpha 2-plasmin inhibitor peptide had the greatest incorporation efficiency out of the FXIII substrate peptides studied, and conjugates of the peptide and oligonucleotide tethers were successfully incorporated into fibrin hydrogels via enzymatic activity. Single-strand complement oligo with either a fluorophore model drug or platelet-derived growth factor-BB (PDGF-BB) could be released on demand via temperature increases. These results demonstrate a strategy that can be used to functionalize traditionally non-stimuli responsive biomaterials suitable for on-demand drug delivery systems (DDS).
Collapse
Affiliation(s)
- Chase S. Linsley
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA 90095, USA; (K.S.); (C.W.); (C.A.A.); (B.J.T.)
- Correspondence: (C.S.L.); (B.M.W.)
| | - Kevin Sung
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA 90095, USA; (K.S.); (C.W.); (C.A.A.); (B.J.T.)
| | - Cameron White
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA 90095, USA; (K.S.); (C.W.); (C.A.A.); (B.J.T.)
| | - Cara A. Abecunas
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA 90095, USA; (K.S.); (C.W.); (C.A.A.); (B.J.T.)
| | - Bill J. Tawil
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA 90095, USA; (K.S.); (C.W.); (C.A.A.); (B.J.T.)
| | - Benjamin M. Wu
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA 90095, USA; (K.S.); (C.W.); (C.A.A.); (B.J.T.)
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Department of Materials Science & Engineering, Samueli School of Engineering, University of California, Los Angeles, CA 90095, USA
- Correspondence: (C.S.L.); (B.M.W.)
| |
Collapse
|
18
|
Avilla-Royo E, Ochsenbein-Kölble N, Vonzun L, Ehrbar M. Biomaterial-based treatments for the prevention of preterm birth after iatrogenic rupture of the fetal membranes. Biomater Sci 2022; 10:3695-3715. [DOI: 10.1039/d2bm00401a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Minimally invasive interventions to ameliorate or correct fetal abnormalities are becoming a clinical reality. However, the iatrogenic premature preterm rupture of the fetal membranes (FMs) (iPPROM), which may result in...
Collapse
|
19
|
Kulkarni NS, Chauhan G, Goyal M, Sarvepalli S, Gupta V. Development of Gelatin Methacrylate (GelMa) Hydrogels for Versatile Intracavitary Applications: In-vitro Characterization and Ex-vivo Performance Assessment. Biomater Sci 2022; 10:4492-4507. [DOI: 10.1039/d2bm00022a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Applicability of hydrogels as drug delivery systems is on the rise due to their highly tunable degree of polymeric crosslinking to attain varying rates of payload release. Sustaining the release...
Collapse
|
20
|
3D Bioprinting for fabrication of tissue models of COVID-19 infection. Essays Biochem 2021; 65:503-518. [PMID: 34028514 DOI: 10.1042/ebc20200129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/19/2022]
Abstract
Over the last few decades, the world has witnessed multiple viral pandemics, the current severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) pandemic being the worst and most devastating one, claiming millions of lives worldwide. Physicians, scientists, and engineers worldwide have joined hands in dealing with the current situation at an impressive speed and efficiency. One of the major reasons for the delay in response is our limited understanding of the mechanism of action and individual effects of the virus on different tissues and organs. Advances in 3D bioprinting have opened up a whole new area to explore and utilize the technology in fabricating models of these tissues and organs, recapitulating in vivo environment. These biomimetic models can not only be utilized in learning the infection pathways and drug toxicology studies but also minimize the need for animal models and shorten the time span for human clinical trials. The current review aims to integrate the existing developments in bioprinting techniques, and their implementation to develop tissue models, which has implications for SARS-CoV-2 infection. Future translation of these models has also been discussed with respect to the pandemic.
Collapse
|
21
|
Lopes SV, Collins MN, Reis RL, Oliveira JM, Silva-Correia J. Vascularization Approaches in Tissue Engineering: Recent Developments on Evaluation Tests and Modulation. ACS APPLIED BIO MATERIALS 2021; 4:2941-2956. [DOI: 10.1021/acsabm.1c00051] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Soraia V. Lopes
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maurice N. Collins
- Bernal Institute, School of Engineering, University of Limerick, Limerick V94 T9PX, Ireland
| | - Rui L. Reis
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Silva-Correia
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
22
|
Liu R, Zuo R, Hudalla GA. Harnessing molecular recognition for localized drug delivery. Adv Drug Deliv Rev 2021; 170:238-260. [PMID: 33484737 PMCID: PMC8274479 DOI: 10.1016/j.addr.2021.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
A grand challenge in drug delivery is providing the right dose, at the right anatomic location, for the right duration of time to maximize therapeutic efficacy while minimizing off-target toxicity and other deleterious side-effects. Two general modalities are receiving broad attention for localized drug delivery. In the first, referred to as "targeted accumulation", drugs or drug carriers are engineered to have targeting moieties that promote their accumulation at a specific tissue site from circulation. In the second, referred to as "local anchoring", drugs or drug carriers are inserted directly into the tissue site of interest where they persist for a specified duration of time. This review surveys recent advances in harnessing molecular recognition between proteins, peptides, nucleic acids, lipids, and carbohydrates to mediate targeted accumulation and local anchoring of drugs and drug carriers.
