1
|
Burnham AJ, Toma AI, Shah D, Cha T, Kaimari S, Behara M, Sekar KPC, Kamalakar A, Willett N, Botchwey E, Goudy SL. FTY720P-treated macrophages in PEG-4MAL hydrogels promote oral wound healing. Cytotherapy 2025; 27:338-349. [PMID: 39665738 PMCID: PMC11810592 DOI: 10.1016/j.jcyt.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/21/2024] [Accepted: 11/03/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND AIMS Oral wound healing involves hemostasis, inflammation, proliferation and tissue remodeling. The oral cavity is a complex wound healing environment because of the presence of saliva, a high bacterial burden and ongoing physical trauma from eating. The inflammatory component of wound healing balances the polarization of macrophages in healing tissues between M1 inflammatory macrophages and M2 anti-inflammatory macrophages. M2 macrophages secrete anti-inflammatory and pro-regenerative cytokines and chemokines, which aid wound healing. Fingolimod, or FTY720, a Food and Drug Administration-approved sphingosine-1-phosphate modulator, has been implicated in inducing the polarization of macrophages to the M2 phenotype. In this study, we investigated whether macrophage pre-treatment with phosphorylated FTY720 (FTY720P), the bioactive form of the drug, in a PEG-4MAL hydrogel promotes improved oral wound healing in a critically sized oral mucosal defect model. METHODS AND RESULTS Using cytokine dot blots and Luminex cytokine assays (MilliporeSigma, Burlington, MA, USA), FTY720P-treated murine RAW 264.7 and human THP1-differentiated macrophages in PEG-4MAL hydrogels secreted chemokines and cytokines known to regulate inflammation (e.g., interleukin [IL] 4, IL-13) and induce M2 macrophage polarization (e.g., CCL6, CCL22), leukocyte migration (e.g., CXCL2, CCL2, CCL12, CCL22), angiogenesis (e.g., vascular endothelial growth factor) and epithelialization (e.g., IL-1, IL-17, IL-22). In vitro, FTY720P-treated cells induced chemotaxis of macrophages and fibroblasts in Transwell assays (Corning, Corning, NY, USA) and oral epithelial scratch wound closure. In a murine oronasal fistula (ONF) model of oral wound healing, the local application of FTY720P-treated macrophages in PEG-4MAL hydrogels significantly increased wound closure (75% closure) relative to non-treated cells (40% closure) and blank hydrogel controls (25% closure) (P < 0.0001). Flow cytometry of mouse palatal tissue showed that application of FTY720P-treated macrophage hydrogels to ONFs significantly increased day 7 percentage of M1 and M2 macrophages, mesenchymal stromal cells and CD19+ B cells. Significantly fewer neutrophils, monocytes, CD4+/CD8+ T cells and endothelial cells were observed in the FTY720P-treated macrophage defects, suggesting that FTY720P-treated macrophages in hydrogels promote oral wound healing via suppression of granulation and resolution of inflammation and promotion of tissue maturation. CONCLUSIONS Our data provide new insights into the use of potential FTY720P-treated macrophage therapies for oral wound healing and have clinical implications for cleft palate and oral surgery.
Collapse
Affiliation(s)
- Andre J Burnham
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA; Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Afra I Toma
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Daniel Shah
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Tim Cha
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sundus Kaimari
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Monica Behara
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | | - Archana Kamalakar
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nick Willett
- Department of Bioengineering, University of Oregon, Eugene, Oregon, USA
| | - Edward Botchwey
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Steven L Goudy
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; Department of Otolaryngology, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.
| |
Collapse
|
2
|
Mouzoura P, Marazioti A, Gkartziou F, Metsiou DN, Antimisiaris SG. Potential of Liposomal FTY720 for Bone Regeneration: Proliferative, Osteoinductive, Chemoattractive, and Angiogenic Properties Compared to Free Bioactive Lipid. Int J Nanomedicine 2025; 20:239-265. [PMID: 39802384 PMCID: PMC11724662 DOI: 10.2147/ijn.s494512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction FTY720 bioactive lipid has proliferative, osteoinductive, chemo attractive, and angiogenic properties, being thus a potential exogenous administered agent for promotion of bone regeneration. Herein we developed FTY720-loaded liposomes as a potential delivery system that could retain and prolong the bioactivity of the bioactive lipid and at the same time reduce its cytotoxicity (at high doses). Methods FTY720 liposomes were prepared by thin-lipid hydration and microfluidic flow focusing, and evaluated for their ability to induce proliferation, osteoinduction, and chemoattraction in three cell types: MC3T3-E1 pre-osteoblast cells, L929 fibroblast cells, and ATDC5 chondrogenic cells. The angiogenic activity of free and liposomal FTY720 was investigated using a chick chorioallantoic membrane assay. NBD-FTY720 cellular uptake was quantitated using flow cytometry and morphologically assessed by confocal microscopy. Implicated cellular signaling mechanisms were investigated by quantifying phosphorylated MAPK and CREB proteins. Results FTY720 liposomes (~80-110 nm) with low polydispersity and ~100% loading were prepared using both methods. FTY720 demonstrated the ability to increase cell proliferation at 10-300nM doses but was cytotoxic at doses>400nM while the corresponding liposomal-FTY720 doses were non-cytotoxic, proving its reduced toxicity. In several cases (cells and doses), FTY720 liposomes demonstrated increased osteogenic differentiation of cells, proliferation, and migration compared to free FTY720, whereas both FTY720 forms demonstrated substantial angiogenic activity. Liposomal FTY720 cellular uptake was substantially higher than that of free FTY720 in some cases, a fact that may be connected to its higher bioactivity. Increased phosphorylated MAPK and CREB protein concentrations provided information about the potential cellular signaling mechanisms involved in FTY720-induced osteogenesis. Discussion The current results confirm the high potential of FTY720 bioactive lipid, especially in its liposomal form, that demonstrated substantial reduction of cytotoxicity and prolonged preservation of the lipids bioactivity (compared to the free lipid), for accelerated treatment of bone defects. Interestingly, the current studies prove the potential of FTY720, especially in its liposomal form, to promote reprogramming of L929 fibroblasts into osteoblasts, a novel finding deserving future exploitation.
Collapse
Affiliation(s)
- Panagiota Mouzoura
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
| | - Antonia Marazioti
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
- Laboratory of Basic Sciences, Department of Physiotherapy, University of the Peloponnese, Sparti, 23100, Greece
| | - Foteini Gkartziou
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
| | - Despoina-Nektaria Metsiou
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
| | - Sophia G Antimisiaris
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
- FORTH/ICE‑ΗΤ, Institute of Chemical Engineering Sciences, Platani, 26504, Greece
| |
Collapse
|
3
|
Behara M, Goudy S. FTY720 in immuno-regenerative and wound healing technologies for muscle, epithelial and bone regeneration. Front Physiol 2023; 14:1148932. [PMID: 37250137 PMCID: PMC10213316 DOI: 10.3389/fphys.2023.1148932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
In 2010, the FDA approved the administration of FTY720, S1P lipid mediator, as a therapy to treat relapsing forms of multiple sclerosis. FTY720 was found to sequester pro-inflammatory lymphocytes within the lymph node, preventing them from causing injury to the central nervous system due to inflammation. Studies harnessing the anti-inflammatory properties of FTY720 as a pro-regenerative strategy in wound healing of muscle, bone and mucosal injuries are currently being performed. This in-depth review discusses the current regenerative impact of FTY720 due to its anti-inflammatory effect stratified into an assessment of wound regeneration in the muscular, skeletal, and epithelial systems. The regenerative effect of FTY720 in vivo was characterized in three animal models, with differing delivery mechanisms emerging in the last 20 years. In these studies, local delivery of FTY720 was found to increase pro-regenerative immune cell phenotypes (neutrophils, macrophages, monocytes), vascularization, cell proliferation and collagen deposition. Delivery of FTY720 to a localized wound environment demonstrated increased bone, muscle, and mucosal regeneration through changes in gene and cytokine production primarily by controlling the local immune cell phenotypes. These changes in gene and cytokine production reduced the inflammatory component of wound healing and increased the migration of pro-regenerative cells (neutrophils and macrophages) to the wound site. The application of FTY720 delivery using a biomaterial has demonstrated the ability of local delivery of FTY720 to promote local wound healing leveraging an immunomodulatory mechanism.
