51
|
Garske DS, Schmidt-Bleek K, Ellinghaus A, Dienelt A, Gu L, Mooney DJ, Duda GN, Cipitria A. Alginate Hydrogels for In Vivo Bone Regeneration: The Immune Competence of the Animal Model Matters. Tissue Eng Part A 2020; 26:852-862. [DOI: 10.1089/ten.tea.2019.0310] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Daniela S. Garske
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Anke Dienelt
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Luo Gu
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- Department of Materials Science and Engineering, Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - David J. Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Georg N. Duda
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Amaia Cipitria
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
52
|
Gu Y, Zhuang R, Xie X, Bai Y. Osteogenic stimulation of human dental pulp stem cells with self‐setting biphasic calcium phosphate cement. J Biomed Mater Res B Appl Biomater 2020; 108:1669-1678. [PMID: 31769191 DOI: 10.1002/jbm.b.34512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Yingzhi Gu
- Department of OrthodonticsBeijing Stomatological Hospital, Capital Medical University Beijing China
| | - Rui Zhuang
- Department of Oral and Maxillofacial SurgeryBeijing Stomatological Hospital, Capital Medical University Beijing China
| | - Xianju Xie
- Department of OrthodonticsBeijing Stomatological Hospital, Capital Medical University Beijing China
| | - Yuxing Bai
- Department of OrthodonticsBeijing Stomatological Hospital, Capital Medical University Beijing China
| |
Collapse
|
53
|
Icriverzi M, Dinca V, Moisei M, Evans RW, Trif M, Roseanu A. Lactoferrin in Bone Tissue Regeneration. Curr Med Chem 2020; 27:838-853. [PMID: 31258057 DOI: 10.2174/0929867326666190503121546] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/15/2018] [Accepted: 12/13/2018] [Indexed: 11/22/2022]
Abstract
Among the multiple properties exhibited by lactoferrin (Lf), its involvement in bone regeneration processes is of great interest at the present time. A series of in vitro and in vivo studies have revealed the ability of Lf to promote survival, proliferation and differentiation of osteoblast cells and to inhibit bone resorption mediated by osteoclasts. Although the mechanism underlying the action of Lf in bone cells is still not fully elucidated, it has been shown that its mode of action leading to the survival of osteoblasts is complemented by its mitogenic effect. Activation of several signalling pathways and gene expression, in an LRPdependent or independent manner, has been identified. Unlike the effects on osteoblasts, the action on osteoclasts is different, with Lf leading to a total arrest of osteoclastogenesis. Due to the positive effect of Lf on osteoblasts, the potential use of Lf alone or in combination with different biologically active compounds in bone tissue regeneration and the treatment of bone diseases is of great interest. Since the bioavailability of Lf in vivo is poor, a nanotechnology- based strategy to improve the biological properties of Lf was developed. The investigated formulations include incorporation of Lf into collagen membranes, gelatin hydrogel, liposomes, loading onto nanofibers, porous microspheres, or coating onto silica/titan based implants. Lf has also been coupled with other biologically active compounds such as biomimetic hydroxyapatite, in order to improve the efficacy of biomaterials used in the regulation of bone homeostasis. This review aims to provide an up-to-date review of research on the involvement of Lf in bone growth and healing and on its use as a potential therapeutic factor in bone tissue regeneration.
Collapse
Affiliation(s)
- Madalina Icriverzi
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania.,University of Bucharest, Faculty of Biology, Bucharest, Romania
| | - Valentina Dinca
- National Institute for Laser, Plasma and Radiation Physics, Magurele RO-077125, Romania
| | - Magdalena Moisei
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Robert W Evans
- Brunel University, School of Engineering and Design, London, United Kingdom
| | - Mihaela Trif
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Anca Roseanu
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
54
|
Wang J, Chen X, Yang X, Guo B, Li D, Zhu X, Zhang X. Positive role of calcium phosphate ceramics regulated inflammation in the osteogenic differentiation of mesenchymal stem cells. J Biomed Mater Res A 2020; 108:1305-1320. [PMID: 32064734 DOI: 10.1002/jbm.a.36903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 02/05/2023]
Abstract
Recently, researches have confirmed the crucial role of inflammatory response in Ca-P ceramic-induced osteogenesis, however, the underlying mechanism has not yet been fully understood. In this study, BCP and β-TCP ceramics were used as material models to investigate the effect of physicochemical properties on inflammatory response in vitro. The results showed that BCP and β-TCP could support macrophages attachment, proliferation, and spreading favorably, as well as promote gene expressions of inflammatory related cytokines (IL-1, IL-6, MCP-1, and TNF-α) and growth factors (TGF-β, FGF, PDGF, VEGF, IGF, and EGF). BCP showed a facilitating function on the gene expressions earlier than β-TCP. Further coculture experiments performed in vitro demonstrated that the CMs containing various increased cytokines for macrophages pre-culture could significantly promote MSCs osteogenic differentiation, which was confirmed by the gene expressions of osteogenic specific markers and the intracellular OCN product accumulation under the stimulation of BCP and β-TCP ceramics. Further evidence was found from the formation of mineralized nodules in BCM and TCM. In addition, this study showed a concise relationship between Ca-P ceramic induced inflammation and its osteoinductivity that the increased cytokines and growth factors from macrophages could promote MSCs osteogenic differentiation.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xuening Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Bo Guo
- Department of Ophthalmology, West China Hospital of Sichuan University, Chengdu, China
| | - Danyang Li
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
55
|
Borem R, Madeline A, Bowman M, Gill S, Tokish J, Mercuri J. Differential Effector Response of Amnion- and Adipose-Derived Mesenchymal Stem Cells to Inflammation; Implications for Intradiscal Therapy. J Orthop Res 2019; 37:2445-2456. [PMID: 31287173 DOI: 10.1002/jor.24412] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/25/2019] [Indexed: 02/04/2023]
Abstract
Intervertebral disc degeneration (IVDD) is a progressive condition marked by tissue destruction and inflammation. The therapeutic effector functions of mesenchymal stem cells (MSCs) makes them an attractive therapy for patients with IVDD. While several sources of MSCs exist, the optimal choice for use in the inflamed IVD remains a significant question. Adipose (AD)- and amnion (AM)-derived MSCs have several advantages compared with other sources, however, no study has directly compared the impact of IVDD inflammation on their effector functions. Human MSCs were cultured in media with or without supplementation of interleukin-1β (IL-1β) and tumor necrosis factor-α at concentrations reportedly produced by IVDD cells. MSC proliferation and production of pro- and anti-inflammatory cytokines were quantified following 24 and 48 h of culture. Additionally, the osteogenic and chondrogenic potential of AD- and AM-MSCs was characterized via histology and biochemical analysis following 28 days of culture. In inflammatory culture, AM-MSCs produced significantly more anti-inflammatory IL-10 (14.47 ± 2.39 pg/ml; p = 0.004) and larger chondrogenic pellets (5.67 ± 0.26 mm2 ; p = 0.04) with greater percent area staining positively for glycosaminoglycan (82.03 ± 3.26%; p < 0.001) compared with AD-MSCs (0.00 ± 0.00 pg/ml; 2.76 ± 0.18 mm2 ; 34.75 ± 2.49%; respectively). Conversely, AD-MSCs proliferated more resulting in higher cell numbers (221,000 ± 8,021 cells; p = 0.048) and produced higher concentrations of pro-inflammatory cytokines prostaglandin E2 (1,118.30 ± 115.56 pg/ml; p = 0.030) and IL-1β (185.40 ± 7.63 pg/ml; p = 0.010) compared with AM-MSCs (109,667 ± 5,696 cells; 1,291.40 ± 78.47 pg/ml; 144.10 ± 4.57 pg/ml; respectively). AD-MSCs produced more mineralized extracellular matrix (3.34 ± 0.05 relative absorbance units [RAU]; p < 0.001) compared with AM-MSCs (1.08 ± 0.06 RAU). Under identical inflammatory conditions, a different effector response was observed with AM-MSCs producing more anti-inflammatories and demonstrating enhanced chondrogenesis compared with AD-MSCs, which produced more pro-inflammatory cytokines and demonstrated enhanced osteogenesis. These findings may begin to help inform researchers which MSC source may be optimal for IVD regeneration. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2445-2456, 2019.
