1
|
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
|
2
|
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] [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
|
3
|
Li Q, Chang B, Dong H, Liu X. Functional microspheres for tissue regeneration. Bioact Mater 2022; 25:485-499. [PMID: 37056261 PMCID: PMC10087113 DOI: 10.1016/j.bioactmat.2022.07.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/02/2022] Open
Abstract
As a new type of injectable biomaterials, functional microspheres have attracted increasing attention in tissue regeneration because they possess some advantageous properties compared to other biomaterials, including hydrogels. A variety of bio-inspired microspheres with unique structures and properties have been developed as cellular carriers and drug delivery vehicles in recent years. In this review, we provide a comprehensive summary of the progress of functional and biodegradable microspheres that have been used for tissue regeneration over the last two decades. First, we briefly introduce the biomaterials and general methods for microsphere fabrication. Next, we focus on the newly developed technologies for preparing functional microspheres, including macroporous microspheres, nanofibrous microspheres, hollow microspheres, core-shell structured microspheres, and surface-modified functional microspheres. After that, we discuss the application of functional microspheres for tissue regeneration, specifically for bone, cartilage, dental, neural, cardiac, and skin tissue regeneration. Last, we present our perspectives and future directions of functional microspheres as injectable carriers for the future advancement of tissue regeneration.
Collapse
|
4
|
Sung B, Kim M, Abelmann L. Magnetic microgels and nanogels: Physical mechanisms and biomedical applications. Bioeng Transl Med 2021; 6:e10190. [PMID: 33532590 PMCID: PMC7823133 DOI: 10.1002/btm2.10190] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Soft micro- and nanostructures have been extensively developed for biomedical applications. The main focus has been on multifunctional composite materials that combine the advantages of hydrogels and colloidal particles. Magnetic microgels and nanogels can be realized by hybridizing stimuli-sensitive gels and magnetic nanoparticles. They are of particular interest since they can be controlled in a wide range of biological environments by using magnetic fields. In this review, we elucidate physical principles underlying the design of magnetic microgels and nanogels for biomedical applications. Particularly, this article provides a comprehensive and conceptual overview on the correlative structural design and physical functionality of the magnetic gel systems under the concept of colloidal biodevices. To this end, we begin with an overview of physicochemical mechanisms related to stimuli-responsive hydrogels and transport phenomena and summarize the magnetic properties of inorganic nanoparticles. On the basis of the engineering principles, we categorize and summarize recent advances in magnetic hybrid microgels and nanogels, with emphasis on the biomedical applications of these materials. Potential applications of these hybrid microgels and nanogels in anticancer treatment, protein therapeutics, gene therapy, bioseparation, biocatalysis, and regenerative medicine are highlighted. Finally, current challenges and future opportunities in the design of smart colloidal biodevices are discussed.
Collapse
Affiliation(s)
- Baeckkyoung Sung
- KIST Europe Forschungsgesellschaft mbHSaarbrückenGermany
- Department of Biological SciencesKent State UniversityKentOhioUSA
- Division of Energy and Environment TechnologyUniversity of Science and TechnologyDaejeonRepublic of Korea
| | - Min‐Ho Kim
- Department of Biological SciencesKent State UniversityKentOhioUSA
| | - Leon Abelmann
- KIST Europe Forschungsgesellschaft mbHSaarbrückenGermany
- MESA+ Institute for Nanotechnology, University of TwenteEnschedeThe Netherlands
| |
Collapse
|
5
|
Mora-Boza A, Mancipe Castro LM, Schneider RS, Han WM, García AJ, Vázquez-Lasa B, San Román J. Microfluidics generation of chitosan microgels containing glycerylphytate crosslinker for in situ human mesenchymal stem cells encapsulation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111716. [PMID: 33545868 PMCID: PMC8237249 DOI: 10.1016/j.msec.2020.111716] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/04/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive source for cell therapies because of their multiple beneficial properties, i.e. via immunomodulation and secretory factors. Microfluidics is particularly attractive for cell encapsulation since it provides a rapid and reproducible methodology for microgel generation of controlled size and simultaneous cell encapsulation. Here, we report the fabrication of hMSC-laden microcarriers based on in situ ionotropic gelation of water-soluble chitosan in a microfluidic device using a combination of an antioxidant glycerylphytate (G1Phy) compound and tripolyphosphate (TPP) as ionic crosslinkers (G1Phy:TPP-microgels). These microgels showed homogeneous size distributions providing an average diameter of 104 ± 12 μm, somewhat lower than that of control (127 ± 16 μm, TPP-microgels). The presence of G1Phy in microgels maintained cell viability over time and upregulated paracrine factor secretion under adverse conditions compared to control TPP-microgels. Encapsulated hMSCs in G1Phy:TPP-microgels were delivered to the subcutaneous space of immunocompromised mice via injection, and the delivery process was as simple as the injection of unencapsulated cells. Immediately post-injection, equivalent signal intensities were observed between luciferase-expressing microgel-encapsulated and unencapsulated hMSCs, demonstrating no adverse effects of the microcarrier on initial cell survival. Cell persistence, inferred by bioluminescence signal, decreased exponentially over time showing relatively higher half-life values for G1Phy:TPP-microgels compared to TPP-microgels and unencapsulated cells. In overall, results position the microfluidics generated G1Phy:TPP-microgels as a promising microcarrier for supporting hMSC survival and reparative activities.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; CIBER-BBN, Health Institute Carlos III, Madrid, Spain
| | - Lina M Mancipe Castro
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rebecca S Schneider
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Woojin M Han
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Blanca Vázquez-Lasa
- Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; CIBER-BBN, Health Institute Carlos III, Madrid, Spain.