Collapse
Affiliation(s)
- Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
23
|
Utilisation of Chick Embryo Chorioallantoic Membrane as a Model Platform for Imaging-Navigated Biomedical Research. Cells 2021; 10:cells10020463. [PMID: 33671534 PMCID: PMC7926796 DOI: 10.3390/cells10020463] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
The fertilised chick egg and particularly its chorioallantoic membrane (CAM) have drawn continuing interest in biomedicine and bioengineering fields, especially for research on vascular study, cancer, drug screening and development, cell factors, stem cells, etc. This literature review systemically introduces the CAM's structural evolution, functions, vascular features and the circulation system, and cell regulatory factors. It also presents the major and updated applications of the CAM in assays for pharmacokinetics and biodistribution, drug efficacy and toxicology testing/screening in preclinical pharmacological research. The time course of CAM applications for different assays and their advantages and limitations are summarised. Among these applications, two aspects are emphasised: (1) potential utility of the CAM for preclinical studies on vascular-disrupting agents (VDAs), promising for anti-cancer vascular-targeted therapy, and (2) modern imaging technologies, including modalities and their applications for real-time visualisation, monitoring and evaluation of the changes in CAM vasculature as well as the interactions occurring after introducing the tested medical, pharmaceutical and biological agents into the system. The aim of this article is to help those working in the biomedical field to familiarise themselves with the chick embryo CAM as an alternative platform and to utilise it to design and optimise experimental settings for their specific research topics.
Collapse
|
24
|
Emelianov VY, Preobrazhenskaia EV, Nikolaev NS. Evaluating the Effectiveness of Biophysical Methods of Osteogenesis Stimulation: Review. TRAUMATOLOGY AND ORTHOPEDICS OF RUSSIA 2021; 27:86-96. [DOI: https:/doi.org/10.21823/2311-2905-2021-27-1-86-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Background. Stimulation of osteogenesis (SO) by biophysical methods has been widely used in practice to accelerate healing or stimulate the healing of fractures with non-unions, since the middle of the XIX century. SO can be carried out by direct current electrostimulation, or indirectly by low-intensity pulsed ultrasound, capacitive electrical coupling stimulation, and pulsed electromagnetic field stimulation. SO simulates natural physiological processes: in the case of electrical stimulation, it changes the electromagnetic potential of damaged cell tissues in a manner similar to normal healing processes, or in the case of low-intensity pulsed ultrasound, it produces weak mechanical effects on the fracture area. SO increases the expression of factors and signaling pathways responsible for tissue regeneration and bone mineralization and ultimately accelerates bone union.The purpose of this review was to present the most up-to-date data from laboratory and clinical studies of the effectiveness of SO.Material and Methods. The results of laboratory studies and the final results of metaanalyses for each of the four SO methods published from 1959 to 2020 in the PubMed, EMBASE, and eLibrary databases are reviewed.Conclusion. The use of SO effectively stimulates the healing of fractures with the correct location of the sensors, compliance with the intensity and time of exposure, as well as the timing of use for certain types of fractures. In case of non-union or delayed union of fractures, spondylodesis, arthrodesis, preference should be given to non-invasive methods of SO. Invasive direct current stimulation can be useful for non-union of long bones, spondylodesis with the risk of developing pseudoarthrosis.
Collapse
|
25
|
Emelianov VY, Preobrazhenskaia EV, Nikolaev NS. Evaluating the Effectiveness of Biophysical Methods of Osteogenesis Stimulation: Review. TRAUMATOLOGY AND ORTHOPEDICS OF RUSSIA 2021; 27:86-96. [DOI: 10.21823/2311-2905-2021-27-1-86-96] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background. Stimulation of osteogenesis (SO) by biophysical methods has been widely used in practice to accelerate healing or stimulate the healing of fractures with non-unions, since the middle of the XIX century. SO can be carried out by direct current electrostimulation, or indirectly by low-intensity pulsed ultrasound, capacitive electrical coupling stimulation, and pulsed electromagnetic field stimulation. SO simulates natural physiological processes: in the case of electrical stimulation, it changes the electromagnetic potential of damaged cell tissues in a manner similar to normal healing processes, or in the case of low-intensity pulsed ultrasound, it produces weak mechanical effects on the fracture area. SO increases the expression of factors and signaling pathways responsible for tissue regeneration and bone mineralization and ultimately accelerates bone union.The purpose of this review was to present the most up-to-date data from laboratory and clinical studies of the effectiveness of SO.Material and Methods. The results of laboratory studies and the final results of metaanalyses for each of the four SO methods published from 1959 to 2020 in the PubMed, EMBASE, and eLibrary databases are reviewed.Conclusion. The use of SO effectively stimulates the healing of fractures with the correct location of the sensors, compliance with the intensity and time of exposure, as well as the timing of use for certain types of fractures. In case of non-union or delayed union of fractures, spondylodesis, arthrodesis, preference should be given to non-invasive methods of SO. Invasive direct current stimulation can be useful for non-union of long bones, spondylodesis with the risk of developing pseudoarthrosis.