Collapse
Affiliation(s)
- Ryan Borem
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Allison Madeline
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Mackenzie Bowman
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Sanjitpal Gill
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634.,Department of Orthopaedic Surgery, Medical Group of the Carolinas-Pelham, Spartanburg Regional Healthcare System, Greer, South Carolina, 29651
| | - John Tokish
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634.,Department of Orthopaedic Surgery, Mayo Clinic, Phoenix, Arizona, 85054
| | - Jeremy Mercuri
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| |
Collapse
|
56
|
Croes M, van der Wal BCH, Vogely HC. Impact of Bacterial Infections on Osteogenesis: Evidence From In Vivo Studies. J Orthop Res 2019; 37:2067-2076. [PMID: 31329305 PMCID: PMC6771910 DOI: 10.1002/jor.24422] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/15/2019] [Indexed: 02/04/2023]
Abstract
The clinical impact of bacterial infections on bone regeneration has been incompletely quantified and documented. As a result, controversy exists about the optimal treatment strategy to maximize healing of a contaminated defect. Animal models are extremely useful in this respect, as they can elucidate how a bacterial burden influences quantitative healing of various types of defects relative to non-infected controls. Moreover, they may demonstrate how antibacterial treatment and/or bone grafting techniques facilitate the osteogenic response in the harsh environment of a bacterial infection. Finally, it a well-known contradiction that osteomyelitis is characterized by uncontrolled bone remodeling and bone loss, but at the same time, it can be associated with excessive new bone apposition. Animal studies can provide a better understanding of how osteolytic and osteogenic responses are related to each other during infection. This review discusses the in vivo impact of bacterial infection on osteogenesis by addressing the following questions (i) How does osteomyelitis affect the radiographic bone appearance? (ii) What is the influence of bacterial infection on histological bone healing? (iii) How do bacterial infections affect quantitative bone healing? (iv) What is the effect of antibacterial treatment on the healing outcome during infection? (v) What is the efficacy of osteoinductive proteins in infected bones? (vi) What is the balance between the osteoclastic and osteoblastic response during bacterial infections? (vii) What is the mechanism of the observed pro-osteogenic response as observed in osteomyelitis? © 2019 The Authors. Journal of Orthopaedic Research© published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 37:2067-2076, 2019.
Collapse
Affiliation(s)
- Michiel Croes
- Department of OrthopaedicsUniversity Medical Center UtrechtHeidelberglaan 1003508 GAUtrechtThe Netherlands
| | - Bart C. H. van der Wal
- Department of OrthopaedicsUniversity Medical Center UtrechtHeidelberglaan 1003508 GAUtrechtThe Netherlands
| | - H. Charles Vogely
- Department of OrthopaedicsUniversity Medical Center UtrechtHeidelberglaan 1003508 GAUtrechtThe Netherlands
| |
Collapse
|
57
|
Goodman SB, Pajarinen J, Yao Z, Lin T. Inflammation and Bone Repair: From Particle Disease to Tissue Regeneration. Front Bioeng Biotechnol 2019; 7:230. [PMID: 31608274 PMCID: PMC6761220 DOI: 10.3389/fbioe.2019.00230] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 09/06/2019] [Indexed: 12/17/2022] Open
Abstract
When presented with an adverse stimulus, organisms evoke an immediate, pre-programmed, non-specific innate immune response. The purpose of this reaction is to maintain the organism's biological integrity and function, mitigate or eradicate the injurious source, and re-establish tissue homeostasis. The initial stage of this protective reaction is acute inflammation, which normally reduces or terminates the offending stimulus. As the inflammatory reaction recedes, the stage of tissue repair and regeneration follows. If the above sequence of events is perturbed, reconstitution of normal biological form and function will not be achieved. Dysregulation of these activities may result in incomplete healing, fibrosis, or chronic inflammation. Our laboratory has studied the reaction to wear particles from joint replacements as a paradigm for understanding the biological pathways of acute and chronic inflammation, and potential translational treatments to reconstitute lost bone. As inflammation is the cornerstone for healing in all anatomical locations, the concepts developed have relevance to tissue engineering and regenerative medicine in all organ systems. To accomplish our goal, we developed novel in vitro and in vivo models (including the murine femoral continuous intramedullary particle infusion model), translational strategies including modulation of macrophage chemotaxis and polarization, and methods to interfere with key transcription factors NFκB and MyD88. We purposefully modified MSCs to facilitate bone healing in inflammatory scenarios: by preconditioning the MSCs, and by genetically modifying MSCs to first sense NFκB activation and then overexpress the anti-inflammatory pro-regenerative cytokine IL-4. These advancements provide significant translational opportunities to enhance healing in bone and other organs.