| | - Julio San Román
- Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; CIBER-BBN, Health Institute Carlos III, Madrid, Spain
| |
Collapse
|
6
|
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]
|
7
|
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
|
8
|
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
|
9
|
Bioactive Molecules for Skin Repair and Regeneration: Progress and Perspectives. Stem Cells Int 2019; 2019:6789823. [PMID: 32082386 PMCID: PMC7012201 DOI: 10.1155/2019/6789823] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 10/25/2019] [Indexed: 12/26/2022] Open
Abstract
Skin regeneration is a vexing problem in the field of regenerative medicine. A bioactive molecule-based strategy has been frequently used in skin wound healing in recent years. Bioactive molecules are practical tools for regulating cellular processes and have been applied to control cellular differentiation, dedifferentiation, and reprogramming. In this review, we focus on recent progress in the use of bioactive molecules in skin regenerative medicine, by which desired cell types can be generated in vitro for cell therapy and conventional therapeutics can be developed to repair and regenerate skin in vivo through activation of the endogenous repairing potential. We further prospect that the bioactive molecule-base method might be one of the promising strategies to achieve in situ skin regeneration in the future.
Collapse
|
10
|
Adipose-Derived Stem Cells in Bone Tissue Engineering: Useful Tools with New Applications. Stem Cells Int 2019; 2019:3673857. [PMID: 31781238 PMCID: PMC6875209 DOI: 10.1155/2019/3673857] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose stem cells (ASCs) are a crucial element in bone tissue engineering (BTE). They are easy to harvest and isolate, and they are available in significative quantities, thus offering a feasible and valid alternative to other sources of mesenchymal stem cells (MSCs), like bone marrow. Together with an advantageous proliferative and differentiative profile, they also offer a high paracrine activity through the secretion of several bioactive molecules (such as growth factors and miRNAs) via a sustained exosomal release which can exert efficient conditioning on the surrounding microenvironment. BTE relies on three key elements: (1) scaffold, (2) osteoprogenitor cells, and (3) bioactive factors. These elements have been thoroughly investigated over the years. The use of ASCs has offered significative new advancements in the efficacy of each of these elements. Notably, the phenotypic study of ASCs allowed discovering cell subpopulations, which have enhanced osteogenic and vasculogenic capacity. ASCs favored a better vascularization and integration of the scaffolds, while improvements in scaffolds' materials and design tried to exploit the osteogenic features of ASCs, thus reducing the need for external bioactive factors. At the same time, ASCs proved to be an incredible source of bioactive, proosteogenic factors that are released through their abundant exosome secretion. ASC exosomes can exert significant paracrine effects in the surroundings, even in the absence of the primary cells. These paracrine signals recruit progenitor cells from the host tissues and enhance regeneration. In this review, we will focus on the recent discoveries which have involved the use of ASCs in BTE. In particular, we are going to analyze the different ASCs' subpopulations, the interaction between ASCs and scaffolds, and the bioactive factors which are secreted by ASCs or can induce their osteogenic commitment. All these advancements are ultimately intended for a faster translational and clinical application of BTE.