Collapse
|
26
|
Catanzano O, Quaglia F, Boateng JS. Wound dressings as growth factor delivery platforms for chronic wound healing. Expert Opin Drug Deliv 2021; 18:737-759. [PMID: 33338386 DOI: 10.1080/17425247.2021.1867096] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Years of tissue engineering research have clearly demonstrated the potential of integrating growth factors (GFs) into scaffolds for tissue regeneration, a concept that has recently been applied to wound dressings. The old concept of wound dressings that only take a passive role in wound healing has now been overtaken, and advanced dressings which can take an active part in wound healing, are of current research interest.Areas covered: In this review we will focus on the recent strategies for the delivery of GFs to wound sites with an emphasis on the different approaches used to achieve fine tuning of spatial and temporal concentrations to achieve therapeutic efficacy.Expert opinion: The use of GFs to accelerate wound healing and reduce scar formation is now considered a feasible therapeutic approach in patients with a high risk of infections and complications. The integration of micro - and nanotechnologies into wound dressings could be the key to overcome the inherent instability of GFs and offer adequate control over the release rate. Many investigations have led to encouraging outcomes in various in vitro and in vivo wound models, and it is expected that some of these technologies will satisfy clinical needs and will enter commercialization.
Collapse
Affiliation(s)
- Ovidio Catanzano
- Institute for Polymers Composites and Biomaterials (IPCB) - CNR, Pozzuoli, Italy
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Joshua S Boateng
- School of Science, Faculty of Engineering and Science, University of Greenwich, Medway, Central Avenue, Chatham Maritime, Kent, UK
| |
Collapse
|
27
|
Hydrogels as Drug Delivery Systems: A Review of Current Characterization and Evaluation Techniques. Pharmaceutics 2020; 12:pharmaceutics12121188. [PMID: 33297493 PMCID: PMC7762425 DOI: 10.3390/pharmaceutics12121188] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022] Open
Abstract
Owing to their tunable properties, controllable degradation, and ability to protect labile drugs, hydrogels are increasingly investigated as local drug delivery systems. However, a lack of standardized methodologies used to characterize and evaluate drug release poses significant difficulties when comparing findings from different investigations, preventing an accurate assessment of systems. Here, we review the commonly used analytical techniques for drug detection and quantification from hydrogel delivery systems. The experimental conditions of drug release in saline solutions and their impact are discussed, along with the main mathematical and statistical approaches to characterize drug release profiles. We also review methods to determine drug diffusion coefficients and in vitro and in vivo models used to assess drug release and efficacy with the goal to provide guidelines and harmonized practices when investigating novel hydrogel drug delivery systems.
Collapse
|
28
|
Zhou F, Hong Y, Liang R, Zhang X, Liao Y, Jiang D, Zhang J, Sheng Z, Xie C, Peng Z, Zhuang X, Bunpetch V, Zou Y, Huang W, Zhang Q, Alakpa EV, Zhang S, Ouyang H. Rapid printing of bio-inspired 3D tissue constructs for skin regeneration. Biomaterials 2020; 258:120287. [DOI: 10.1016/j.biomaterials.2020.120287] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/14/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
|
29
|
Zheng Z, Yu C, Wei H. Injectable Hydrogels as Three-Dimensional Network Reservoirs for Osteoporosis Treatment. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:430-454. [PMID: 33086984 DOI: 10.1089/ten.teb.2020.0168] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite tremendous progresses made in the field of tissue engineering over the past several decades, it remains a significant challenge for the treatment of osteoporosis (OP) due to the lack of appropriate carriers to improve the bioavailability of therapeutic agents and the unavailability of artificial bone matrix with desired properties for the replacement of damaged bone regions. Encouragingly, the development of injectable hydrogels for the treatment of OP has attracted increasing attention in recent years because they can serve either as a reservoir for various therapeutic species or as a perfect filler for bone injuries with irregular shapes. However, the relationship between the complicated pathological mechanism of OP and the properties of diverse polymeric materials lacks elucidation, which clearly hampers the clinical application of injectable hydrogels for the efficient treatment of OP. To clarify this relationship, this article summarized both localized and systematic treatment of OP using an injectable hydrogel-based strategy. Specifically, the pathogenesis of OP and the limitations of current treatment approaches were first analyzed. We further focused on the use of hydrogels loaded with various therapeutic substances following a classification standard of the encapsulated cargoes for OP treatment with an emphasis on the application and precautions of each category. A concluding remark on existing challenges and future directions of this rapidly developing research area was finally made. Impact statement Effective osteoporosis (OP) treatment remains a significant challenge due substantially to the unavailability of appropriate drug carriers and artificial matrices with desired properties to promote bone repair and replace damaged regions. For this purpose, this review focused on the development of diverse injectable hydrogel systems for the delivery of various therapeutic agents, including drugs, stem cells, and nucleic acids, for effective increase in bone mass and favorable osteogenesis. The summarized important guidelines are believed to promote clinical development and translation of hydrogels for the efficient treatment of OP and OP-related bone damages toward improved life quality of millions of patients.