Collapse
Affiliation(s)
- Monica Behara
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Steven Goudy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- Department of Otolaryngology, Emory University, Atlanta, GA, United States
| |
Collapse
|
4
|
Yang C, Liu Y, Wang Z, Lin M, Liu C. Controlled mechanical loading improves bone regeneration by regulating type H vessels in a S1Pr1-dependent manner. FASEB J 2022; 36:e22530. [PMID: 36063128 DOI: 10.1096/fj.202200339rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair and result in delayed unions or non-unions. Dynamic mechanical stimulation promotes bone formation, when applied at the correct time frame, with optimal loading magnitude, frequency, and repetition. Controlled mechanical loading significantly increases osteogenic cells during the matrix deposition phase of bone repair. In the bone defect, the blood vessel network guides the initial bone formation activities. A unique blood vessel subtype (Type H) exists in bone, which expresses high levels of CD31 and endomucin, and functions to couple angiogenesis and osteogenesis. However, how this form of controlled mechanical loading regulates the Type H vessels and promotes bone formation is still not clear. Sphingosine 1-phosphate (S1P) participates in the bone anabolic process and is a key regulator of the blood vessel. Its receptor, sphingosine 1-phosphate receptor 1 (S1Pr1), is a mechanosensitive protein that regulates vascular integrity. Therefore, we hypothesis that controlled anabolic mechanical loading promotes bone repair by acting on Type H vessels. To study the effect of S1Pr1 on loading induced-bone repair, we utilized a stabilized tibial defect model, which allows for the application of anabolic mechanical loading. Mechanical loading upregulated S1Pr1 within the entire defect, with up to 80% expressed in blood vessels, as observed by deep tissue imaging. Additionally, S1Pr1 antagonism by W146 inhibited the anabolic effects of mechanical loading. We showed that mechanical loading or activating S1Pr1 could induce YAP nuclear translocation, a key regulator in the cell's mechanical response, in endothelial cells (ECs) in vitro. Inhibition of S1Pr1 in endothelial cells by siRNA reduced loading-induced YAP nuclear translocation and expressions of angiogenic genes. In vivo, YAP nuclear translocation in Type H vessels was up-regulated after mechanical loading but was inhibited by antagonizing S1Pr1. S1Pr1 agonist, FTY720, increased bone volume and Type H vessel volume, similar to that of mechanical stimulation. In conclusion, controlled anabolic mechanical loading enhanced bone formation mainly through Type H vessels in a S1Pr1-dependent manner.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
5
|
Tabanez AP, de Campos Soriani Azevedo M, Melchiades JL, Fonseca AC, Francisconi CF, Colavite PM, Biguetti CC, de Oliveira Rodini Pegoraro C, Trombone APF, Garlet GP. FTY720 administration results in a M2 associated immunoregulatory effect that positively influences the outcome of alveolar bone repair outcome in mice. Bone 2022; 163:116506. [PMID: 35902072 DOI: 10.1016/j.bone.2022.116506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/02/2022]
Abstract
The alveolar bone repair process may be influenced by multiple local and systemic factors, which include immune system cells and mediators. Macrophages allegedly play important roles in the repair process, and the transition of an initial inflammatory M1 profile into a pro-reparative M2 profile theoretically contributes to a favorable repair outcome. In this context, considering immunoregulatory molecules as potential targets for improving bone repair, this study evaluated the role of the immunoregulatory molecule FTY720, previously described to favor the development of the M2 phenotype, in the process of alveolar bone healing in C57Bl/6 (WT) mice. Experimental groups submitted to tooth extraction and maintained under control conditions or treated with FTY720 were evaluated by microtomographic (μCT), histomorphometric, immunohistochemical and molecular analysis to characterize healing and host response features at 0, 1, 3, 7 and 14 days. Our results demonstrated that the FTY720 group presented higher bone tissue density, higher bone tissue volume, greater tissue volume fraction, greater number and thickness of trabeculae and a higher number of osteoblasts and osteoclasts than the control group. Accordingly, the bone markers BMP2, BMP7, ALPL, SOST and RANK mRNA expressions increased in the FTY720 treated group. Furthermore, the levels of FIZZ, ARG2 and IL-10 mRNA increased in the FTY720 group together with the presence of CD206+ cells, suggesting that the boost of bone formation mediated by FTY720 involves an increased polarization and activity of M2 macrophages in healing sites. Thus, our results demonstrate that FTY720 favored the process of alveolar bone repair, probably trough a strengthened M2 response, associated with an increased expression of markers osteogenic differentiation and activity markers. Immunoregulatory strategies based in the modulation of macrophage polarization profile can comprise effective tools to improve the bone repair process.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Cláudia Cristina Biguetti
- School of Dentistry of Bauru, University of São Paulo (FOB/USP), Bauru, SP, Brazil; Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States of America
| | | | | | | |
Collapse
|
6
|
Khayambashi P, Iyer J, Pillai S, Upadhyay A, Zhang Y, Tran SD. Hydrogel Encapsulation of Mesenchymal Stem Cells and Their Derived Exosomes for Tissue Engineering. Int J Mol Sci 2021; 22:E684. [PMID: 33445616 PMCID: PMC7827932 DOI: 10.3390/ijms22020684] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
Tissue engineering has been an inveterate area in the field of regenerative medicine for several decades. However, there remains limitations to engineer and regenerate tissues. Targeted therapies using cell-encapsulated hydrogels, such as mesenchymal stem cells (MSCs), are capable of reducing inflammation and increasing the regenerative potential in several tissues. In addition, the use of MSC-derived nano-scale secretions (i.e., exosomes) has been promising. Exosomes originate from the multivesicular division of cells and have high therapeutic potential, yet neither self-replicate nor cause auto-immune reactions to the host. To maintain their biological activity and allow a controlled release, these paracrine factors can be encapsulated in biomaterials. Among the different types of biomaterials in which exosome infusion is exploited, hydrogels have proven to be the most user-friendly, economical, and accessible material. In this paper, we highlight the importance of MSCs and MSC-derived exosomes in tissue engineering and the different biomaterial strategies used in fabricating exosome-based biomaterials, to facilitate hard and soft tissue engineering.
Collapse
Affiliation(s)
| | | | | | | | | | - Simon D. Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada; (P.K.); (J.I.); (S.P.); (A.U.); (Y.Z.)
| |
Collapse
|
7
|
Sok MCP, Baker N, McClain C, Lim HS, Turner T, Hymel L, Ogle M, Olingy C, Palacios JI, Garcia JR, Srithar K, García AJ, Qiu P, Botchwey EA. Dual delivery of IL-10 and AT-RvD1 from PEG hydrogels polarize immune cells towards pro-regenerative phenotypes. Biomaterials 2021; 268:120475. [PMID: 33321293 PMCID: PMC11129952 DOI: 10.1016/j.biomaterials.2020.120475] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 09/29/2020] [Accepted: 10/18/2020] [Indexed: 02/06/2023]
Abstract
Inflammation after traumatic injury or surgical intervention is both a protective tissue response leading to regeneration and a potential cause of wound complications. One potentially successful strategy to harness to pro-regenerative roles of host inflammation is the localized delivery of bioactive materials to induce immune suppressive cellular responses by cells responding to injury. In this study, we designed a fully synthetic poly (ethylene) glycol (PEG)-based hydrogel to release the specialized pro-resolving lipid mediator aspirin-triggered resolvin-D1 (AT-RvD1) and recombinant human interleukin 10 (IL-10). We utilized a unique side-by-side internally controlled implant design wherein bioactive hydrogels were implanted adjacent to control hydrogels devoid of immune modulatory factors in the dorsal skinfold window chamber. We also explored single-immune cell data with unsupervised approaches such as SPADE. First, we show that RGD-presenting hydrogel delivery results in enhanced immune cell recruitment to the site of injury. We then use intra-vital imaging to assess cellular recruitment and microvascular remodeling to show an increase in the caliber and density of local microvessels. Finally, we show that the recruitment and re-education of mononuclear phagocytes by combined delivery IL-10 and AT-RvD1 localizes immune suppressive subsets to the hydrogel, including CD206+ macrophages (M2a/c) and IL-10 expressing dendritic cells in the context of chronic inflammation following surgical tissue disruption. These data demonstrate the potential of combined delivery on the recruitment of regenerative cell subsets involved in wound healing complications.