Collapse
Affiliation(s)
- Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States.,Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jukka Pajarinen
- Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Stanford University, Stanford, CA, United States
| | - Tzuhua Lin
- Orthopaedic Research Laboratories, Stanford University, Stanford, CA, United States
| |
Collapse
|
58
|
Hidaka K, Mikuni-Takagaki Y, Wada-Takahashi S, Saita M, Kawamata R, Sato T, Kawata A, Miyamoto C, Maehata Y, Watabe H, Tani-Ishii N, Hamada N, Takahashi SS, Deguchi S, Takeuchi R. Low-Intensity Pulsed Ultrasound Prevents Development of Bisphosphonate-Related Osteonecrosis of the Jaw-Like Pathophysiology in a Rat Model. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:1721-1732. [PMID: 31006496 DOI: 10.1016/j.ultrasmedbio.2019.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/21/2018] [Accepted: 02/15/2019] [Indexed: 06/09/2023]
Abstract
We developed a rat model of bisphosphonate-related osteonecrosis of the jaw (BRONJ) by removing a maxillary molar tooth (M1) from ovariectomized rats after treatment with alendronate. To mimic periodontitis, some of the rats were administered Porphyromonas gingivalis (p. gingivalis) at the M1 site every 2 to 3 d for 2 wk. Rats pretreated with alendronate plus p. gingivalis showed delayed healing of socket epithelia, periosteal reaction of alveolar bone formation and lower bone mineral density in the alveolus above adjacent M2 teeth. These abnormalities were prevented by tooth socket exposure to 20 min/d low-intensity pulsed ultrasound (LIPUS), which restored diminished expression of RANKL, Bcl-2, IL-6, Hsp70, NF-κB and TNF-α messenger ribonucleic acids in remote bone marrow, suggesting LIPUS prevented development of BRONJ-like pathophysiology in rat by inducing systemic responses for regeneration, in addition to accelerating local healing. Non-invasive treatment by LIPUS, as well as low-level laser therapy, may be useful for medication-related osteonecrosis of the jaw patients.
Collapse
Affiliation(s)
- Kouki Hidaka
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan; Department of Oral Interdisciplinary Medicine, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Yuko Mikuni-Takagaki
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan.
| | - Satoko Wada-Takahashi
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Makiko Saita
- Department of Oral Interdisciplinary Medicine, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Ryota Kawamata
- Department of Dentomaxillofacial Diagnosis and Treatment, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Takenori Sato
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Akira Kawata
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Chihiro Miyamoto
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Yojiro Maehata
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Hirotaka Watabe
- Department of Oral Interdisciplinary Medicine, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Nobuyuki Tani-Ishii
- Department of Oral Interdisciplinary Medicine, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Nobushiro Hamada
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Shun-Suke Takahashi
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Shinji Deguchi
- Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan
| | - Ryohei Takeuchi
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka, Japan; Joint Surgery Center, Kawasaki Saiwai Hospital, Kawasaki, Japan
| |
Collapse
|
59
|
Farto-Vaamonde X, Auriemma G, Aquino RP, Concheiro A, Alvarez-Lorenzo C. Post-manufacture loading of filaments and 3D printed PLA scaffolds with prednisolone and dexamethasone for tissue regeneration applications. Eur J Pharm Biopharm 2019; 141:100-110. [PMID: 31112767 DOI: 10.1016/j.ejpb.2019.05.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 10/26/2022]
Abstract
Strategies to load prednisolone or dexamethasone in preformed poly(L-lactic acid) (PLA) filaments and 3D printed scaffolds were explored as a way of personalizing the drug, the dose and the release profile for regenerative medicine purposes. Instead of starting from a PLA filament preloaded with a given content of drug, we explored two more versatile strategies. The first one involved the soaking of PLA filaments into a drug solution prepared in a solvent that reversibly swelled PLA; during 3D printing the melting of PLA contributed to the efficient integration (encapsulation) of the drug inside the printed strand. The second strategy consisted in first printing the 3D PLA scaffolds followed by soaking in a suitable drug solution in order to exploit the higher specific surface of the printed strands compared to the filament. Sustained release profiles were recorded when either prednisolone or dexamethasone were loaded in preformed PLA filaments, while rapid release was recorded for 3D PLA scaffolds loaded after printing. The combination of the two proposed methods reported here opened the possibility of creating concentration gradients of different drugs in the same scaffold exhibiting distinct release patterns. Namely, the strand core contained an active ingredient to be slowly released, while the surface was covered with other active ingredient that could be rapidly delivered. The feasibility of this approach was confirmed through dual loading of dexamethasone in the filament and of prednisolone on the preformed scaffold. Drug-loaded scaffolds were characterized in terms of printability, structural characteristics (DSC, XRD), mechanical properties, biodegradation, and ability to promote cell attachment and proliferation. Finally, anti-inflammatory response and osteoinductive properties were verified in cell cultures.
Collapse
Affiliation(s)
- Xián Farto-Vaamonde
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Giulia Auriemma
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano (SA), Italy
| | - Rita Patrizia Aquino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano (SA), Italy
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
60
|
Chang Y, Cho B, Kim S, Kim J. Direct conversion of fibroblasts to osteoblasts as a novel strategy for bone regeneration in elderly individuals. Exp Mol Med 2019; 51:1-8. [PMID: 31073120 PMCID: PMC6509166 DOI: 10.1038/s12276-019-0251-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/24/2018] [Accepted: 01/28/2019] [Indexed: 12/31/2022] Open
Abstract
Mortality caused by age-related bone fractures or osteoporosis is steadily increasing worldwide as the population ages. The pace of the development of bone regeneration engineering to treat bone fractures has consequently increased in recent years. A range of techniques for bone regeneration, such as immunotherapy, allografts, and hydrogel therapy, have been devised. Cell-based therapies using bone marrow-derived mesenchymal stem cells and induced pluripotent stem cells derived from somatic cells are considered to be suitable approaches for bone repair. However, these cell-based therapies suffer from a number of limitations in terms of efficiency and safety. Somatic cells can also be directly differentiated into osteoblasts by several transcription factors. As osteoblasts play a central role in the process of bone formation, the direct reprogramming of fibroblasts into osteoblasts may hence be a new way to treat bone fractures in elderly individuals. Here, we review recent developments regarding the therapeutic potential of the direct reprogramming of cells for bone regeneration. Reprogramming cells that produce connective tissue to form bone instead could help prevent fractures in the elderly. Bones weaken with age, and fractures are a significant health risk in ageing populations. Most current bone regeneration treatments use stem cells, which can differentiate into any type of cell and have infinite capacity to divide; however, they are difficult to source and can lead to tumor formation. Jongpil Kim at Dongguk University in South Korea and coworkers have reviewed a new method that uses genetic signals to transform connective tissue-forming cells into bone-producing cells. The reprogrammed cells have been shown to generate new bone at the desired site, and because they have already lost their capacity for infinite division, tumor formation risk is greatly reduced. This method shows promise to expand treatment options for fractures and osteoporosis.