Collapse
Affiliation(s)
- Zhi Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study and School of Pharmaceutical Science, University of South China, Hengyang, China
| | - Cuiyun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study and School of Pharmaceutical Science, University of South China, Hengyang, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study and School of Pharmaceutical Science, University of South China, Hengyang, China
| |
Collapse
|
30
|
Development of Injectable Thermosensitive Chitosan-Based Hydrogels for Cell Encapsulation. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10186550] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The three-dimensional complexity of the native extracellular matrix (ECM) suggests switching from 2D to 3D culture systems for providing the cells with an architecture more similar to the physiological environment. Reproducing the three-dimensionality in vitro can guarantee beneficial effects in terms of cell growth, adhesion, proliferation, and/or their differentiation. Hydrogels have the same tailorable physico-chemical and biological characteristics as ECM materials. In this study, we propose a thermoresponsive chitosan-based hydrogel that gels thanks to the addition of organic and inorganic salt solutions (beta-glycerolphosphate and sodium hydrogen carbonate) and is suitable for cell encapsulation allowing obtaining 3D culture systems. Physico-chemical analyses showed that the hydrogel formulations jellify at physiological conditions (37 °C, pH 7.4), are stable in vitro up to three weeks, have high swelling ratios and mechanical stiffness suitable for cellular encapsulation. Moreover, preliminary biological tests underlined the pronounced biocompatibility of the system. Therefore, these chitosan-based hydrogels are proposed as valid biomaterials for cell encapsulation.
Collapse
|
31
|
Victorelli FD, Cardoso VMDO, Ferreira NN, Calixto GMF, Fontana CR, Baltazar F, Gremião MPD, Chorilli M. Chick embryo chorioallantoic membrane as a suitable in vivo model to evaluate drug delivery systems for cancer treatment: A review. Eur J Pharm Biopharm 2020; 153:273-284. [DOI: 10.1016/j.ejpb.2020.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022]
|
32
|
Oliva N, Almquist BD. Spatiotemporal delivery of bioactive molecules for wound healing using stimuli-responsive biomaterials. Adv Drug Deliv Rev 2020; 161-162:22-41. [PMID: 32745497 DOI: 10.1016/j.addr.2020.07.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/23/2020] [Indexed: 12/28/2022]
Abstract
Wound repair is a fascinatingly complex process, with overlapping events in both space and time needed to pave a pathway to successful healing. This additional complexity presents challenges when developing methods for the controlled delivery of therapeutics for wound repair and tissue engineering. Unlike more traditional applications, where biomaterial-based depots increase drug solubility and stability in vivo, enhance circulation times, and improve retention in the target tissue, when aiming to modulate wound healing, there is a desire to enable localised, spatiotemporal control of multiple therapeutics. Furthermore, many therapeutics of interest in the context of wound repair are sensitive biologics (e.g. growth factors), which present unique challenges when designing biomaterial-based delivery systems. Here, we review the diverse approaches taken by the biomaterials community for creating stimuli-responsive materials that are beginning to enable spatiotemporal control over the delivery of therapeutics for applications in tissue engineering and regenerative medicine.
Collapse
|
33
|
Zafar MS, Amin F, Fareed MA, Ghabbani H, Riaz S, Khurshid Z, Kumar N. Biomimetic Aspects of Restorative Dentistry Biomaterials. Biomimetics (Basel) 2020; 5:E34. [PMID: 32679703 PMCID: PMC7557867 DOI: 10.3390/biomimetics5030034] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Biomimetic has emerged as a multi-disciplinary science in several biomedical subjects in recent decades, including biomaterials and dentistry. In restorative dentistry, biomimetic approaches have been applied for a range of applications, such as restoring tooth defects using bioinspired peptides to achieve remineralization, bioactive and biomimetic biomaterials, and tissue engineering for regeneration. Advancements in the modern adhesive restorative materials, understanding of biomaterial-tissue interaction at the nano and microscale further enhanced the restorative materials' properties (such as color, morphology, and strength) to mimic natural teeth. In addition, the tissue-engineering approaches resulted in regeneration of lost or damaged dental tissues mimicking their natural counterpart. The aim of the present article is to review various biomimetic approaches used to replace lost or damaged dental tissues using restorative biomaterials and tissue-engineering techniques. In addition, tooth structure, and various biomimetic properties of dental restorative materials and tissue-engineering scaffold materials, are discussed.
Collapse
Affiliation(s)
- Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah, Al Munawwarah 41311, Saudi Arabia;
- Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan
| | - Faiza Amin
- Science of Dental Materials Department, Dow Dental College, Dow University of Health Sciences, Karachi 74200, Pakistan;
| | - Muhmmad Amber Fareed
- Adult Restorative Dentistry, Dental Biomaterials and Prosthodontics Oman Dental College, Muscat 116, Sultanate of Oman;
| | - Hani Ghabbani
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah, Al Munawwarah 41311, Saudi Arabia;
| | - Samiya Riaz
- School of Dental Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Zohaib Khurshid
- Department of Prosthodontics and Dental Implantology, College of Dentistry, King Faisal University, Al-Ahsa 31982, Saudia Arabia;
| | - Naresh Kumar
- Department of Science of Dental Materials, Dow University of Health Sciences, Karachi 74200, Pakistan;
| |
Collapse
|
34
|
Wright ME, Yu JK, Jain D, Maeda A, Yeh SCA, DaCosta RS, Lin CP, Santerre JP. Engineering functional microvessels in synthetic polyurethane random-pore scaffolds by harnessing perfusion flow. Biomaterials 2020; 256:120183. [PMID: 32622017 DOI: 10.1016/j.biomaterials.2020.120183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/24/2022]
Abstract
Recently reported biomaterial-based approaches toward prevascularizing tissue constructs rely on biologically or structurally complex scaffolds that are complicated to manufacture and sterilize, and challenging to customize for clinical applications. In the current work, a prevascularization method for soft tissue engineering that uses a non-patterned and non-biological scaffold is proposed. Human fibroblasts and HUVECs were seeded on an ionomeric polyurethane-based hydrogel and cultured for 14 days under medium perfusion. A flow rate of 0.05 mL/min resulted in a greater lumen density in the constructs relative to 0.005 and 0.5 mL/min, indicating the critical importance of flow magnitude in establishing microvessels. Constructs generated at 0.05 mL/min perfusion flow were implanted in a mouse subcutaneous model and intravital imaging was used to characterize host blood perfusion through the construct after 2 weeks. Engineered microvessels were functional (i.e. perfused with host blood and non-leaky) and neovascularization of the construct by host vessels was enhanced relative to non-prevascularized constructs. We report on the first strategy toward engineering functional microvessels in a tissue construct using non-bioactive, non-patterned synthetic polyurethane materials.