Collapse
Affiliation(s)
- Mary Caitlin P Sok
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Emory University Medical Scientist Training Program, USA
| | - Nusaiba Baker
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Emory University Medical Scientist Training Program, USA
| | - Claire McClain
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hong Seo Lim
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Thomas Turner
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Lauren Hymel
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Molly Ogle
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Claire Olingy
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Joshua I Palacios
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - José R Garcia
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Krithik Srithar
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Peng Qiu
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Edward A Botchwey
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
8
|
Sartawi Z, Waeber C, Schipani E, Ryan KB. Development of electrospun polymer scaffolds for the localized and controlled delivery of siponimod for the management of critical bone defects. Int J Pharm 2020; 590:119956. [DOI: 10.1016/j.ijpharm.2020.119956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 12/20/2022]
|
9
|
Menger MM, Laschke MW, Orth M, Pohlemann T, Menger MD, Histing T. Vascularization Strategies in the Prevention of Nonunion Formation. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:107-132. [PMID: 32635857 DOI: 10.1089/ten.teb.2020.0111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Delayed healing and nonunion formation are major challenges in orthopedic surgery, which require the development of novel treatment strategies. Vascularization is considered one of the major prerequisites for successful bone healing, providing an adequate nutrient supply and allowing the infiltration of progenitor cells to the fracture site. Hence, during the last decade, a considerable number of studies have focused on the evaluation of vascularization strategies to prevent or to treat nonunion formation. These involve (1) biophysical applications, (2) systemic pharmacological interventions, and (3) tissue engineering, including sophisticated scaffold materials, local growth factor delivery systems, cell-based techniques, and surgical vascularization approaches. Accumulating evidence indicates that in nonunions, these strategies are indeed capable of improving the process of bone healing. The major challenge for the future will now be the translation of these strategies into clinical practice to make them accessible for the majority of patients. If this succeeds, these vascularization strategies may markedly reduce the incidence of nonunion formation. Impact statement Delayed healing and nonunion formation are a major clinical problem in orthopedic surgery. This review provides an overview of vascularization strategies for the prevention and treatment of nonunions. The successful translation of these strategies in clinical practice is of major importance to achieve adequate bone healing.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Marcel Orth
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Tina Histing
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
10
|
Bone regenerative potential of the selective sphingosine 1-phosphate receptor modulator siponimod: In vitro characterisation using osteoblast and endothelial cells. Eur J Pharmacol 2020; 882:173262. [PMID: 32534075 DOI: 10.1016/j.ejphar.2020.173262] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/31/2022]
Abstract
The repair of critical bone defects remains a significant therapeutic challenge. While the implantation of drug-eluting scaffolds is an option, a drug with the optimal pharmacological properties has not yet been identified. Agents acting at sphingosine 1-phosphate (S1P) receptors have been considered, but those investigated so far do not discriminate between the five known S1P receptors. This work was undertaken to investigate the potential of the specific S1P1/5 modulator siponimod as a bone regenerative agent, by testing in vitro its effect on cell types critical to the bone regeneration process. hFOB osteoblasts and HUVEC endothelial cells were treated with siponimod and other S1P receptor modulators and investigated for changes in intracellular cyclic AMP content, viability, proliferation, differentiation, attachment and cellular motility. Siponimod showed no effect on the viability and proliferation of osteoblasts and endothelial cells, but increased osteoblast differentiation (as shown by increased alkaline phosphatase activity). Furthermore, siponimod significantly increased endothelial cell motility in scratch and transwell migration assays. These effects on osteoblast differentiation and endothelial cell migration suggest that siponimod may be a potential agent for the stimulation of localised differentiation of osteoblasts in critical bone defects.
Collapse
|
11
|
Ranmuthu CDS, Ranmuthu CKI, Russell JC, Singhania D, Khan WS. Evaluating the Effect of Non-cellular Bioactive Glass-Containing Scaffolds on Osteogenesis and Angiogenesis in in vivo Animal Bone Defect Models. Front Bioeng Biotechnol 2020; 8:430. [PMID: 32478053 PMCID: PMC7240009 DOI: 10.3389/fbioe.2020.00430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
The use of bone scaffolds to replace injured or diseased bone has many advantages over the currently used autologous and allogeneic options in clinical practice. This systematic review evaluates the current evidence for non-cellular scaffolds containing bioactive glass on osteogenesis and angiogenesis in animal bone defect models. Studies that reported results of osteogenesis via micro-CT and results of angiogenesis via Microfil perfusion or immunohistochemistry were included in the review. A literature search of PubMed, EMBASE and Scopus was carried out in November 2019 from which nine studies met the inclusion and exclusion criteria. Despite the significant heterogeneity in the composition of the scaffolds used in each study, it could be concluded that scaffolds containing bioactive glass improve bone regeneration in these models, both by osteogenic and angiogenic measures. Incorporation of additional elements into the glass network, using additives, and using biochemical factors generally had a beneficial effect. Comparing the different compositions of non-cellular bioactive glass containing scaffolds is however difficult due to the heterogeneity in bioactive glass compositions, fabrication methods and biochemical additives used.
Collapse
Affiliation(s)
| | | | - Jodie C. Russell
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Disha Singhania
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Wasim S. Khan
- Division of Trauma and Orthopaedics, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
12
|
Rothe R, Hauser S, Neuber C, Laube M, Schulze S, Rammelt S, Pietzsch J. Adjuvant Drug-Assisted Bone Healing: Advances and Challenges in Drug Delivery Approaches. Pharmaceutics 2020; 12:E428. [PMID: 32384753 PMCID: PMC7284517 DOI: 10.3390/pharmaceutics12050428] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
Bone defects of critical size after compound fractures, infections, or tumor resections are a challenge in treatment. Particularly, this applies to bone defects in patients with impaired bone healing due to frequently occurring metabolic diseases (above all diabetes mellitus and osteoporosis), chronic inflammation, and cancer. Adjuvant therapeutic agents such as recombinant growth factors, lipid mediators, antibiotics, antiphlogistics, and proangiogenics as well as other promising anti-resorptive and anabolic molecules contribute to improving bone healing in these disorders, especially when they are released in a targeted and controlled manner during crucial bone healing phases. In this regard, the development of smart biocompatible and biostable polymers such as implant coatings, scaffolds, or particle-based materials for drug release is crucial. Innovative chemical, physico- and biochemical approaches for controlled tailor-made degradation or the stimulus-responsive release of substances from these materials, and more, are advantageous. In this review, we discuss current developments, progress, but also pitfalls and setbacks of such approaches in supporting or controlling bone healing. The focus is on the critical evaluation of recent preclinical studies investigating different carrier systems, dual- or co-delivery systems as well as triggered- or targeted delivery systems for release of a panoply of drugs.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (S.S.); (S.R.)
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (S.S.); (S.R.)
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, 01307 Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (R.R.); (S.H.); (C.N.); (M.L.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| |
Collapse
|
13
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part II - Modulation of angiogenesis. Clin Hemorheol Microcirc 2020; 73:409-438. [PMID: 31177206 DOI: 10.3233/ch-199103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The treatment of critical-size bone defects following complicated fractures, infections or tumor resections is a major challenge. The same applies to fractures in patients with impaired bone healing due to systemic inflammatory and metabolic diseases. Despite considerable progress in development and establishment of new surgical techniques, design of bone graft substitutes and imaging techniques, these scenarios still represent unresolved clinical problems. However, the development of new active substances offers novel potential solutions for these issues. This work discusses therapeutic approaches that influence angiogenesis or hypoxic situations in healing bone and surrounding tissue. In particular, literature on sphingosine-1-phosphate receptor modulators and nitric oxide (NO•) donors, including bi-functional (hybrid) compounds like NO•-releasing cyclooxygenase-2 inhibitors, was critically reviewed with regard to their local and systemic mode of action.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
14
|
Liu L, Zhao F, Chen X, Luo M, Yang Z, Cao X, Miao G, Chen D, Chen X. Local delivery of FTY720 in mesoporous bioactive glass improves bone regeneration by synergistically immunomodulating osteogenesis and osteoclastogenesis. J Mater Chem B 2020; 8:6148-6158. [DOI: 10.1039/d0tb00982b] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
FTY720@MBGs improve bone regeneration by synergistically immunomodulating osteogenesis and osteoclastogenesis.