Collapse
Affiliation(s)
- Yujung Chang
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Byounggook Cho
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Siyoung Kim
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Jongpil Kim
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea. .,Department of Chemistry, Dongguk University, 30, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea.
| |
Collapse
|
61
|
Xue H, Tao D, Weng Y, Fan Q, Zhou S, Zhang R, Zhang H, Yue R, Wang X, Wang Z, Sun Y. Glycosylation of dentin matrix protein 1 is critical for fracture healing via promoting chondrogenesis. Front Med 2019; 13:575-589. [DOI: 10.1007/s11684-019-0693-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/25/2019] [Indexed: 10/26/2022]
|
62
|
Souza DCD, Abreu HDLD, Oliveira PVD, Capelo LP, Passos-Bueno MR, Catalani LH. A fast degrading PLLA composite with a high content of functionalized octacalcium phosphate mineral phase induces stem cells differentiation. J Mech Behav Biomed Mater 2019; 93:93-104. [DOI: 10.1016/j.jmbbm.2019.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/02/2019] [Accepted: 02/03/2019] [Indexed: 01/24/2023]
|
63
|
Bucher CH, Schlundt C, Wulsten D, Sass FA, Wendler S, Ellinghaus A, Thiele T, Seemann R, Willie BM, Volk HD, Duda GN, Schmidt-Bleek K. Experience in the Adaptive Immunity Impacts Bone Homeostasis, Remodeling, and Healing. Front Immunol 2019; 10:797. [PMID: 31031773 PMCID: PMC6474158 DOI: 10.3389/fimmu.2019.00797] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
Bone formation as well as bone healing capacity is known to be impaired in the elderly. Although bone formation is outpaced by bone resorption in aged individuals, we hereby present a novel path that considerably impacts bone formation and architecture: Bone formation is substantially reduced in aged individual owing to the experience of the adaptive immunity. Thus, immune-aging in addition to chronological aging is a potential risk factor, with an experienced immune system being recognized as more pro-inflammatory. The role of the aging immune system on bone homeostasis and on the bone healing cascade has so far not been considered. Within this study mice at different age and immunological experience were analyzed toward bone properties. Healing was assessed by introducing an osteotomy, immune cells were adoptively transferred to disclose the difference in biological vs. chronological aging. In vitro studies were employed to test the interaction of immune cell products (cytokines) on cells of the musculoskeletal system. In metaphyseal bone, immune-aging affects bone homeostasis by impacting bone formation capacity and thereby influencing mass and microstructure of bone trabeculae leading to an overall reduced mechanical competence as found in bone torsional testing. Furthermore, bone formation is also impacted during bone regeneration in terms of a diminished healing capacity observed in young animals who have an experienced human immune system. We show the impact of an experienced immune system compared to a naïve immune system, demonstrating the substantial differences in the healing capacity and bone homeostasis due to the immune composition. We further showed that in vivo mechanical stimulation changed the immune system phenotype in young mice toward a more naïve composition. While this rescue was found to be significant in young individuals, aged mice only showed a trend toward the reconstitution of a more naïve immune phenotype. Considering the immune system's experience level in an individual, will likely allow one to differentiate (stratify) and treat (immune-modulate) patients more effectively. This work illustrates the relevance of including immune diagnostics when discussing immunomodulatory therapeutic strategies for the progressively aging population of the industrial countries.
Collapse
Affiliation(s)
- Christian H Bucher
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Schlundt
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dag Wulsten
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - F Andrea Sass
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Wendler
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Thiele
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ricarda Seemann
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina M Willie
- Department of Pediatric Surgery, Faculty of Medicine, McGill University, Shriners Hospital for Children, Montreal, QC, Canada
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
64
|
Li X, Luo W, Hu J, Chen Y, Yu T, Yang J, Dong S, Tian X, Sun L. Interleukin-27 prevents LPS-induced inflammatory osteolysis by inhibiting osteoclast formation and function. Am J Transl Res 2019; 11:1154-1169. [PMID: 30972153 PMCID: PMC6456512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/21/2019] [Indexed: 06/09/2023]
Abstract
Osteolysis is a serious complication of several chronic inflammatory diseases and is closely associated with a local chronic inflammatory reaction with a variety of causes. Inflammatory factors and osteoclastogenesis can enhance bone erosion. Interleukin-27 (IL-27) is speculated to play an important role in the physiological immune response. However, there are few studies on its effects on osteoclastogenesis. In this study, IL-27 was shown to inhibit receptor activator nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis. The gene expression levels of osteoclast (OC)-specific genes, such as nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) and C-FOS, which are essential for OC differentiation and bone resorption, were significantly reduced. Further investigating the underlying mechanism, we found that IL-27 significantly reduced RANKL-induced osteoclastogenesis by inhibiting the phosphorylation of IκB and phosphorylation of nuclear factor κB (NF-κB) p65. Furthermore, IL-27 was shown to inhibit lipopolysaccharide (LPS)-induced osteolysis in vivo. Collectively, these results indicate that IL-27 may be a potential candidate for the treatment of osteolytic diseases.
Collapse
Affiliation(s)
- Xianghe Li
- Guizhou Medical UniversityGuiyang 550025, China
| | - Wei Luo
- Guizhou Medical UniversityGuiyang 550025, China
| | - Junxian Hu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University)Chongqing 400038, China
| | - Yueqi Chen
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University)Chongqing 400038, China
| | - Tao Yu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University)Chongqing 400038, China
| | - Jing Yang
- Center of Trauma of Daping Hospital, Third Military Medical University (Army Medical University)Chongqing 400038, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University)Chongqing 400038, China
| | | | - Li Sun
- Department of Orthopedics, Guizhou Province People’s HospitalGuiyang 550002, China
| |
Collapse
|
65
|
Mast cells in mastocytosis and allergy - Important player in metabolic and immunological homeostasis. Adv Med Sci 2019; 64:124-130. [PMID: 30641273 DOI: 10.1016/j.advms.2018.08.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/27/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022]
Abstract
The role of mast cell (MC) activity in pathophysiology is complex and challenging and its clinical effects are difficult to predict. Apart from the known role of MCs in basic immunological processes and allergy, underlined is their importance in bone mineralization and in regulation of autoimmune reactions. Mast cell mediators, especially those released from mast cells in degranulation, but also those released constitutively, are important both in metabolic and immunological processes. Mastocytosis is a heterogeneous group of disorders characterized by accumulation of MC in one or more organs. There are scientific data indicating that mastocytosis patients are at increased risk of osteoporosis in the systemic form of the disease and children with cutaneous mastocytosis have a higher rate of hypogammaglobulinemia. Moreover, the origin of osteoporosis in patients with allergy is no longer considered as linked to steroid therapy only, but to the mast cell mediators' activity as well. There are indications that osteoporosis symptoms in this group of patients may develop independently of the cumulative steroids' dose. Thus, the influence of mast cells on metabolic and immunologic processes in allergic patients should be investigated. The assessment of mast cell activity and burden in mastocytosis may be used to guide clinical management of patients with allergy.