Collapse
|
11
|
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
|
12
|
Clouet J, Fusellier M, Camus A, Le Visage C, Guicheux J. Intervertebral disc regeneration: From cell therapy to the development of novel bioinspired endogenous repair strategies. Adv Drug Deliv Rev 2019; 146:306-324. [PMID: 29705378 DOI: 10.1016/j.addr.2018.04.017] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 03/29/2018] [Accepted: 04/24/2018] [Indexed: 12/15/2022]
Abstract
Low back pain (LBP), frequently associated with intervertebral disc (IVD) degeneration, is a major public health concern. LBP is currently managed by pharmacological treatments and, if unsuccessful, by invasive surgical procedures, which do not counteract the degenerative process. Considering that IVD cell depletion is critical in the degenerative process, the supplementation of IVD with reparative cells, associated or not with biomaterials, has been contemplated. Recently, the discovery of reparative stem/progenitor cells in the IVD has led to increased interest in the potential of endogenous repair strategies. Recruitment of these cells by specific signals might constitute an alternative strategy to cell transplantation. Here, we review the status of cell-based therapies for treating IVD degeneration and emphasize the current concept of endogenous repair as well as future perspectives. This review also highlights the challenges of the mobilization/differentiation of reparative progenitor cells through the delivery of biologics factors to stimulate IVD regeneration.
Collapse
Affiliation(s)
- Johann Clouet
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; CHU Nantes, Pharmacie Centrale, PHU 11, Nantes F-44093, France; Université de Nantes, UFR Sciences Biologiques et Pharmaceutiques, Nantes F-44035, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France
| | - Marion Fusellier
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Department of Diagnostic Imaging, CRIP, National Veterinary School (ONIRIS), Nantes F-44307, France
| | - Anne Camus
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France
| | - Catherine Le Visage
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France
| | - Jérôme Guicheux
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France; CHU Nantes, PHU4 OTONN, Nantes, F-44093, France.
| |
Collapse
|
13
|
Kumar Meena L, Rather H, Kedaria D, Vasita R. Polymeric microgels for bone tissue engineering applications – a review. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2019.1570512] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Lalit Kumar Meena
- Biomaterials & Biomimetics laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, India
| | - Hilal Rather
- Biomaterials & Biomimetics laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, India
| | - Dhaval Kedaria
- Biomaterials & Biomimetics laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, India
| | - Rajesh Vasita
- Biomaterials & Biomimetics laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, India
| |
Collapse
|
14
|
Kong W, Qi Z, Xia P, Chang Y, Li H, Qu Y, Pan S, Yang X. Local delivery of FTY720 and NSCs on electrospun PLGA scaffolds improves functional recovery after spinal cord injury. RSC Adv 2019; 9:17801-17811. [PMID: 35520542 PMCID: PMC9064641 DOI: 10.1039/c9ra01717h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level. FTY720, also known as fingolimod, has recently been reported to exert a positive effect on the recovery from a spinal cord injury. Through local delivery to the lesion site, FTY720 effectively integrates with biomaterials, and the systemic adverse effects are alleviated. However, the effects of the proper mass ratio of FTY720 in biomaterials on neural stem cell (NSC) proliferation and differentiation, as well as functional recovery after SCI, have not been thoroughly investigated. In our study, we fabricated electrospun poly (lactide-co-glycolide) (PLGA)/FTY720 scaffolds at different mass ratios (0.1%, 1%, and 10%) and characterized these scaffolds. The effects of electrospun PLGA/FTY720 scaffolds on NSC proliferation and differentiation were measured. Then, a rat model of spinal transection was established to investigate the effects of PLGA/FTY720 scaffolds loaded with NSCs. Notably, 1% PLGA/FTY720 scaffolds exerted the best effects on the proliferation and differentiation of NSCs and 10% PLGA/FTY720 was cytotoxic to NSCs. Based on the Basso, Beattie, and Bresnahan (BBB) score, HE staining and immunofluorescence staining, the PLGA/FTY720 scaffold loaded with NSCs effectively promoted the recovery of spinal cord function. Thus, FTY720 properly integrated with electrospun PLGA scaffolds, and electrospun PLGA/FTY720 scaffolds loaded with NSCs may have potential applications for SCI as a nerve implant. Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level.![]()
Collapse
Affiliation(s)
- Weijian Kong
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Zhiping Qi
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Peng Xia
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yuxin Chang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Hongru Li
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yunpeng Qu
- Department of Cardiovascular Medicine
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Su Pan
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| |
Collapse
|
15
|
Dai Z, Huang S. Functional Dynamics Inside Nano- or Microscale Bio-Hybrid Systems. Front Chem 2018; 6:621. [PMID: 30619829 PMCID: PMC6305405 DOI: 10.3389/fchem.2018.00621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/30/2018] [Indexed: 11/22/2022] Open
Abstract
Soft nano- or microgels made by natural or synthetic polymers have been investigated intensively because of their board applications. Due to their porosity and biocompatibility, nano- or microgels can be integrated with various biologics to form a bio-hybrid system. They can support living cells as a scaffold; entrap bioactive molecules as a drug carrier or encapsulate microorganisms as a semi-permeable membrane. Especially, researchers have created various modes of functional dynamics into these bio-hybrid systems. From one side, the encapsulating materials can respond to the external stimulus and release the cargo. From the other side, cells can respond to physical, or chemical properties of the matrix and differentiate into a specific cell type. With recent advancements of synthetic biology, cells can be further programed to respond to certain signals, and express therapeutics or other functional proteins for various purposes. Thus, the integration of nano- or microgels and programed cells becomes a potential candidate in applications spanning from biotechnology to new medicines. This brief review will first talk about several nano- or microgels systems fabricated by natural or synthetic polymers, and further discuss their applications when integrated with various types of biologics. In particular, we will concentrate on the dynamics embedded in these bio-hybrid systems, to dissect their designs and sophisticated functions.