Collapse
Affiliation(s)
- Lu Liu
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Fujian Zhao
- Stomatological Hospital
- Southern Medical University
- Guangzhou 510280
- China
| | - Xiaoyu Chen
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Man Luo
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Zhen Yang
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Xiaodong Cao
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| | - Guohou Miao
- Key Laboratory of Oral Medicine
- Guangzhou Institute of Oral Disease
- Stomatology Hospital of Guangzhou Medical University
- Guangzhou 510140
- China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering
- Beijing Laboratory of Biomedical Materials
- Beijing Research Institute of Orthopaedics and Traumatology
- Beijing JiShuiTan Hospital
- Beijing,100035
| | - Xiaofeng Chen
- School of Materials Science and Engineering
- South China University of Technology
- Guangzhou 510641
- China
- National Engineering Research Centre for Tissue Restoration and Reconstruction
| |
Collapse
|
15
|
Li S, Song C, Yang S, Yu W, Zhang W, Zhang G, Xi Z, Lu E. Supercritical CO 2 foamed composite scaffolds incorporating bioactive lipids promote vascularized bone regeneration via Hif-1α upregulation and enhanced type H vessel formation. Acta Biomater 2019; 94:253-267. [PMID: 31154054 DOI: 10.1016/j.actbio.2019.05.066] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/22/2019] [Accepted: 05/26/2019] [Indexed: 01/27/2023]
Abstract
Bone tissue engineering has substantial potential for the treatment of massive bone defects; however, efficient vascularization coupled with bone regeneration still remains a challenge in this field. In the current study, supercritical carbon dioxide (scCO2) foaming technique was adopted to fabricate mesoporous bioactive glasses (MBGs) particle-poly (lactic-co-glycolic acid) (PLGA) composite scaffolds with appropriate mechanical and degradation properties as well as in vitro bioactivity. The MBG-PLGA scaffolds incorporating the bioactive lipid FTY720 (designated as FTY/MBG-PLGA) exhibited simultaneously sustained release of the bioactive lipid and ions. In addition to providing a favorable microenvironment for cellular adhesion and proliferation, FTY/MBG-PLGA scaffolds significantly facilitated the in vitro osteogenic differentiation of rBMSCs and also markedly stimulated the upregulation of Hif-1α expression via the activation of the Erk1/2 pathway, which mediated the osteogenic and pro-angiogenic effects on rBMSCs. Furthermore, FTY/MBG-PLGA extracts induced superior in vitro angiogenic performance of HUVECs. In vivo evaluation of critical-sized rat calvarial bone defects indicated that FTY/MBG-PLGA scaffolds potently promoted vascularized bone regeneration. Notably, the significantly enhanced formation of type H vessels (CD31hiEmcnhi neo-vessels) was observed in newly formed bone tissue in FTY/MBG-PLGA group, strongly suggesting that FTY720 and therapeutic ions released from the scaffolds synergistically induced more type H vessel formation, which indicated the coupling of angiogenesis and osteogenesis to achieve efficiently vascularized bone regeneration. Overall, the results indicated that the foamed porous MBG-PLGA scaffolds incorporating bioactive lipids achieved desirable vascularization-coupled bone formation and could be a promising strategy for bone regenerative medicine. STATEMENT OF SIGNIFICANCE: Efficacious coupling of vascularizationandbone formation is critical for the restoration of large bone defects. Anoveltechnique was used to fabricate composite scaffolds incorporating bioactive lipids which possessedsynergistic cues of bioactive lipids and therapeutic ions to potently promotebone regenerationas well as vascularization. The underlying molecular mechanism for the osteogenic and pro-angiogenic effects of the compositescaffolds was unveiled. Interestingly, the scaffolds were furtherfoundto enhance the formation oftype H capillarieswithin the bone healing microenvironment to couple angiogenesis to osteogenesis to achieve satisfyingvascularizedbone regeneration.These findings provide a novel strategy to develop efficiently vascularized engineering constructs to treat massive bone defects.
Collapse
Affiliation(s)
- Shuang Li
- Department of Stomatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Chaobo Song
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai, China
| | - Weijun Yu
- College of Stomatology, School of Medicine, Shanghai Jiao Tong University, 390 Yanqiao Road, Shanghai, China
| | - Weiqi Zhang
- Department of Stomatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Guohua Zhang
- Department of Stomatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Zhenhao Xi
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, China.
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.
| |
Collapse
|
16
|
Uskoković V. Mechanism of formation governs the mechanism of release of antibiotics from calcium phosphate nanopowders and cements in a drug-dependent manner. J Mater Chem B 2019; 7:3982-3992. [PMID: 31681475 PMCID: PMC6824273 DOI: 10.1039/c9tb00444k] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The kinetics of drug release from hydroxyapatite (HAp) cements could be tuned by controlling the kinetics of crystallization of their HAp precursor powders during synthesis. Here it is shown that this history of formation affects not only the kinetics, but also the mechanism of release. Cements composed of two HAp powders precipitated under different conditions, one (HAp2) taking twice longer to transform from the amorphous to the crystalline state than the other (HAp1), were mixed at different ratios to tune their drug release kinetics and tested for the release mechanism in conjunction with compositional and microstructural analyses. While the cement component converting to the amorphous phase during gelation (HAp2) exhibited a faster, but also more anomalous, non-Fickian mechanism of release of vancomycin, the cement component retaining its crystalline state all throughout gelation, setting and hardening (HAp1) stabilized at the ideal, Fickian diffusion case corresponding to the Korsmeyer-Peppas exponent value of 0.45 ± 0.02. This effect got reversed for the other antibiotic studied as a drug, ciprofloxacin, in which case HAp2 exhibited the ideal, Fickian diffusion with n = 0.45 ± 0.02 and the increase in the content of the cement component retaining its crystallinity during gelation, setting and hardening (HAp1) steadily shifted the mechanism of release to more anomalous, non-Fickian types. This has indicated that the molecular structure of the drug is an essential determinant of the mechanism of release and that the design of a carrier for a universally tunable release of drugs based on the passive transport is likely impossible. Preliminary assays involving the addition of chitosan or gelatin as polymeric components to HAp led to the inclusion of swelling and erosion as additional effects by which the drug escapes the carrier and shifted the release toward less diffusional and more multimodal mechanisms. With regard to the microstructural and compositional effects governing the release mechanism and kinetics, the retention of a finite concentration of slit-like pores of the amorphous precursor in HAp2 and its lower surface energy and lesser drug binding potential in the gelled, amorphous state, but also its possibly less stable and more diffusive particle surface and higher structural water content were elaborated as potential reasons explaining the distinct rates and mechanisms of release from the two HAp powders with different histories of formation.
Collapse
Affiliation(s)
- Vuk Uskoković
- Department of Bioengineering, University of Illinois, Chicago, IL, USA
| |
Collapse
|
17
|
Basu S, Pacelli S, Feng Y, Lu Q, Wang J, Paul A. Harnessing the Noncovalent Interactions of DNA Backbone with 2D Silicate Nanodisks To Fabricate Injectable Therapeutic Hydrogels. ACS NANO 2018; 12:9866-9880. [PMID: 30189128 PMCID: PMC6563937 DOI: 10.1021/acsnano.8b02434] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Injectable hydrogels present several advantages over prefabricated scaffolds including ease of delivery, shear-thinning property, and broad applicability in the fields of drug delivery and tissue engineering. Here, we report an approach to develop injectable hydrogels with sustained drug release properties, exploiting the chemical nature of the DNA backbone and silicate nanodisks. A two-step gelation method is implemented for generating a combination of noncovalent network points, leading to a physically cross-linked hydrogel. The first step initiates the development of an interconnected structure by utilizing DNA denaturation and rehybridization mechanism to form hydrogen bonds between complementary base pairs of neighboring DNA strands. The anisotropic charge distribution of two-dimensional silicate nanodisks (nSi) makes them an active center in the second step of the gelation process. Silicate nanodisks create additional network points via attractive electrostatic interactions with the DNA backbone, thereby enhancing the mechanical resilience of the formulated hydrogel. The thermally stable hydrogels displayed an increase in elasticity and yield stress as a function of nSi concentration. They were able to form self-supporting structures post injection due to their rapid recovery after removal of cyclic stress. Moreover, the presence of nanosilicate was shown to modulate the release of a model osteogenic drug dexamethasone (Dex). The bioactivity of released Dex was confirmed from in vitro osteogenic differentiation of human adipose stem cells and in vivo bone formation in a rat cranial bone defect model. Overall, our DNA-based nanocomposite hydrogel obtained from a combination of noncovalent network points can serve as an injectable material for bone regeneration and carrier for sustained release of therapeutics.
Collapse
Affiliation(s)
- Sayantani Basu
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - Settimio Pacelli
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - Yi Feng
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Qinghua Lu
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Arghya Paul
- BioIntel Research Laboratory, Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
18
|
Novel Lipid Signaling Mediators for Mesenchymal Stem Cell Mobilization during Bone Repair. Cell Mol Bioeng 2018; 11:241-253. [PMID: 29983824 DOI: 10.1007/s12195-018-0532-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Introduction Mesenchymal stem and progenitor cells (MSCs), which normally reside in the bone marrow, are critical to bone health and can be recruited to sites of traumatic bone injury, contributing to new bone formation. The ability to control the trafficking of MSCs provides therapeutic potential for improving traumatic bone healing and therapy for genetic bone diseases such as hypophosphatasia. Methods In this study, we explored the sphingosine-1-phosphate (S1P) signaling axis as a means to control the mobilization of MSCs into blood and possibly to recruit MSCs enhancing bone growth. Results Loss of S1P receptor 3 (S1PR3) leads to an increase in circulating CD45-/CD29+/CD90+/Sca1 putative mesenchymal progenitor cells, suggesting that blocking S1PR3 may stimulate MSCs to leave the bone marrow. Antagonism of S1PR3 with the small molecule VPC01091 stimulated acute migration of CD45-/CD29+/CD90+/Sca1+ MSCs into the blood as early as 1.5 hours after treatment. VPC01091 administration also increased ectopic bone formation induced by BMP-2 and significantly increased new bone formation in critically sized rat cranial defects, suggesting that mobilized MSCs may home to injuries to contribute to healing. We also explored the possibility of combining S1P manipulation of endogenous host cell occupancy with exogenous MSC transplantation for potential use in combination therapies. Importantly, reducing niche occupancy of host MSCs with VPC01091 does not impede engraftment of exogenous MSCs. Conclusions Our studies suggest that MSC mobilization through S1PR3 antagonism is a promising strategy for endogenous tissue engineering and improving MSC delivery to treat bone diseases.