Collapse
|
66
|
Kohno Y, Lin T, Pajarinen J, Romero-Lopez M, Maruyama M, Huang JF, Nathan K, Yao Z, Goodman SB. Osteogenic ability of rat bone marrow concentrate is at least as efficacious as mesenchymal stem cells in vitro. J Biomed Mater Res B Appl Biomater 2019; 107:2500-2506. [PMID: 30779478 DOI: 10.1002/jbm.b.34340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 11/18/2018] [Accepted: 01/26/2019] [Indexed: 01/01/2023]
Abstract
Cell therapy using bone marrow concentrate (BMC) or purified and expanded mesenchymal stem cells (MSCs) has been shown to have a promising osteogenic capacity. However, few studies have directly compared their relative osteogenic ability. The aim of this study was to compare the osteogenic ability of BMC isolated by density gradient centrifugation with bone marrow-derived MSCs in vitro using the cells of 3-month-old Sprague-Dawley rats. The isolated cells were seeded onto 24-well plates (1 × 105 cells/well) and cultured in control growth media, osteogenic media with dexamethasone, or media without dexamethasone (which simulated the in vivo tissue environment). Alkaline phosphatase activity at week 2, osteocalcin using quantitative real-time polymerase chain reaction at week 4, and Alizarin red staining at week 4 were evaluated. In the osteogenic media with dexamethasone, BMC showed equivalent (osteocalcin) or even greater (Alizarin red staining) osteogenic ability compared to MSCs, suggesting that cross-talk among various cells in the BMC leads to greater osteogenesis. Furthermore, in the osteogenic media without dexamethasone, BMC showed equivalent (osteocalcin) or a trend for greater (Alizarin red staining) bone formation than MSCs alone. Our results suggest that BMC has at least comparable bone regeneration potential to MSCs. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B:2500-2506, 2019.
Collapse
Affiliation(s)
- Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Monica Romero-Lopez
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Jhih-Fong Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Karthik Nathan
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
67
|
Li L, Lu H, Zhao Y, Luo J, Yang L, Liu W, He Q. Functionalized cell-free scaffolds for bone defect repair inspired by self-healing of bone fractures: A review and new perspectives. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 98:1241-1251. [PMID: 30813005 DOI: 10.1016/j.msec.2019.01.075] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/15/2018] [Accepted: 01/17/2019] [Indexed: 12/20/2022]
Abstract
Studies have demonstrated that scaffolds, a component of bone tissue engineering, play an indispensable role in bone repair. However, these scaffolds involving ex-vivo cultivated cells seeded have disadvantages in clinical practice, such as limited autologous cells, time-consuming cell expansion procedures, low survival rate and immune-rejection issues. To overcome these disadvantages, recent focus has been placed on the design of functionalized cell-free scaffolds, instead of cell-seeded scaffolds, that can reduplicate the natural self-healing events of bone fractures, such as inflammation, cell recruitment, vascularization, and osteogenic differentiation. New approaches and applications in tissue engineering and regenerative medicine continue to drive the development of functionalized cell-free scaffolds for bone repair. In this review, the self-healing processes were highlighted, and approaches for the functionalization were summarized. Also, ongoing efforts and breakthroughs in the field of functionalization for bone defect repair were discussed. Finally, a brief summery and new perspectives for functionalization strategies were presented to provide guidelines for further efforts in the design of bioinspired cell-free scaffolds.
Collapse
Affiliation(s)
- Li Li
- Institute for Clean Energy & Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing 400715, PR China; Orthopedic Department, Southwest Hospital, Army Medical University, Chongqing 400038, PR China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Orthopedic Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, PR China
| | - Hongwei Lu
- Orthopedic Department, Southwest Hospital, Army Medical University, Chongqing 400038, PR China
| | - Yulan Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Jiangming Luo
- Center of Joint Surgery, Southwest Hospital, Army Medical University, Chongqing 400038, PR China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Qingyi He
- Institute for Clean Energy & Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing 400715, PR China; Orthopedic Department, Southwest Hospital, Army Medical University, Chongqing 400038, PR China; Orthopedic Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
68
|
Longoni A, Knežević L, Schepers K, Weinans H, Rosenberg AJWP, Gawlitta D. The impact of immune response on endochondral bone regeneration. NPJ Regen Med 2018; 3:22. [PMID: 30510772 PMCID: PMC6265275 DOI: 10.1038/s41536-018-0060-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/26/2018] [Indexed: 12/29/2022] Open
Abstract
Tissue engineered cartilage substitutes, which induce the process of endochondral ossification, represent a regenerative strategy for bone defect healing. Such constructs typically consist of multipotent mesenchymal stromal cells (MSCs) forming a cartilage template in vitro, which can be implanted to stimulate bone formation in vivo. The use of MSCs of allogeneic origin could potentially improve the clinical utility of the tissue engineered cartilage constructs in three ways. First, ready-to-use construct availability can speed up the treatment process. Second, MSCs derived and expanded from a single donor could be applied to treat several patients and thus the costs of the medical interventions would decrease. Finally, it would allow more control over the quality of the MSC chondrogenic differentiation. However, even though the envisaged clinical use of allogeneic cell sources for bone regeneration is advantageous, their immunogenicity poses a significant obstacle to their clinical application. The aim of this review is to increase the awareness of the role played by immune cells during endochondral ossification, and in particular during regenerative strategies when the immune response is altered by the presence of implanted biomaterials and/or cells. More specifically, we focus on how this balance between immune response and bone regeneration is affected by the implantation of a cartilaginous tissue engineered construct of allogeneic origin.