Collapse
Affiliation(s)
- Meghan Ee Wright
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Jonathan K Yu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Devika Jain
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Azusa Maeda
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Shu-Chi A Yeh
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph S DaCosta
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
35
|
Huang CH, Lei KF. Impedimetric quantification of migration speed of cancer cells migrating along a Matrigel-filled microchannel. Anal Chim Acta 2020; 1121:67-73. [DOI: 10.1016/j.aca.2020.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 12/21/2022]
|
36
|
Sanada F, Fujikawa T, Shibata K, Taniyama Y, Rakugi H, Morishita R. Therapeutic Angiogenesis Using HGF Plasmid. Ann Vasc Dis 2020; 13:109-115. [PMID: 32595785 PMCID: PMC7315247 DOI: 10.3400/avd.ra.20-00035] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 12/27/2022] Open
Abstract
Hepatocyte growth factor (HGF) is secreted from stromal and mesenchymal cells, and its receptor cMet is expressed on various types of cells such as smooth muscle cells, fibroblast, and endothelial cells. HGF stimulates epithelial and endothelial cell proliferation, motility, and morphogenesis in a paracrine and autocrine manner, organizing multistep of angiogenesis in many organs. In addition, HGF is recognized as a potent anti-inflammatory and anti-fibrotic growth factor, which has been proved in several animal studies, including neointimal hyperplasia and acute myocardial infarction model in rodent. Thus, as compared to other angiogenic growth factors, HGF exerts multiple effects on ischemic tissues, accompanied by the regression of tissue inflammation and fibrosis. These data suggest the therapeutic potential of the HGF for peripheral artery disease as it being accompanied with chronic tissue inflammation and fibrosis. In the present narrative review, the pleiotropic action of the HGF that differentiates it from other angiogenic growth factors is discussed first, and later, outcomes of the human clinical study with gene therapy are overviewed.
Collapse
Affiliation(s)
- Fumihiro Sanada
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tatsuya Fujikawa
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kana Shibata
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiaki Taniyama
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
37
|
Ervolino De Oliveira C, Dourado MR, Sawazaki-Calone Í, Costa De Medeiros M, Rossa Júnior C, De Karla Cervigne N, Esquiche León J, Lambert D, Salo T, Graner E, Coletta RD. Activin A triggers angiogenesis via regulation of VEGFA and its overexpression is associated with poor prognosis of oral squamous cell carcinoma. Int J Oncol 2020; 57:364-376. [PMID: 32377747 DOI: 10.3892/ijo.2020.5058] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/15/2020] [Indexed: 11/05/2022] Open
Abstract
Poor prognosis associated with the dysregulated expression of activin A in a number of malignancies has been related to with numerous aspects of tumorigenesis, including angiogenesis. The present study investigated the prognostic significance of activin A immunoexpression in blood vessels and cancer cells in a number of oral squamous cell carcinoma (OSCC) cases and applied in vitro strategies to determine the impact of activin A on angiogenesis. In a cohort of 95 patients with OSCC, immunoexpression of activin A in both blood vessels and tumor cells was quantified and the association with clinicopathological parameters and survival was analyzed. Effects of activin A on the tube formation, proliferation and migration of human umbilical vein endothelial cells (HUVECs) were evaluated in gain‑of‑function (treatment with recombinant activin A) or loss‑of‑function [treatment with activin A‑antagonist follistatin or by stable transfection with short hairpin RNA (shRNA) targeting activin A] conditions. Conditioned medium from an OSCC cell line with shRNA‑mediated depletion of activin A was also tested. The profile of pro‑ and anti‑angiogenic factors regulated by activin A was assessed with a human angiogenesis quantitative PCR (qPCR) array. Vascular endothelial growth factor A (VEGFA) and its major isoforms were evaluated by reverse transcription‑qPCR and ELISA. Activin A expression in blood vessels demonstrated an independent prognostic value in the multivariate analysis with a hazard ratio of 2.47 [95% confidence interval (CI), 1.30‑4.71; P=0.006) for disease‑specific survival and 2.09 (95% CI, 1.07‑4.08l: P=0.03) for disease‑free survival. Activin A significantly increased tubular formation of HUVECs concomitantly with an increase in proliferation. This effect was validated by reduced proliferation and tubular formation of HUVECs following inhibition of activin A by follistatin or shRNA, as well as by treatment of HUVECs with conditioned medium from activin A‑depleted OSCC cells. Activin A‑knockdown increased the migration of HUVECs. In addition, activin A stimulated the phosphorylation of SMAD2/3 and the expression and production of total VEGFA, significantly enhancing the expression of its pro‑angiogenic isoform 121. The present findings suggest that activin A is a predictor of the prognosis of patients with OSCC, and provide evidence that activin A, in an autocrine and paracrine manner, may contribute to OSCC angiogenesis through differential expression of the isoform 121 of VEGFA.