Collapse
|
19
|
Sphingosine 1-phosphate (S1P) signalling: Role in bone biology and potential therapeutic target for bone repair. Pharmacol Res 2017; 125:232-245. [PMID: 28855094 DOI: 10.1016/j.phrs.2017.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/30/2022]
Abstract
The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.
Collapse
|
20
|
Meshcheryakova A, Mechtcheriakova D, Pietschmann P. Sphingosine 1-phosphate signaling in bone remodeling: multifaceted roles and therapeutic potential. Expert Opin Ther Targets 2017; 21:725-737. [PMID: 28524744 PMCID: PMC5470107 DOI: 10.1080/14728222.2017.1332180] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Sphingolipids belong to a complex class of lipid molecules that are crucially involved in the regulation of important biological processes including proliferation, migration and apoptosis. Given the significant progress made in understanding the sphingolipid pathobiology of several diseases, sphingolipid-related checkpoints emerge as attractive targets. Recent data indicate the multifaceted contribution of the sphingolipid machinery to osteoclast – osteoblast crosstalk, representing one of the pivotal interactions underlying bone homeostasis. Imbalances in the interplay of osteoblasts and osteoclasts might lead to bone-related diseases such as osteoporosis, rheumatoid arthritis, and bone metastases. Areas covered: We summarize and analyze the progress made in bone research in the context of the current knowledge of sphingolipid-related mechanisms regulating bone remodeling. Particular emphasis was given to bioactive sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs). Moreover, the mechanisms of how dysregulations of this machinery cause bone diseases, are covered. Expert opinion: In the context of bone diseases, pharmacological interference with sphingolipid machinery may lead to novel directions in therapeutic strategies. Implementation of knowledge derived from in vivo animal models and in vitro studies using pharmacological agents to manipulate the S1P/S1PRs axes suggests S1PR2 and S1PR3 as potential drug targets, particularly in conjunction with technology for local drug delivery.
Collapse
Affiliation(s)
- Anastasia Meshcheryakova
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| | - Diana Mechtcheriakova
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| | - Peter Pietschmann
- a Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
21
|
Seaman SA, Cao Y, Campbell CA, Peirce SM. Arteriogenesis in murine adipose tissue is contingent on CD68 + /CD206 + macrophages. Microcirculation 2017; 24:10.1111/micc.12341. [PMID: 27976451 PMCID: PMC5432396 DOI: 10.1111/micc.12341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/05/2016] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The surgical transfer of skin, fat, and/or muscle from a donor site to a recipient site within the same patient is a widely performed procedure in reconstructive surgeries. A surgical pretreatment strategy that is intended to increase perfusion in the flap, termed "flap delay," is a commonly employed technique by plastic surgeons prior to flap transplantation. Here, we explored whether CD68+ /CD206+ macrophages are required for arteriogenesis within the flap by performing gain-of-function and loss-of-function studies in a previously published flap delay murine model. METHODS AND RESULTS Local injection of M2-polarized macrophages into the flap resulted in an increase in collateral vessel diameter. Application of a thin biomaterial film loaded with a pharmacological agent (FTY720), which has been previously shown to recruit CD68+ /CD206+ macrophages to remodeling tissue, increased CD68+ /CD206+ cell recruitment and collateral vessel enlargement. Conversely, when local macrophage populations were depleted within the inguinal fat pad via clodronate liposome delivery, we observed fewer CD68+ cells accompanied by diminished collateral vessel enlargement. CONCLUSIONS Our study underscores the importance of macrophages during microvascular adaptations that are induced by flap delay. These studies suggest a mechanism for a translatable therapeutic target that may be used to enhance the clinical flap delay procedure.
Collapse
Affiliation(s)
- Scott A. Seaman
- Department of Biomedical Engineering, University of Virginia
| | - Yiqi Cao
- Department of Biomedical Engineering, University of Virginia
| | | | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia
- Department of Plastic Surgery, University of Virginia
| |
Collapse
|
22
|
Sok MCP, Tria MC, Olingy CE, San Emeterio CL, Botchwey EA. Aspirin-Triggered Resolvin D1-modified materials promote the accumulation of pro-regenerative immune cell subsets and enhance vascular remodeling. Acta Biomater 2017; 53:109-122. [PMID: 28213094 DOI: 10.1016/j.actbio.2017.02.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/18/2022]
Abstract
Many goals in tissue engineering rely on modulating cellular localization and polarization of cell signaling, including the inhibition of inflammatory infiltrate, facilitation of inflammatory cell egress, and clearance of apoptotic cells. Omega-3 polyunsaturated fatty acid-derived resolvins are gaining increasing recognition for their essential roles in inhibition of neutrophil invasion into inflamed tissue and promotion of macrophage phagocytosis of cellular debris as well as their egress to the lymphatics. Biomaterial-based release of lipid mediators is a largely under-explored approach that provides a method to manipulate local lipid signaling gradients in vivo and direct the recruitment and/or polarization of anti-inflammatory cell subsets to suppress inflammatory signaling and enhance angiogenesis and tissue regeneration. The goal of this study was to encapsulate Aspirin-Triggered Resolvin D1 (AT-RvD1) into a degradable biomaterial in order to elucidate the effects of sustained, localized delivery in a model of sterile inflammation. Flow cytometric and imaging analysis at both 1 and 3days after injury showed that localized AT-RvD1 delivery was able significantly increase the accumulation of anti-inflammatory monocytes and M2 macrophages while limiting the infiltration of neutrophils. Additionally, cytokine profiling and longitudinal vascular analysis revealed a shift towards a pro-angiogenic profile with increased concentrations of VEGF and SDF-1α, and increased arteriolar diameter and tortuosity. These results demonstrate the ability of locally-delivered AT-RvD1 to increase pro-regenerative immune subpopulations and promote vascular remodeling. STATEMENT OF SIGNIFICANCE This work is motivated by our efforts to explore the underlying mechanisms of inflammation resolution after injury and to develop biomaterial-based approaches to amplify endogenous mechanisms of resolution and repair. Though specific lipid mediators have been identified that actively promote the resolution of inflammation, biomaterial-based localized delivery of these mediators has been largely unexplored. We loaded Aspirin-Triggered Resolvin D1 into a PLGA scaffold and examined the effects of sustained, localized delivery on the innate immune response. We found that biomaterial delivery of resolvin was able to enhance the accumulation of pro-regenerative populations of immune cells, including anti-inflammatory monocytes, population that has never before been shown to respond to resolvin treatment, and also enhance vascular remodeling in response to tissue injury.
Collapse
Affiliation(s)
- Mary Caitlin P Sok
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Maxianne C Tria
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Claire E Olingy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Cheryl L San Emeterio
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Edward A Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA.
| |
Collapse
|
23
|
Ogle ME, Olingy CE, Awojoodu AO, Das A, Ortiz RA, Cheung HY, Botchwey EA. Sphingosine-1-Phosphate Receptor-3 Supports Hematopoietic Stem and Progenitor Cell Residence Within the Bone Marrow Niche. Stem Cells 2017; 35:1040-1052. [PMID: 28026131 DOI: 10.1002/stem.2556] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/19/2016] [Accepted: 12/08/2016] [Indexed: 12/29/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) egress from bone marrow (BM) during homeostasis and at increased rates during stress; however, the mechanisms regulating their trafficking remain incompletely understood. Here we describe a novel role for lipid receptor, sphingosine-1-phosphate receptor 3 (S1PR3), in HSPC residence within the BM niche. HSPCs expressed increased levels of S1PR3 compared to differentiated BM cells. Pharmacological antagonism or knockout (KO) of S1PR3 mobilized HSPCs into blood circulation, suggesting that S1PR3 influences niche localization. S1PR3 antagonism suppressed BM and plasma SDF-1, enabling HSPCs to migrate toward S1P-rich plasma. Mobilization synergized with AMD3100-mediated antagonism of CXCR4, which tethers HSPCs in the niche, and recovered homing deficits of AMD3100-treated grafts. S1PR3 antagonism combined with AMD3100 improved re-engraftment and survival in lethally irradiated recipients. Our studies indicate that S1PR3 and CXCR4 signaling cooperate to maintain HSPCs within the niche under homeostasis. These results highlight an important role for S1PR3 in HSPC niche occupancy and trafficking that can be harnessed for both rapid clinical stem cell mobilization and re-engraftment strategies, as well as the opportunity to design novel therapeutics for control of recruitment, homing, and localization through bioactive lipid signaling. Stem Cells 2017;35:1040-1052.