Collapse
Affiliation(s)
- A Longoni
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,Regenerative Medicine Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - L Knežević
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,3Faculty of Health Sciences, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD UK
| | - K Schepers
- 4Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - H Weinans
- 5Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands.,6Department of Rheumatology, University Medical Center Utrecht, Utrecht University, 3584CX Utrecht, The Netherlands.,7Department of Biomechanical Engineering, Delft University of Technology, 2628CD Delft, The Netherlands
| | - A J W P Rosenberg
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands
| | - D Gawlitta
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,Regenerative Medicine Center Utrecht, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
69
|
Chang Q, Cai J, Wang Y, Yang R, Xing M, Lu F. Large adipose tissue generation in a mussel-inspired bioreactor of elastic-mimetic cryogel and platelets. J Tissue Eng 2018; 9:2041731418808633. [PMID: 30505425 PMCID: PMC6259050 DOI: 10.1177/2041731418808633] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022] Open
Abstract
Soft tissue generation, especially in large tissue, is a major challenge in reconstructive surgery to treat congenital deformities, posttraumatic repair, and cancer rehabilitation. The concern is along with the donor site morbidity, donor tissue shortage, and flap necrosis. Here, we report a dissection-free adipose tissue chamber-based novel guided adipose tissue regeneration strategy in a bioreactor of elastic gelatin cryogel and polydopamine-assisted platelet immobilization intended to improve angiogenesis and generate large adipose tissue in situ. In order to have matched tissue mechanics, we used 5% gelatin cryogel as growth substrate of bioreactor. Platelets from the platelet-rich plasma were then immobilized onto the gelatin cryogel with the aid of polydopamine to form a biomimetic bioreactor (polydopamine/gelatin cryogel/platelet). Platelets on the substrate led to a sustained high release in both platelet-derived growth factor and vascular endothelial growth factor compared with non-polydopamine-assisted group. The formed bioreactor was then transferred to a tissue engineering chamber and then inserted above inguinal fat pad of rats without flap dissection. This integrate strategy significantly boomed the vessel density, stimulated cellular proliferation, and upregulated macrophage infiltration. There was a noticeable rise in the expression of dual-angiogenic growth factors (platelet-derived growth factor and vascular endothelial growth factor) in chamber fluid; host cell migration and host fibrous protein secretion coordinated with gelatin cryogel degradation. The regenerated adipose tissue volume gained threefold larger than control group (p < 0.05) with less fibrosis tissue. These results indicate that a big well-vascularized three-dimensional mature adipose tissue can be regenerated using elastic gel, polydopamine, platelets, and small fat tissue.
Collapse
Affiliation(s)
- Qiang Chang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, Canada
| | - Junrong Cai
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ruijia Yang
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, Canada
| | - Malcolm Xing
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, Canada.,State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
70
|
Tang R, Yi J, Yang J, Chen Y, Luo W, Dong S, Fei J. Interleukin-37 inhibits osteoclastogenesis and alleviates inflammatory bone destruction. J Cell Physiol 2018; 234:7645-7658. [PMID: 30414292 PMCID: PMC6587950 DOI: 10.1002/jcp.27526] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/10/2018] [Indexed: 12/30/2022]
Abstract
Excessive osteoclast formation is one of the important pathological features of inflammatory bone destruction. Interleukin‐37 (IL‐37) is an anti‐inflammatory agent that is present throughout the body, but it displays low physiological retention. In our study, high levels of the IL‐37 protein were detected in clinical specimens from patients with bone infections. However, the impact of IL‐37 on osteoclast formation remains unclear. Next, IL‐37 alleviated the inflammatory bone destruction in the mouse in vivo. We used receptor activator of nuclear factor‐κB ligand and lipopolysaccharide to trigger osteoclastogenesis under physiological and pathological conditions to observe the role of IL‐37 in this process and explore the potential mechanism of this phenomenon. In both induction models, IL‐37 exerted inhibitory effects on osteoclast differentiation and bone resorption. Furthermore, IL‐37 decreased the phosphorylation of inhibitor of κBα and p65 and the expression of nuclear factor of activated T cells c1, while the dimerization inhibitor of myeloid differentiation factor 88 reversed the effects. These data provide evidence that IL‐37 modulates osteoclastogenesis and a theoretical basis for the clinical application of IL‐37 as a treatment for bone loss–related diseases.
Collapse
Affiliation(s)
- Ruohui Tang
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jin Yi
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jing Yang
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yueqi Chen
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei Luo
- Department of Osteological, Guizhou Province People's Hospital, Guiyang, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jun Fei
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| |
Collapse
|
71
|
Fahmy-Garcia S, Mumcuoglu D, de Miguel L, Dieleman V, Witte-Bouma J, van der Eerden BCJ, van Driel M, Eglin D, Verhaar JAN, Kluijtmans SGJM, van Osch GJVM, Farrell E. Novel In Situ Gelling Hydrogels Loaded with Recombinant Collagen Peptide Microspheres as a Slow-Release System Induce Ectopic Bone Formation. Adv Healthc Mater 2018; 7:e1800507. [PMID: 30230271 DOI: 10.1002/adhm.201800507] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Indexed: 01/06/2023]
Abstract
New solutions for large bone defect repair are needed. Here, in situ gelling slow release systems for bone induction are assessed. Collagen-I based Recombinant Peptide (RCP) microspheres (MSs) are produced and used as a carrier for bone morphogenetic protein 2 (BMP-2). The RCP-MSs are dispersed in three hydrogels: high mannuronate (SLM) alginate, high guluronate (SLG) alginate, and thermoresponsive hyaluronan derivative (HApN). HApN+RCP-MS forms a gel structure at 32 ºC or above, while SLM+RCP-MS and SLG+RCP-MS respond to shear stress displaying thixotropic behavior. Alginate formulations show sustained release of BMP-2, while there is minimal release from HApN. These formulations are injected subcutaneously in rats. SLM+RCP-MS and SLG+RCP-MS loaded with BMP-2 induce ectopic bone formation as revealed by X-ray tomography and histology, whereas HApN+RCP-MS do not. Vascularization occurs within all the formulations studied and is significantly higher in SLG+MS and HApN+RCP-MS than in SLM+RCP-MS. Inflammation (based on macrophage subset staining) decreases over time in both alginate groups, but increases in the HApN+RCP-MS condition. It is shown that a balance between inflammatory cell infiltration, BMP-2 release, and vascularization, achieved in the SLG+RCP-MS alginate condition, is optimal for the induction of de novo bone formation.