Collapse
Affiliation(s)
| | - Maurício Rocha Dourado
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP 13414‑018, Brazil
| | - Íris Sawazaki-Calone
- Department of Oral Pathology and Oral Medicine, Dentistry School, Western Paraná State University, Cascavel, PR 85819‑170, Brazil
| | - Marcell Costa De Medeiros
- Departament of Diagnosis and Surgery, School of Dentistry at Araraquara, Araraquara, SP 14801‑385, Brazil
| | - Carlos Rossa Júnior
- Departament of Diagnosis and Surgery, School of Dentistry at Araraquara, Araraquara, SP 14801‑385, Brazil
| | | | - Jorge Esquiche León
- Departament of Stomatology, Public Oral Health and Forensic Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040‑904, Brazil
| | - Daniel Lambert
- Integrated Biosciences, School of Clinical Dentistry and Sheffield Cancer Centre, University of Sheffield, Sheffield S10 2TG, UK
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
| | - Edgard Graner
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP 13414‑018, Brazil
| | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP 13414‑018, Brazil
| |
Collapse
|
38
|
Tissue Engineering and Regenerative Medicine in Craniofacial Reconstruction and Facial Aesthetics. J Craniofac Surg 2020; 31:15-27. [PMID: 31369496 DOI: 10.1097/scs.0000000000005840] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The craniofacial region is anatomically complex and is of critical functional and cosmetic importance, making reconstruction challenging. The limitations of current surgical options highlight the importance of developing new strategies to restore the form, function, and esthetics of missing or damaged soft tissue and skeletal tissue in the face and cranium. Regenerative medicine (RM) is an expanding field which combines the principles of tissue engineering (TE) and self-healing in the regeneration of cells, tissues, and organs, to restore their impaired function. RM offers many advantages over current treatments as tissue can be engineered for specific defects, using an unlimited supply of bioengineered resources, and does not require immunosuppression. In the craniofacial region, TE and RM are being increasingly used in preclinical and clinical studies to reconstruct bone, cartilage, soft tissue, nerves, and blood vessels. This review outlines the current progress that has been made toward the engineering of these tissues for craniofacial reconstruction and facial esthetics.
Collapse
|
39
|
Page DJ, Clarkin CE, Mani R, Khan NA, Dawson JI, Evans ND. Injectable nanoclay gels for angiogenesis. Acta Biomater 2019; 100:378-387. [PMID: 31541735 DOI: 10.1016/j.actbio.2019.09.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 01/16/2023]
Abstract
The retention and sustained activity of therapeutic proteins at delivery sites are goals of regenerative medicine. Vascular endothelial growth factor (VEGF) has significant potential in promoting the growth and regeneration of blood vessels but is intrinsically labile. This is exacerbated by the inflammatory microenvironments at sites requiring regeneration. For VEGF to be efficacious, it may require a carrier that stabilises it, protects it from degradation and retains it at the site of interest. In this study, we tested the hypothesis that injectable nanoclay gels comprising Laponite™ XLG (a synthetic hectorite clay) can stabilise VEGF and retain it in the active form for therapeutic delivery. To achieve this, VEGF was incorporated in Laponite gels and its activity tested at a range of concentrations using in vitro cell culture tubulogenesis assays and in vivo angiogenesis assays. We found that VEGF-Laponite gels enhanced tubulogenesis in a dose-dependent manner in vitro. When administered subcutaneously in vivo, Laponite was retained at the injection site for up to a period of three weeks and promoted a 4-fold increase in blood vessel formation compared with that of alginate or vehicle controls as confirmed by CD31 staining. Notably, as compared to alginate, Laponite gels did not release VEGF, indicating a strong interaction between the growth factor and the nanoclay and suggesting that Laponite enhancement of VEGF efficacy is due to its retention at the implantation site for a prolonged period. Our approach provides a robust method for the delivery of bioactive recombinant VEGF without the necessity for complex hydrogel or protein engineering. STATEMENT OF SIGNIFICANCE: In medicine, it is important to deliver drugs to a particular location in the body. Often, however, the drugs are quickly broken down and carried away in the blood before they can exert their effect. In this study, we used a type of synthetic clay, called Laponite™, to preserve a molecule, named VEGF, that stimulates the growth of blood vessels. Previously, we have been able to bind VEGF to the surface of clays, but the clay is not effective when injected or applied as a gel. Herein, we show that we can mix VEGF with the clay and that it strongly stimulates blood vessel growth. We speculate that this would be a useful material for skin wound healing.