Collapse
Affiliation(s)
- Molly E Ogle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Claire E Olingy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Anthony O Awojoodu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Anusuya Das
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Rafael A Ortiz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Hoi Yin Cheung
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Edward A Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
24
|
Wang T, Krieger J, Huang C, Das A, Francis MP, Ogle R, Botchwey E. Enhanced osseous integration of human trabecular allografts following surface modification with bioactive lipids. Drug Deliv Transl Res 2016; 6:96-104. [PMID: 26169381 DOI: 10.1007/s13346-015-0244-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this study, we used extracellular matrix (ECM) gels and human bone allograft as matrix vehicles to deliver the sphingolipid growth factor FTY720 to rodent models of tibial fracture and a critical-sized cranial defect. We show that FTY720 released from injectable ECM gels may accelerate callous formation and resolution and bone volume in a mouse tibial fracture model. We then show that FTY720 binds directly to human trabecular allograft bone and releases over 1 week in vitro. Rat critical-sized cranial defects treated with FTY720-coated grafts show increases in vascularization and bone deposition, with histological and micro-computed topography (microCT) evidence of enhanced bone formation within the graft and defect void. Immunohistochemical analysis suggests that osteogenesis within FTY720-coated grafts is associated with reduced CD68(+) macrophage infiltration and recruitment of CD29(+) bone progenitor cells. Matrix binding of FTY720 thus represents a promising and robust bone regeneration strategy with potential clinical translatability.
Collapse
Affiliation(s)
- Tiffany Wang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive Rm 1311, Atlanta, GA, 30332, USA
| | - Jack Krieger
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive Rm 1311, Atlanta, GA, 30332, USA
| | - Cynthia Huang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | - Anusuya Das
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, USA
| | | | - Roy Ogle
- School of Medical Diagnostic and Translational Science, Old Dominion University, Norfolk, VA, 23529, USA
| | - Edward Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive Rm 1311, Atlanta, GA, 30332, USA.
| |
Collapse
|
25
|
Resop RS, Douaisi M, Craft J, Jachimowski LCM, Blom B, Uittenbogaart CH. Sphingosine-1-phosphate/sphingosine-1-phosphate receptor 1 signaling is required for migration of naive human T cells from the thymus to the periphery. J Allergy Clin Immunol 2016; 138:551-557.e8. [PMID: 27056271 PMCID: PMC7007110 DOI: 10.1016/j.jaci.2015.12.1339] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 12/04/2015] [Accepted: 12/16/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND The mechanisms that govern the egress of mature thymocytes from the human thymus to the periphery remain understudied yet are of utmost importance to the field of basic immunology, as well as T-cell reconstitution in various immunodeficiencies. We examined the expression and function of sphingosine-1-phosphate (S1P) receptors in human thymocyte egress. OBJECTIVES We aimed to determine whether S1P receptors (S1P-Rs) play a role in mature human thymocyte egress and to identify the thymocyte population or populations that express S1P-Rs and respond to S1P by migrating across a concentration gradient. METHODS Human thymocytes were exposed to S1P in Transwell plate migration assays coupled to flow cytometry to evaluate the response to S1P of thymocytes at different stages of maturation. Constitutive S1P-R expression was quantified by means of real-time PCR in sorted thymocyte subsets and flow cytometry. S1P-R1 and Kruppel-like factor 2 expression were monitored after S1P exposure by using flow cytometry and quantitative PCR. RESULTS S1P-R1 was the prevalent S1P receptor on mature human thymocytes (CD3(hi)CD27(+)CD69(-)), the population that also demonstrated the greatest response to S1P in migration assays. Pretreatment with FTY720, an S1P-R1 nonselective modulator significantly reduced migration and suggested a role for S1P-R2 in retaining thymocytes in the tissue. Lastly, surface S1P-R1 expression, as well S1PR1 and Kruppel-like factor 2 (KLF2) transcripts, were significantly decreased in mature thymocytes on exposure to S1P. CONCLUSION Mature human thymocytes rely on S1P-R1 to migrate toward S1P. Taken in the context of murine work demonstrating that S1P is required for thymocyte egress to the periphery, our data highlight a new key chemokine for human thymocyte egress.
Collapse
Affiliation(s)
- Rachel S Resop
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, Calif; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Calif
| | - Marc Douaisi
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, Calif
| | - Joshua Craft
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, Calif
| | | | - Bianca Blom
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Christel H Uittenbogaart
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, Calif; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; UCLA AIDS Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Calif; Department of Pediatrics, University of California Los Angeles, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Mele L, Vitiello PP, Tirino V, Paino F, De Rosa A, Liccardo D, Papaccio G, Desiderio V. Changing Paradigms in Cranio-Facial Regeneration: Current and New Strategies for the Activation of Endogenous Stem Cells. Front Physiol 2016; 7:62. [PMID: 26941656 PMCID: PMC4764712 DOI: 10.3389/fphys.2016.00062] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/09/2016] [Indexed: 12/20/2022] Open
Abstract
Craniofacial area represent a unique district of human body characterized by a very high complexity of tissues, innervation and vascularization, and being deputed to many fundamental function such as eating, speech, expression of emotions, delivery of sensations such as taste, sight, and earing. For this reasons, tissue loss in this area following trauma or for example oncologic resection, have a tremendous impact on patients' quality of life. In the last 20 years regenerative medicine has emerged as one of the most promising approach to solve problem related to trauma, tissue loss, organ failure etc. One of the most powerful tools to be used for tissue regeneration is represented by stem cells, which have been successfully implanted in different tissue/organs with exciting results. Nevertheless, both autologous and allogeneic stem cell transplantation raise many practical and ethical concerns that make this approach very difficult to apply in clinical practice. For this reason different cell free approaches have been developed aiming to the mobilization, recruitment, and activation of endogenous stem cells into the injury site avoiding exogenous cells implant but instead stimulating patients' own stem cells to repair the lesion. To this aim many strategies have been used including functionalized bioscaffold, controlled release of stem cell chemoattractants, growth factors, BMPs, Platelet-Rich-Plasma, and other new strategies such as ultrasound wave and laser are just being proposed. Here we review all the current and new strategies used for activation and mobilization of endogenous stem cells in the regeneration of craniofacial tissue.
Collapse
Affiliation(s)
- Luigi Mele
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Pietro Paolo Vitiello
- Medical Oncology, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara," Second University of Naples Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Francesca Paino
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Alfredo De Rosa
- Department of Odontology and Surgery, Second University of Naples Naples, Italy
| | - Davide Liccardo
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Section of Biotechnology and Medical Histology and Embryology, Second University of Naples Naples, Italy
| |
Collapse
|
27
|
Das A, Segar CE, Chu Y, Wang TW, Lin Y, Yang C, Du X, Ogle RC, Cui Q, Botchwey EA. Bioactive lipid coating of bone allografts directs engraftment and fate determination of bone marrow-derived cells in rat GFP chimeras. Biomaterials 2015; 64:98-107. [PMID: 26125501 DOI: 10.1016/j.biomaterials.2015.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 12/31/2022]
Abstract
Bone grafting procedures are performed to treat wounds incurred during wartime trauma, accidents, and tumor resections. Endogenous mechanisms of repair are often insufficient to ensure integration between host and donor bone and subsequent restoration of function. We investigated the role that bone marrow-derived cells play in bone regeneration and sought to increase their contributions by functionalizing bone allografts with bioactive lipid coatings. Polymer-coated allografts were used to locally deliver the immunomodulatory small molecule FTY720 in tibial defects created in rat bone marrow chimeras containing genetically-labeled bone marrow for monitoring cell origin and fate. Donor bone marrow contributed significantly to both myeloid and osteogenic cells in remodeling tissue surrounding allografts. FTY720 coatings altered the phenotype of immune cells two weeks post-injury, which was associated with increased vascularization and bone formation surrounding allografts. Consequently, degradable polymer coating strategies that deliver small molecule growth factors such as FTY720 represent a novel therapeutic strategy for harnessing endogenous bone marrow-derived progenitors and enhancing healing in load-bearing bone defects.