Collapse
Affiliation(s)
- Shorouk Fahmy-Garcia
- Department of Orthopedics; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
- Department of Internal Medicine; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
| | - Didem Mumcuoglu
- Department of Orthopedics; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
- Fujifilm Manufacturing Europe B.V.; Oudenstaart 1 5047TK Tilburg The Netherlands
| | - Laura de Miguel
- Fujifilm Manufacturing Europe B.V.; Oudenstaart 1 5047TK Tilburg The Netherlands
| | - Veerle Dieleman
- Department of Oral and Maxillofacial Surgery; Special Dental Care and Orthodontics; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery; Special Dental Care and Orthodontics; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
| | | | - Marjolein van Driel
- Department of Internal Medicine; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
| | - David Eglin
- AO Research Institute Davos; Clavadelerstrasse 8 7270 Davos Switzerland
| | - Jan A. N. Verhaar
- Department of Orthopedics; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
| | | | - Gerjo J. V. M. van Osch
- Department of Orthopedics; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
- Department of Otorhinolaryngology; Head and Neck Surgery; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery; Special Dental Care and Orthodontics; Erasmus MC; Wytemaweg 80 3015CN Rotterdam The Netherlands
| |
Collapse
|
72
|
Dzobo K, Thomford NE, Senthebane DA, Shipanga H, Rowe A, Dandara C, Pillay M, Motaung KSCM. Advances in Regenerative Medicine and Tissue Engineering: Innovation and Transformation of Medicine. Stem Cells Int 2018; 2018:2495848. [PMID: 30154861 PMCID: PMC6091336 DOI: 10.1155/2018/2495848] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/22/2018] [Accepted: 07/08/2018] [Indexed: 02/08/2023] Open
Abstract
Humans and animals lose tissues and organs due to congenital defects, trauma, and diseases. The human body has a low regenerative potential as opposed to the urodele amphibians commonly referred to as salamanders. Globally, millions of people would benefit immensely if tissues and organs can be replaced on demand. Traditionally, transplantation of intact tissues and organs has been the bedrock to replace damaged and diseased parts of the body. The sole reliance on transplantation has created a waiting list of people requiring donated tissues and organs, and generally, supply cannot meet the demand. The total cost to society in terms of caring for patients with failing organs and debilitating diseases is enormous. Scientists and clinicians, motivated by the need to develop safe and reliable sources of tissues and organs, have been improving therapies and technologies that can regenerate tissues and in some cases create new tissues altogether. Tissue engineering and/or regenerative medicine are fields of life science employing both engineering and biological principles to create new tissues and organs and to promote the regeneration of damaged or diseased tissues and organs. Major advances and innovations are being made in the fields of tissue engineering and regenerative medicine and have a huge impact on three-dimensional bioprinting (3D bioprinting) of tissues and organs. 3D bioprinting holds great promise for artificial tissue and organ bioprinting, thereby revolutionizing the field of regenerative medicine. This review discusses how recent advances in the field of regenerative medicine and tissue engineering can improve 3D bioprinting and vice versa. Several challenges must be overcome in the application of 3D bioprinting before this disruptive technology is widely used to create organotypic constructs for regenerative medicine.
Collapse
Affiliation(s)
- Kevin Dzobo
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Dimakatso Alice Senthebane
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Hendrina Shipanga
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Arielle Rowe
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Collet Dandara
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Michael Pillay
- Department of Biotechnology, Faculty of Applied and Computer Sciences, Vaal University of Technology, Vanderbijlpark 1900, South Africa
| | | |
Collapse
|
73
|
Lai WY, Feng SW, Chan YH, Chang WJ, Wang HT, Huang HM. In Vivo Investigation into Effectiveness of Fe₃O₄/PLLA Nanofibers for Bone Tissue Engineering Applications. Polymers (Basel) 2018; 10:E804. [PMID: 30960729 PMCID: PMC6404065 DOI: 10.3390/polym10070804] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 01/08/2023] Open
Abstract
Fe₃O₄ nanoparticles were loaded into poly-l-lactide (PLLA) with concentrations of 2% and 5%, respectively, using an electrospinning method. In vivo animal experiments were then performed to evaluate the potential of the Fe₃O₄/PLLA nanofibrous material for bone tissue engineering applications. Bony defects with a diameter of 4 mm were prepared in rabbit tibias. Fe₃O₄/PLLA nanofibers were grafted into the drilled defects and histological examination and computed tomography (CT) image detection were performed after an eight-week healing period. The histological results showed that the artificial bony defects grafted with Fe₃O₄/PLLA nanofibers exhibited a visibly higher bone healing activity than those grafted with neat PLLA. In addition, the quantitative results from CT images revealed that the bony defects grafted with 2% and 5% Fe₃O₄/PLLA nanofibers, respectively, showed 1.9- and 2.3-fold increases in bone volume compared to the control blank sample. Overall, the results suggest that the Fe₃O₄/PLLA nanofibers fabricated in this study may serve as a useful biomaterial for future bone tissue engineering applications.
Collapse
Affiliation(s)
- Wei-Yi Lai
- School of Organic and Polymeric, National Taipei University of Technology, Taipei 10608, Taiwan.
| | - Sheng-Wei Feng
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ya-Hui Chan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Wei-Jen Chang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Dental Department, Taipei Medical University Shuang-Ho Hospital, New Taipei City 23561, Taiwan.
| | - Hsin-Ta Wang
- School of Organic and Polymeric, National Taipei University of Technology, Taipei 10608, Taiwan.
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Biomedical Optomechatronics, College of Biomedical Engineering, Taipei 11031, Taiwan.
| |
Collapse
|
74
|
Hachemi Y, Rapp AE, Picke AK, Weidinger G, Ignatius A, Tuckermann J. Molecular mechanisms of glucocorticoids on skeleton and bone regeneration after fracture. J Mol Endocrinol 2018; 61:R75-R90. [PMID: 29588427 PMCID: PMC5976078 DOI: 10.1530/jme-18-0024] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/27/2018] [Indexed: 12/29/2022]
Abstract
Glucocorticoid hormones (GCs) have profound effects on bone metabolism. Via their nuclear hormone receptor - the GR - they act locally within bone cells and modulate their proliferation, differentiation, and cell death. Consequently, high glucocorticoid levels - as present during steroid therapy or stress - impair bone growth and integrity, leading to retarded growth and glucocorticoid-induced osteoporosis, respectively. Because of their profound impact on the immune system and bone cell differentiation, GCs also affect bone regeneration and fracture healing. The use of conditional-mutant mouse strains in recent research provided insights into the cell-type-specific actions of the GR. However, despite recent advances in system biology approaches addressing GR genomics in general, little is still known about the molecular mechanisms of GCs and GR in bone cells. Here, we review the most recent findings on the molecular mechanisms of the GR in general and the known cell-type-specific actions of the GR in mesenchymal cells and their derivatives as well as in osteoclasts during bone homeostasis, GC excess, bone regeneration and fracture healing.