Collapse
|
40
|
Roberts IV, Bukhary D, Valdivieso CYL, Tirelli N. Fibrin Matrices as (Injectable) Biomaterials: Formation, Clinical Use, and Molecular Engineering. Macromol Biosci 2019; 20:e1900283. [PMID: 31769933 DOI: 10.1002/mabi.201900283] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/14/2019] [Indexed: 12/19/2022]
Abstract
This review focuses on fibrin, starting from biological mechanisms (its production from fibrinogen and its enzymatic degradation), through its use as a medical device and as a biomaterial, and finally discussing the techniques used to add biological functions and/or improve its mechanical performance through its molecular engineering. Fibrin is a material of biological (human, and even patient's own) origin, injectable, adhesive, and remodellable by cells; further, it is nature's most common choice for an in situ forming, provisional matrix. Its widespread use in the clinic and in research is therefore completely unsurprising. There are, however, areas where its biomedical performance can be improved, namely achieving a better control over mechanical properties (and possibly higher modulus), slowing down degradation or incorporating cell-instructive functions (e.g., controlled delivery of growth factors). The authors here specifically review the efforts made in the last 20 years to achieve these aims via biomimetic reactions or self-assembly, as much via formation of hybrid materials.
Collapse
Affiliation(s)
- Iwan Vaughan Roberts
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Deena Bukhary
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.,Department of Pharmaceutical Science, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | | | - Nicola Tirelli
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.,Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, via Morego 30, 16163, Genova, Italy
| |
Collapse
|
41
|
|
42
|
Denatured acellular dermal matrix seeded with bone marrow mesenchymal stem cells for wound healing in mice. Burns 2019; 45:1685-1694. [DOI: 10.1016/j.burns.2019.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/02/2019] [Accepted: 04/16/2019] [Indexed: 12/15/2022]
|
43
|
Ullah I, Abu-Dawud R, Busch JF, Rabien A, Erguen B, Fischer I, Reinke P, Kurtz A. VEGF – Supplemented extracellular matrix is sufficient to induce endothelial differentiation of human iPSC. Biomaterials 2019; 216:119283. [DOI: 10.1016/j.biomaterials.2019.119283] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 01/13/2023]
|
44
|
Pensa NW, Curry AS, Reddy MS, Bellis SL. Sustained delivery of the angiogenic QK peptide through the use of polyglutamate domains to control peptide release from bone graft materials. J Biomed Mater Res A 2019; 107:2764-2773. [DOI: 10.1002/jbm.a.36779] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 01/19/2023]
Affiliation(s)
- Nicholas W. Pensa
- Department of Biomedical EngineeringUniversity of Alabama at Birmingham Birmingham Alabama
| | - Andrew S. Curry
- Department of Biomedical EngineeringUniversity of Alabama at Birmingham Birmingham Alabama
| | - Michael S. Reddy
- School of DentistryUniversity of California San Francisco California
| | - Susan L. Bellis
- Department of Cell, Developmental, and Integrative BiologyUniversity of Alabama at Birmingham Birmingham Alabama
| |
Collapse
|
45
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
46
|
Engineered biomaterials to mitigate growth factor cost in cell biomanufacturing. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2018.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
47
|
In Vitro Characterization of Hypoxia Preconditioned Serum (HPS)-Fibrin Hydrogels: Basis for an Injectable Biomimetic Tissue Regeneration Therapy. J Funct Biomater 2019; 10:jfb10020022. [PMID: 31086048 PMCID: PMC6616457 DOI: 10.3390/jfb10020022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 01/03/2023] Open
Abstract
Blood-derived growth factor preparations have long been employed to improve perfusion and aid tissue repair. Among these, platelet-rich plasma (PRP)-based therapies have seen the widest application, albeit with mixed clinical results to date. Hypoxia-preconditioned blood products present an alternative to PRP, by comprising the complete wound healing factor-cascade, i.e., hypoxia-induced peripheral blood cell signaling, in addition to platelet-derived factors. This study set out to characterize the preparation of hypoxia preconditioned serum (HPS), and assess the utility of HPS–fibrin hydrogels as vehicles for controlled factor delivery. Our findings demonstrate the positive influence of hypoxic incubation on HPS angiogenic potential, and the individual variability of HPS angiogenic factor concentration. HPS–fibrin hydrogels can rapidly retain HPS factor proteins and gradually release them over time, while both functions appear to depend on the fibrin matrix mass. This offers a means of controlling factor retention/release, through adjustment of HPS fibrinogen concentration, thus allowing modulation of cellular angiogenic responses in a growth factor dose-dependent manner. This study provides the first evidence that HPS–fibrin hydrogels could constitute a new generation of autologous/bioactive injectable compositions that provide biochemical and biomaterial signals analogous to those mediating physiological wound healing. This therefore establishes a rational foundation for their application towards biomimetic tissue regeneration.