Collapse
Affiliation(s)
- Zhuojun Dai
- Institute for Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | |
Collapse
|
16
|
Zheng L, Huang D. [Effects of FTY720-P on EphA2-EphrinA2 bidirectional signaling in osteoclasts]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:575-580. [PMID: 29806345 DOI: 10.7507/1002-1892.201710109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To investigate the effects of FTY720-P on EphA2-EphrinA2 bidirectional signaling in osteoclasts. Methods Murine RAW264.7 macrophages were induced into osteoclasts by dexamethasone and 1α, 25-dihydroxyvitamin D 3, and identified by tartrate resistant acid phosphatase (TRAP) staining. Then, the osteoclasts were divided into 2 groups. The osteoclasts were treated with 400 ng/mL FTY720-P in experimental group and without FTY720-P in control group, respectively. After 48 hours of culture, the cells in 2 groups were detected by real-time fluorescent quantitative PCR, Western blot, and immunofluorescence staining. The expressions of EphA2, EphrinA2, RhoA, and the bone reconstruction associated proteins[bone morphogenetic protein 2 (BMP-2) and transform growth factor β 1 (TGF-β 1)]were analyzed and compared. Results RAW264.7 cells were successfully induced into osteoclasts identified by TRAP staining. Compared with control group, the relative expressions of EphA2 and EphrinA2 mRNAs and proteins in experimental group significantly decreased after 48 hours ( P<0.05), and the relative expression of RhoA protein also significantly decreased ( P<0.05). The relative expressions of BMP-2 and TGF-β 1 mRNAs were significantly increased ( P<0.05), and those protein expressions were enhanced. Conclusion FTY720-P can down-regulate the expression of RhoA and promote the expressions of TGF- β 1 and BMP-2 by affecting the transduction of EphA2-EphrinA2 bidirectional signaling in osteoclasts.
Collapse
Affiliation(s)
- Libin Zheng
- Guangdong Medical University, Zhanjiang Guangdong, 524023, P.R.China
| | - Dong Huang
- Guangdong Medical University, Zhanjiang Guangdong, 524023, P.R.China;Department of Trauma Orthopedics, Guangdong Second Provincial Central Hospital, Guangzhou Guangdong, 510000,
| |
Collapse
|
17
|
Zheng L, Huang D. [Effect of FTY720-P on the differentiation and maturation of MC3T3-E1 cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:285-290. [PMID: 29806276 PMCID: PMC8414265 DOI: 10.7507/1002-1892.201710108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/15/2018] [Indexed: 11/03/2022]
Abstract
Objective To investigate the effect of FTY720-P on the differentiation and maturation of MC3T3-E1 cells. Methods The MC3T3-E1 cells were divided into the experimental group and the control group. In the experimental group, the cells were induced by the medium containing 400 ng/mL FTY720-P (chloroform as solubilizer) in vitro. In the control group, the cells were cultured with the medium only containing chloroform. The cell morphology of 2 groups were observed by inverted phase contrast microscope; the expression of osteoblast related protein (collagen type Ⅰ and collagen type Ⅲ) was detected by immunofluorescence staining; the alkaline phosphatase (ALP) staining and alizarin red staining were used to observe the formation of osteoblasts and the formation of mineralized nodules in 2 groups; and the TUNEL fluorescence assay was used to detect the cell apoptosis. Results After 48 hours of culture, the cells of 2 groups had grown into slender fusiform at the bottom of the bottle, and there was no significant difference in cell morphology between 2 groups. Immunofluorescence staining showed that the expression of collagen type Ⅰ was positive in the experimental group and weakly positive in the control group; the integrated absorbance ( IA) value of the experimental group was 187 600±7 944, which was significantly higher than that of the control group (14 230±1 070) ( t=43.680, P=0.001). The expression of collagen type Ⅲ was weakly positive in the experimental group and the control group, and there was no significant difference in IA value between 2 groups ( t=1.976, P=0.119). ALP staining and alizarin red staining were positive in the experimental group and negative in the control group. TUNEL staining was positive in the experimental group and negative in the control group; the rate of TUNEL staining positive cells in the experimental group was 35.82%±2.99%, which was significantly higher than that in the control group (2.28%±0.51%) ( t=23.420, P=0.002). Conclusion FTY720-P can promote the osteogenic differentiation of MC3T3-E1 cells with speeding up maturation and mineralization of extracellular matrix and affect the apoptosis of the cells.