Collapse
Affiliation(s)
- Anusuya Das
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Claire E Segar
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Yihsuan Chu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Tiffany W Wang
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Yong Lin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Chunxi Yang
- Department of Orthopaedic Surgery, Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Xeujun Du
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Roy C Ogle
- School of Medical Diagnostic and Translational Sciences, Old Dominion University, Norfolk, VA, USA
| | - Quanjun Cui
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA, USA
| | - Edward A Botchwey
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
28
|
Han Q, Yang P, Wu Y, Meng S, Sui L, Zhang L, Yu L, Tang Y, Jiang H, Xuan D, Kaplan DL, Kim SH, Tu Q, Chen J. Epigenetically Modified Bone Marrow Stromal Cells in Silk Scaffolds Promote Craniofacial Bone Repair and Wound Healing. Tissue Eng Part A 2015; 21:2156-65. [PMID: 25923143 DOI: 10.1089/ten.tea.2014.0484] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epigenetic regulation of gene expression is a central mechanism that governs cell stemness, determination, commitment, and differentiation. It has been recently found that PHF8, a major H4K20/H3K9 demethylase, plays a critical role in craniofacial and bone development. In this study, we hypothesize that PHF8 promotes osteoblastogenesis by epigenetically regulating the expression of a nuclear matrix protein, special AT-rich sequence-binding protein 2 (SATB2) that plays pivotal roles in skeletal patterning and osteoblast differentiation. Our results showed that expression levels of PHF8 and SATB2 in preosteoblasts and bone marrow stromal cells (BMSCs) increased simultaneously during osteogenic induction. Overexpressing PHF8 in these cells upregulated the expression of SATB2, Runx2, osterix, and bone matrix proteins. Conversely, knockdown of PHF8 reduced the expression of these genes. Furthermore, ChIP assays confirmed that PHF8 specifically bound to the transcription start site (TSS) of the SATB2 promoter, and the expression of H3K9me1 at the TSS region of SATB2 decreased in PHF8 overexpressed group. Implantation of the BMSCs overexpressing PHF8 with silk protein scaffolds promoted bone regeneration in critical-sized defects in mouse calvaria. Taken together, our results demonstrated that PHF8 epigenetically modulates SATB2 activity, triggering BMSCs osteogenic differentiation and facilitating bone formation and regeneration in biodegradable silk scaffolds.
Collapse
Affiliation(s)
- Qianqian Han
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts.,2 Shandong Provincial Key Lab of Oral Biomedicine , Jinan, China .,3 Guangdong Provincial Stomatological Hospital , Guangzhou, China
| | - Pishan Yang
- 2 Shandong Provincial Key Lab of Oral Biomedicine , Jinan, China .,4 Department of Periodontology, School of Stomatology, Shandong University , Jinan, China
| | - Yuwei Wu
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Shu Meng
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Lei Sui
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Lan Zhang
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Liming Yu
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Yin Tang
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Hua Jiang
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Dongying Xuan
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts.,3 Guangdong Provincial Stomatological Hospital , Guangzhou, China
| | - David L Kaplan
- 5 Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
| | - Sung Hoon Kim
- 6 Cancer Preventive Material Development Research Center (CPMDRC) and Institute, College of Oriental Medicine, Kyung Hee University , Seoul, Korea
| | - Qisheng Tu
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts
| | - Jake Chen
- 1 Division of Oral Biology, Tufts University School of Dental Medicine , Boston, Massachusetts.,7 Department of Anatomy and Cell Biology, Tufts University School of Medicine , Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts
| |
Collapse
|
29
|
Ogle ME, Sefcik LS, Awojoodu AO, Chiappa NF, Lynch K, Peirce-Cottler S, Botchwey EA. Engineering in vivo gradients of sphingosine-1-phosphate receptor ligands for localized microvascular remodeling and inflammatory cell positioning. Acta Biomater 2014; 10:4704-4714. [PMID: 25128750 PMCID: PMC4529737 DOI: 10.1016/j.actbio.2014.08.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/30/2014] [Accepted: 08/06/2014] [Indexed: 12/29/2022]
Abstract
Biomaterial-mediated controlled release of soluble signaling molecules is a tissue engineering approach to spatially control processes of inflammation, microvascular remodeling and host cell recruitment, and to generate biochemical gradients in vivo. Lipid mediators, such as sphingosine 1-phosphate (S1P), are recognized for their essential roles in spatial guidance, signaling and highly regulated endogenous gradients. S1P and pharmacological analogs such as FTY720 are therapeutically attractive targets for their critical roles in the trafficking of cells between blood and tissue spaces, both physiologically and pathophysiologically. However, the interaction of locally delivered sphingolipids with the complex metabolic networks controlling the flux of lipid species in inflamed tissue has yet to be elucidated. In this study, complementary in vitro and in vivo approaches are investigated to identify relationships between polymer composition, drug release kinetics, S1P metabolic activity, signaling gradients and spatial positioning of circulating cells around poly(lactic-co-glycolic acid) biomaterials. Results demonstrate that biomaterial-based gradients of S1P are short-lived in the tissue due to degradation by S1P lyase, an enzyme that irreversibly degrades intracellular S1P. On the other hand, in vivo gradients of the more stable compound, FTY720, enhance microvascular remodeling by selectively recruiting an anti-inflammatory subset of monocytes (S1P3(high)) to the biomaterial. Results highlight the need to better understand the endogenous balance of lipid import/export machinery and lipid kinase/phosphatase activity in order to design biomaterial products that spatially control the innate immune environment to maximize regenerative potential.
Collapse
Affiliation(s)
- Molly E. Ogle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Lauren S. Sefcik
- Department of Chemical & Biomolecular Engineering, Lafayette College, 740 High Street, Easton, PA 18042
| | - Anthony O. Awojoodu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Nathan F. Chiappa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Kevin Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903
| | - Shayn Peirce-Cottler
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903
| | - Edward A. Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
30
|
Gupta S, Ahsan I, Mahfooz N, Abdelhamid N, Ramanathan M, Weinstock-Guttman B. Osteoporosis and multiple sclerosis: risk factors, pathophysiology, and therapeutic interventions. CNS Drugs 2014; 28:731-42. [PMID: 24871932 DOI: 10.1007/s40263-014-0173-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory-demyelinating disease of the nervous system. There has been mounting evidence showing that MS is associated with increased risk of osteoporosis and fractures. The development of osteoporosis in MS patients can be related to the cumulative effects of various factors. This review summarizes the common risk factors and physiologic pathways that play a role in development of osteoporosis in MS patients. Physical inactivity and reduced mechanical load on the bones (offsetting gravity) is likely the major contributing factor for osteoporosis in MS. Additional possible factors leading to reduced bone mass are low vitamin D levels, and use of medications such as glucocorticoids and anticonvulsants. The role of the inflammatory processes related to the underlying disease is considered in the context of the complex bone metabolism. The known effect of different MS disease-modifying therapies on bone health is limited. An algorithm for diagnosis and management of osteoporosis in MS is proposed.
Collapse
Affiliation(s)
- Sahil Gupta
- Department of Neurology, State University of New York, Buffalo, NY, USA
| | | | | | | | | | | |
Collapse
|
31
|
Das A, Barker DA, Wang T, Lau CM, Lin Y, Botchwey EA. Delivery of bioactive lipids from composite microgel-microsphere injectable scaffolds enhances stem cell recruitment and skeletal repair. PLoS One 2014; 9:e101276. [PMID: 25077607 PMCID: PMC4117484 DOI: 10.1371/journal.pone.0101276] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 06/04/2014] [Indexed: 01/07/2023] Open
Abstract
In this study, a microgel composed of chitosan and inorganic phosphates was used to deliver poly(lactic-co-glycolic acid) (PLAGA) microspheres loaded with sphingolipid growth factor FTY720 to critical size cranial defects in Sprague Dawley rats. We show that sustained release of FTY720 from injected microspheres used alone or in combination with recombinant human bone morphogenic protein-2 (rhBMP2) improves defect vascularization and bone formation in the presence and absence of rhBMP2 as evaluated by quantitative microCT and histological measurements. Moreover, sustained delivery of FTY720 from PLAGA and local targeting of sphingosine 1-phosphate (S1P) receptors reduces CD45+ inflammatory cell infiltration, promotes endogenous recruitment of CD29+CD90+ bone progenitor cells and enhances the efficacy of rhBMP2 from chitosan microgels. Companion in vitro studies suggest that selective activation of sphingosine receptor subtype-3 (S1P3) via FTY720 treatment induces smad-1 phosphorylation in bone-marrow stromal cells. Additionally, FTY720 enhances stromal cell-derived factor-1 (SDF-1) mediated chemotaxis of CD90+CD11B-CD45- bone progenitor cells in vitro after stimulation with rhBMP2. We believe that use of such small molecule delivery formulations to recruit endogenous bone progenitors may be an attractive alternative to exogenous cell-based therapy.
Collapse
Affiliation(s)
- Anusuya Das
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Daniel A. Barker
- Department of Otolaryngology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tiffany Wang
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Cheryl M. Lau
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Yong Lin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Edward A. Botchwey
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
32
|
Zhang J, Song J. Amphiphilic degradable polymers for immobilization and sustained delivery of sphingosine 1-phosphate. Acta Biomater 2014; 10:3079-90. [PMID: 24631657 DOI: 10.1016/j.actbio.2014.02.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/18/2014] [Accepted: 02/28/2014] [Indexed: 01/13/2023]
Abstract
Controlled delivery of the angiogenic factor sphingosine 1-phosphate (S1P) represents a promising strategy for promoting vascularization during tissue repair and regeneration. In this study, we developed an amphiphilic biodegradable polymer platform for the stable encapsulation and sustained release of S1P. Mimicking the interaction between amphiphilic S1P and its binding proteins, a series of polymers with hydrophilic poly(ethylene glycol) core and lipophilic flanking segments of polylactide and/or poly(alkylated lactide) with different alkyl chain lengths were synthesized. These polymers were electrospun into fibrous meshes, and loaded with S1P in generally high loading efficiencies (>90%). Sustained S1P release from these scaffolds could be tuned by adjusting the alkyl chain length, blockiness and lipophilic block length, achieving 35-55% and 45-80% accumulative releases in the first 8h and by 7 days, respectively. Furthermore, using endothelial cell tube formation assay and chicken chorioallantoic membrane assay, we showed that the different S1P loading doses and release kinetics translated into distinct pro-angiogenic outcomes. These results suggest that these amphiphilic polymers are effective delivery vehicles for S1P and may be explored as tissue engineering scaffolds where the delivery of lipophilic or amphiphilic bioactive factors is desired.