Collapse
|
48
|
Lin Y, Huang S, Zou R, Gao X, Ruan J, Weir MD, Reynolds MA, Qin W, Chang X, Fu H, Xu HHK. Calcium phosphate cement scaffold with stem cell co-culture and prevascularization for dental and craniofacial bone tissue engineering. Dent Mater 2019; 35:1031-1041. [PMID: 31076156 DOI: 10.1016/j.dental.2019.04.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/17/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Calcium phosphate cements (CPCs) mimic nanostructured bone minerals and are promising for dental, craniofacial and orthopedic applications. Vascularization plays a critical role in bone regeneration. This article represents the first review on cutting-edge research on prevascularization of CPC scaffolds to enhance bone regeneration. METHODS This article first presented the prevascularization of CPC scaffolds. Then the co-culture of two cell types in CPC scaffolds was discussed. Subsequently, to further enhance the prevascularization efficacy, tri-culture of three different cell types in CPC scaffolds was presented. RESULTS (1) Arg-Gly-Asp (RGD) incorporation in CPC bone cement scaffold greatly enhanced cell affinity and bone prevascularization; (2) By introducing endothelial cells into the culture of osteogenic cells (co-culture of two different cell types, or bi-culture) in CPC scaffold, the bone defect area underwent much better angiogenic and osteogenic processes when compared to mono-culture; (3) Tri-culture with an additional cell type of perivascular cells (such as pericytes) resulted in a substantially enhanced prevascularization of CPC scaffolds in vitro and more new bone and blood vessels in vivo, compared to bi-culture. Furthermore, biological cell crosstalk and capillary-like structure formation made critical contributions to the bi-culture system. In addition, the pericytes in the tri-culture system substantially promoted stability and maturation of the primary vascular network. SIGNIFICANCE The novel approach of CPC scaffolds with stem cell bi-culture and tri-culture is of great significance in the regeneration of dental, craniofacial and orthopedic defects in clinical practice.
Collapse
Affiliation(s)
- Ying Lin
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Shuheng Huang
- Department of Endodontics, Guanghua School and Hospital of Stomatology & Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510055, China
| | - Rui Zou
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 510182, China
| | - Xianling Gao
- Department of Endodontics, Guanghua School and Hospital of Stomatology & Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510055, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Jianping Ruan
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Michael D Weir
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Wei Qin
- Department of Endodontics, Guanghua School and Hospital of Stomatology & Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510055, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Xiaofeng Chang
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Haijun Fu
- Department of Endodontics, Guanghua School and Hospital of Stomatology & Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510055, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
49
|
Vardar E, Vythilingam G, Pinnagoda K, Engelhardt EM, Zambelli PY, Hubbell JA, Lutolf MP, Frey P, Larsson HM. A bioactive injectable bulking material; a potential therapeutic approach for stress urinary incontinence. Biomaterials 2019; 206:41-48. [PMID: 30925287 DOI: 10.1016/j.biomaterials.2019.03.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Stress urinary incontinence (SUI) is a life changing condition, affecting 20 million women worldwide. In this study, we developed a bioactive, injectable bulking agent that consists of Permacol™ (Medtronic, Switzerland) and recombinant insulin like growth factor-1 conjugated fibrin micro-beads (fib_rIGF-1) for its bulk stability and capacity to induce muscle regeneration. Therefore, Permacol™ formulations were injected in the submucosal space of rabbit bladders. The ability of a bulking material to form a stable and muscle-inducing bulk represents for us a promising therapeutic approach to achieve a long-lasting treatment for SUI. The fib_rIGF-1 showed no adverse effect on human smooth muscle cell metabolic activity and viability in vitro based on AlamarBlue assays and Live/Dead staining. Three months after injection of fib_rIGF-1 together with Permacol™ into the rabbit bladder wall, we observed a smooth muscle tissue like formation within the injected materials. Positive staining for alpha smooth muscle actin, calponin, and caldesmon demonstrated a contractile phenotype of the newly formed smooth muscle tissue. Moreover, the fib_rIGF-1 treated group also improved the neovascularization at the injection site, confirmed by CD31 positive staining compared to bulks made of PermacolTM only. The results of this study encourage us to further develop this injectable, bioactive bulking material towards a future therapeutic approach for a minimal invasive and long-lasting treatment of SUI.
Collapse
Affiliation(s)
- E Vardar
- Experimental Pediatric Urology, Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland; Department of Pediatrics, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - G Vythilingam
- Experimental Pediatric Urology, Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland; Department of Surgery, University of Malaya, Kuala Lumpur, Malaysia
| | - K Pinnagoda
- Department of Pediatrics, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - E M Engelhardt
- Experimental Pediatric Urology, Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - P Y Zambelli
- Department of Pediatrics, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - J A Hubbell
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - M P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland
| | - P Frey
- Experimental Pediatric Urology, Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - H M Larsson
- Experimental Pediatric Urology, Laboratory for Regenerative Medicine and Pharmacobiology, Institute for Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland; Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland.
| |
Collapse
|
50
|
Farina M, Alexander JF, Thekkedath U, Ferrari M, Grattoni A. Cell encapsulation: Overcoming barriers in cell transplantation in diabetes and beyond. Adv Drug Deliv Rev 2019; 139:92-115. [PMID: 29719210 DOI: 10.1016/j.addr.2018.04.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/19/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Cell-based therapy is emerging as a promising strategy for treating a wide range of human diseases, such as diabetes, blood disorders, acute liver failure, spinal cord injury, and several types of cancer. Pancreatic islets, blood cells, hepatocytes, and stem cells are among the many cell types currently used for this strategy. The encapsulation of these "therapeutic" cells is under intense investigation to not only prevent immune rejection but also provide a controlled and supportive environment so they can function effectively. Some of the advanced encapsulation systems provide active agents to the cells and enable a complete retrieval of the graft in the case of an adverse body reaction. Here, we review various encapsulation strategies developed in academic and industrial settings, including the state-of-the-art technologies in advanced preclinical phases as well as those undergoing clinical trials, and assess their advantages and challenges. We also emphasize the importance of stimulus-responsive encapsulated cell systems that provide a "smart and live" therapeutic delivery to overcome barriers in cell transplantation as well as their use in patients.
Collapse
|