Collapse
Affiliation(s)
- Libin Zheng
- Guangdong Medical University, Zhanjiang Guangdong, 524000, P.R.China
| | - Dong Huang
- Guangdong Medical University, Zhanjiang Guangdong, 524000, P.R.China;Department of Trauma Orthopedics, Guangdong Second Provincial Central Hospital, Guangzhou Guangdong, 510000,
| |
Collapse
|
18
|
Krieger JR, Sok MCP, Turner TC, Botchwey EA. Delivery of Immunomodulatory Microparticles in a Murine Model of Rotator Cuff Tear. MRS ADVANCES 2018; 3:1341-1346. [PMID: 30002922 PMCID: PMC6039128 DOI: 10.1557/adv.2018.50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Full thickness rotator cuff tears (RCT) and the associated muscle degeneration that results due to this injury presents a significant clinical burden. The prevention or recovery from this degeneration requires the synchronized behavior of many cells that participate in regeneration. Strategies that tune the inflammatory cascade that is initiated after injury serves as a powerful way to influence tissue repair. Here, we use the local, sustained delivery of the immunomodulatory small molecule FTY720 to examine whether the recruitment of pro-regenerative myeloid cells affects the healing outcome. We find that PLGA microparticles have an atrophic effect on the muscle that is ameliorated with the release of FTY720. However, the inability of FTY720 delivery to induce pro-regenerative monocyte and macrophage recruitment and our findings demonstrating enrichment of CD4+ T cells suggest that effects of this small molecule are context dependent and that the underlying mechanisms behind this RCT associated muscle degeneration require further studies.
Collapse
Affiliation(s)
- Jack R Krieger
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Mary Caitlin P Sok
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Thomas C Turner
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Edward A Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| |
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
|
Patel R, Patel M, Kwak J, Iyer AK, Karpoormath R, Desai S, Rarh V. Polymeric microspheres: a delivery system for osteogenic differentiation. POLYM ADVAN TECHNOL 2017. [DOI: 10.1002/pat.4084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Rajkumar Patel
- School of Electrical and Computer Engineering; The University of Seoul; Seoul 02504 Korea
| | - Madhumita Patel
- Department of Chemistry and Nano Science; Ewha Womans University; Seodaemun-gu Seoul 120-750 South Korea
| | - Jeonghun Kwak
- School of Electrical and Computer Engineering; The University of Seoul; Seoul 02504 Korea
| | - Arun K. Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-Bind) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health, Sciences; Wayne State University; 259 Mack Ave Detroit MI 48201 USA
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, College of Health Sciences; University of Kwa Zulu Natal; Durban 4000 Africa
| | - Shrojal Desai
- Global Infusion Systems R&D at Hospira; Chicago, IL USA
| | - Vimal Rarh
- Department of Chemistry, S.G.T.B. Khalsa College; University of Delhi; Delhi 110007 India
| |
Collapse
|
21
|
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
|
22
|
Quantitative analysis of immune cell subset infiltration of supraspinatus muscle after severe rotator cuff injury. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017; 3:82-93. [PMID: 28706969 DOI: 10.1007/s40883-017-0030-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Rotator cuff tears cause muscle degeneration that is characterized by myofiber atrophy, fatty infiltration, and fibrosis and is minimally responsive to current treatment options. The underlying pathogenesis of rotator cuff muscle degeneration remains to be elucidated, and increasing evidence implicates immune cell infiltration as a significant factor. Because immune cells are comprised of highly heterogeneous subpopulations that exert divergent effects on injured tissue, understanding trafficking and accumulation of immune subpopulations may hold the key to more effective therapies. The present study quantifies subpopulations of immune cells infiltrating the murine supraspinatus muscle after severe rotator cuff injury that includes tenotomy and denervation. Rotator cuff injury stimulates dramatic infiltration of mononuclear phagocytes, enriches mononuclear phagocytes in non-classical subpopulations, and enriches T lymphocytes in TH and Treg subpopulations. The combination of tenotomy plus denervation significantly increases mononuclear phagocyte infiltration, enriches macrophages in the non-classical subpopulation, and decreases T lymphocyte enrichment in TH cells compared to tenotomy alone. Depletion of circulating monocytes via liposomal clodronate accelerates supraspinatus atrophy after tenotomy and denervation. The study may aid rational design of immunologically smart therapies that harness immune cells to enhance outcomes after rotator cuff tears.