Collapse
|
33
|
Delivery of small molecules for bone regenerative engineering: preclinical studies and potential clinical applications. Drug Discov Today 2014; 19:794-800. [PMID: 24508820 DOI: 10.1016/j.drudis.2014.01.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 01/01/2014] [Accepted: 01/31/2014] [Indexed: 12/20/2022]
Abstract
Stimulation of bone regeneration using growth factors is a promising approach for musculoskeletal regenerative engineering. However, common limitations with protein growth factors, such as high manufacturing costs, protein instability, contamination issues, and unwanted immunogenic responses of the host reduce potential clinical applications. New strategies for bone regeneration that involve inexpensive and stable small molecules can obviate these problems and have a significant impact on the treatment of skeletal injury and diseases. Over the past decade, a large number of small molecules with the potential of regenerating skeletal tissue have been reported in the literature. Here, we review this literature, paying specific attention to the prospects for small molecule-based bone-regenerative engineering. We also review the preclinical study of small molecules associated with bone regeneration.
Collapse
|
34
|
Heilmann A, Schinke T, Bindl R, Wehner T, Rapp A, Haffner-Luntzer M, Liedert A, Amling M, Ignatius A. Systemic treatment with the sphingosine-1-phosphate analog FTY720 does not improve fracture healing in mice. J Orthop Res 2013; 31:1845-50. [PMID: 23818033 DOI: 10.1002/jor.22426] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/06/2013] [Indexed: 02/04/2023]
Abstract
Sphingosine-1-phosphate (S1P) has recently been recognized as a crucial coupling molecule of osteoclast and osteoblast activity provoking osteoanabolic effects. Targeting S1P receptors could, therefore, be a potential strategy to support bone formation in osteopenic diseases or in fracture repair. Here we investigated whether systemic treatment with the S1P analog FTY720 (Fingolimod) could improve fracture healing. Twelve-week-old, female C57BL/6 mice received an osteotomy of the femur, which was stabilized using an external fixator. The mice received a daily subcutaneous injection of either FTY720 (6 mg/kg) or vehicle from the third postoperative day. Fracture healing was evaluated after 10 and 21 days using biomechanical testing, µ-computed tomography, and histomorphometry. Because FTY720 is supposed to influence osteoclast recruitment, osteoclasts were identified in the fracture callus by staining for tartrate resistant acid phosphatase (TRAP). There were no significant differences in callus mechanical properties, tissue composition and osteoclast number between the groups, suggesting that systemically applied FTY720 did not influence bone regeneration in this model of regular fracture healing. Even if further studies should test the potency of FTY720 under unfavorable healing conditions, we conclude that the effect of systemically applied FTY720 on fracture healing might be inferior compared to other anabolic treatments. © 2013 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 31:1845-1850, 2013.
Collapse
Affiliation(s)
- Aline Heilmann
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Das A, Segar CE, Hughley BB, Bowers DT, Botchwey EA. The promotion of mandibular defect healing by the targeting of S1P receptors and the recruitment of alternatively activated macrophages. Biomaterials 2013; 34:9853-62. [PMID: 24064148 DOI: 10.1016/j.biomaterials.2013.08.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/07/2013] [Indexed: 02/07/2023]
Abstract
Endogenous signals originating at the site of injury are involved in the paracrine recruitment, proliferation, and differentiation of circulating progenitor and diverse inflammatory cell types. Here, we investigate a strategy to exploit endogenous cell recruitment mechanisms to regenerate injured bone by local targeting and activation of sphingosine-1-phosphate (S1P) receptors. A mandibular defect model was selected for evaluating regeneration of bone following trauma or congenital disease. The particular challenges of mandibular reconstruction are inherent in the complex anatomy and function of the bone given that the area is highly vascularized and in close proximity to muscle. Nanofibers composed of poly(DL-lactide-co-glycolide) (PLAGA) and polycaprolactone (PCL) were used to delivery FTY720, a targeted agonist of S1P receptors 1 and 3. In vitro culture of bone progenitor cells on drug-loaded constructs significantly enhanced SDF1α mediated chemotaxis of bone marrow mononuclear cells. In vivo results show that local delivery of FTY720 from composite nanofibers enhanced blood vessel ingrowth and increased recruitment of M2 alternatively activated macrophages, leading to significant osseous tissue ingrowth into critical sized defects after 12 weeks of treatment. These results demonstrate that local activation of S1P receptors is a regenerative cue resulting in recruitment of wound healing or anti-inflammatory macrophages and bone healing. Use of such small molecule therapy can provide an alternative to biological factors for the clinical treatment of critical size craniofacial defects.
Collapse
Affiliation(s)
- Anusuya Das
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
| | | | | | | | | |
Collapse
|
36
|
Luckanagul J, Lee LA, Nguyen QL, Sitasuwan P, Yang X, Shazly T, Wang Q. Porous alginate hydrogel functionalized with virus as three-dimensional scaffolds for bone differentiation. Biomacromolecules 2012; 13:3949-58. [PMID: 23148483 DOI: 10.1021/bm301180c] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In regenerative medicine, a synthetic extracellular matrix is crucial for supporting stem cells during its differentiation process to integrate into surrounding tissues. Hydrogels are used extensively in biomaterials as synthetic matrices to support the cells. However, to mimic the biological niche of a functional tissue, various chemical functionalities are necessary. We present here, a method of functionalizing a highly porous hydrogel with functional groups by mixing the hydrogel with a plant virus, tobacco mosaic virus (TMV), and its mutant. The implication of this process resides with the three important features of TMV: its well-defined genetic/chemical modularity, its multivalency (TMV capsid is composed of 2130 copies of identical subunits), and its well-defined structural features. Previous studies utilizing the native TMV on two-dimensional supports accelerated mesenchymal stem cell differentiation, and surfaces modified with genetically modified viral particles further enhanced cell attachment and differentiation. Herein we demonstrate that functionalization of a porous alginate scaffold can be achieved by the addition of viral particles with minimal processing and downstream purifications, and the cell attachment and differentiation within the macroporous scaffold can be effectively manipulated by altering the peptide or small molecule displayed on the viral particles.
Collapse
Affiliation(s)
- Jittima Luckanagul
- Department of Chemistry and Biochemistry, University of South Carolina, Medical Chronobiology Laboratory and Center for Colon Cancer Research, WJB Dorn VA Medical Center, South Carolina, United States
| | | | | | | | | | | | | |
Collapse
|
37
|
Culpepper BK, Bonvallet PP, Reddy MS, Ponnazhagan S, Bellis SL. Polyglutamate directed coupling of bioactive peptides for the delivery of osteoinductive signals on allograft bone. Biomaterials 2012. [PMID: 23182349 DOI: 10.1016/j.biomaterials.2012.10.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Allograft bone is commonly used as an alternative to autograft, however allograft lacks many osteoinductive factors present in autologous bone due to processing. In this study, we investigated a method to reconstitute allograft with osteoregenerative factors. Specifically, an osteoinductive peptide from collagen I, DGEA, was engineered to express a heptaglutamate (E7) domain, which binds the hydroxyapatite within bone mineral. Addition of E7 to DGEA resulted in 9× greater peptide loading on allograft, and significantly greater retention after a 5-day interval with extensive washing. When factoring together greater initial loading and retention, the E7 domain directed a 45-fold enhancement of peptide density on the allograft surface. Peptide-coated allograft was also implanted subcutaneously into rats and it was found that E7DGEA was retained in vivo for at least 3 months. Interestingly, E7DGEA peptides injected intravenously accumulated within bone tissue, implicating a potential role for E7 domains in drug delivery to bone. Finally, we determined that, as with DGEA, the E7 modification enhanced coupling of a bioactive BMP2-derived peptide on allograft. These results suggest that E7 domains are useful for coupling many types of bone-regenerative molecules to the surface of allograft to reintroduce osteoinductive signals and potentially advance allograft treatments.
Collapse
Affiliation(s)
- Bonnie K Culpepper
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
38
|
Hutmacher DW, Duda G, Guldberg RE. Endogenous musculoskeletal tissue regeneration. Cell Tissue Res 2012; 347:485-8. [DOI: 10.1007/s00441-012-1357-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 01/31/2012] [Indexed: 01/04/2023]
|