Collapse
|
23
|
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
|
24
|
Methods for Generating Hydrogel Particles for Protein Delivery. Ann Biomed Eng 2016; 44:1946-58. [PMID: 27160672 DOI: 10.1007/s10439-016-1637-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
Proteins represent a major class of therapeutic molecules with vast potential for the treatment of acute and chronic diseases and regenerative medicine applications. Hydrogels have long been investigated for their potential in carrying and delivering proteins. As compared to bulk hydrogels, hydrogel microparticles (microgels) hold promise in improving aspects of delivery owing to their less traumatic route of entry into the body and improved versatility. This review discusses common methods of fabricating microgels, including emulsion polymerization, microfluidic techniques, and lithographic techniques. Microgels synthesized from both natural and synthetic polymers are discussed, as are a series of microgels fashioned from environment-responsive materials.
Collapse
|
25
|
Gohil SV, Kuo C, Adams DJ, Maye P, Rowe DW, Nair LS. Evaluation of the donor cell contribution in rh
BMP
‐2 mediated bone formation with chitosan thermogels using fluorescent protein reporter mice. J Biomed Mater Res A 2016; 104:928-41. [DOI: 10.1002/jbm.a.35634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/18/2015] [Accepted: 12/18/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Shalini V. Gohil
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
| | - Chia‐Ling Kuo
- Connecticut Institute for Clinical and Translational Science, Institute for Systems Genomics, University of ConnecticutFarmington Connecticut06030
| | - Douglas J. Adams
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
| | - Peter Maye
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - David W. Rowe
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - Lakshmi S. Nair
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
- Departments of Material Science and Engineering, Biomedical Engineering and Institute of Material ScienceUniversity of ConnecticutStorrs Connecticut06269
| |
Collapse
|
26
|
Balmayor ER. Targeted delivery as key for the success of small osteoinductive molecules. Adv Drug Deliv Rev 2015; 94:13-27. [PMID: 25959428 DOI: 10.1016/j.addr.2015.04.022] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/20/2015] [Accepted: 04/29/2015] [Indexed: 02/08/2023]
Abstract
Molecules such as growth factors, peptides and small molecules can guide cellular behavior and are thus important for tissue engineering. They are rapidly emerging as promising compounds for the regeneration of tissues of the musculoskeletal system. Growth factors have disadvantages such as high cost, short half-life, supraphysiological amounts needed, etc. Therefore, small molecules may be an alternative. These molecules have been discovered using high throughput screening. Small osteoinductive molecules exhibit several advantages over growth factors owing to their small sizes, such as high stability and non-immunogenicity. These molecules may stimulate directly signaling pathways that are important for osteogenesis. However, systemic application doesn't induce osteogenesis in most cases. Therefore, local administration is needed. This may be achieved by using a bone graft material providing additional osteoconductive properties. These graft materials can also act by themselves as a delivery matrix for targeted and local delivery. Furthermore, vascularization is necessary in the process of osteogenesis. Many of the small molecules are also capable of promoting vascularization of the tissue to be regenerated. Thus, in this review, special attention is given to molecules that are capable of inducing both angiogenesis and osteogenesis simultaneously. Finally, more recent preclinical and clinical uses in bone regeneration of those molecules are described, highlighting the needs for the clinical translation of these promising compounds.
Collapse
|