Collapse
Affiliation(s)
- Yasmine Hachemi
- Institute of Comparative Molecular EndocrinologyUlm University, Ulm, Germany
| | - Anna E Rapp
- Institute of Orthopaedic Research and BiomechanicsUlm University Medical Centre, Ulm, Germany
| | - Ann-Kristin Picke
- Institute of Comparative Molecular EndocrinologyUlm University, Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular BiologyUlm University, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and BiomechanicsUlm University Medical Centre, Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular EndocrinologyUlm University, Ulm, Germany
| |
Collapse
|
75
|
Diomede F, D'Aurora M, Gugliandolo A, Merciaro I, Ettorre V, Bramanti A, Piattelli A, Gatta V, Mazzon E, Fontana A, Trubiani O. A novel role in skeletal segment regeneration of extracellular vesicles released from periodontal-ligament stem cells. Int J Nanomedicine 2018; 13:3805-3825. [PMID: 29988728 PMCID: PMC6029600 DOI: 10.2147/ijn.s162836] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose The combination of oral derived stem cells and 3-D scaffolds is considered advantageous in bone repair. In particular, collagen membranes possess ideal biological properties and can support infiltration and proliferation of osteoblasts, promoting bone regeneration. Our study aimed to develop a new biocompatible osteogenic construct composed of a commercially available collagen membrane (Evolution [Evo]), human periodontal-ligament stem cells (hPDLSCs) enriched with extracellular vesicles (EVs), or polyethylenimine (PEI)-engineered EVs (PEI-EVs). Methods Osteogenic ability and expression of osteogenic genes were evaluated in vitro in hPDLSCs cultured with or without Evo, with Evo and EVs, or PEI-EVs. In addition, the bone-regeneration capacity of Evo, Evo enriched with hPDLSCs, Evo enriched with hPDLSCs and EVs/PEI-EVs was investigated in rats subjected to calvarial defects. Results Our results showed that Evo enriched with EVs and PEI-EVs showed high biocompatibility and osteogenic properties in vitro and in vivo. In addition, quantitative reverse-transcription polymerase chain reaction demonstrated the upregulation of osteogenic genes, such as TGFB1, MMP8, TUFT1, TFIP11, BMP2, and BMP4, in the presence of PEI-EVs. Upregulation of BMP2/4 was confirmed for Evo enriched with PEI-EVs and hPDLSCs both in vitro by Western blot and in vivo by immunofluorescence. Conclusion Our results indicated that Evo enriched with hPDLSCs and PEI-EVs is able to promote a bone-regeneration process for the treatment of calvarium and ossification defects caused by accidental or surgery trauma. In particular, PEI-EVs had a significant role in activation of the osteogenic process.
Collapse
Affiliation(s)
- Francesca Diomede
- Department of Medical, Oral, and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Marco D'Aurora
- Department of Psychological, Health, and Territorial Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Agnese Gugliandolo
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy,
| | - Ilaria Merciaro
- Department of Medical, Oral, and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Valeria Ettorre
- Department of Pharmacy, University "G. d'Annunzio", Chieti, Italy
| | - Alessia Bramanti
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy, .,Eduardo Caianiello Institute of Applied Science and Intelligent Systems (ISASI), National Research Council, Messina, Italy
| | - Adriano Piattelli
- Department of Medical, Oral, and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Valentina Gatta
- Department of Psychological, Health, and Territorial Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Emanuela Mazzon
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy,
| | | | - Oriana Trubiani
- Department of Medical, Oral, and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| |
Collapse
|
76
|
Diomede F, Gugliandolo A, Cardelli P, Merciaro I, Ettorre V, Traini T, Bedini R, Scionti D, Bramanti A, Nanci A, Caputi S, Fontana A, Mazzon E, Trubiani O. Three-dimensional printed PLA scaffold and human gingival stem cell-derived extracellular vesicles: a new tool for bone defect repair. Stem Cell Res Ther 2018; 9:104. [PMID: 29653587 PMCID: PMC5899396 DOI: 10.1186/s13287-018-0850-0] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/27/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The role of bone tissue engineering in the field of regenerative medicine has been a main research topic over the past few years. There has been much interest in the use of three-dimensional (3D) engineered scaffolds (PLA) complexed with human gingival mesenchymal stem cells (hGMSCs) as a new therapeutic strategy to improve bone tissue regeneration. These devices can mimic a more favorable endogenous microenvironment for cells in vivo by providing 3D substrates which are able to support cell survival, proliferation and differentiation. The present study evaluated the in vitro and in vivo capability of bone defect regeneration of 3D PLA, hGMSCs, extracellular vesicles (EVs), or polyethyleneimine (PEI)-engineered EVs (PEI-EVs) in the following experimental groups: 3D-PLA, 3D-PLA + hGMSCs, 3D-PLA + EVs, 3D-PLA + EVs + hGMSCs, 3D-PLA + PEI-EVs, 3D-PLA + PEI-EVs + hGMSCs. METHODS The structural parameters of the scaffold were evaluated using both scanning electron microscopy and nondestructive microcomputed tomography. Nanotopographic surface features were investigated by means of atomic force microscopy. Scaffolds showed a statistically significant mass loss along the 112-day evaluation. RESULTS Our in vitro results revealed that both 3D-PLA + EVs + hGMSCs and 3D-PLA + PEI-EVs + hGMSCs showed no cytotoxicity. However, 3D-PLA + PEI-EVs + hGMSCs exhibited greater osteogenic inductivity as revealed by morphological evaluation and transcriptomic analysis performed by next-generation sequencing (NGS). In addition, in vivo results showed that 3D-PLA + PEI-EVs + hGMSCs and 3D-PLA + PEI-EVs scaffolds implanted in rats subjected to cortical calvaria bone tissue damage were able to improve bone healing by showing better osteogenic properties. These results were supported also by computed tomography evaluation that revealed the repair of bone calvaria damage. CONCLUSION The re-establishing of the integrity of the bone lesions could be a promising strategy in the treatment of accidental or surgery trauma, especially for cranial bones.
Collapse
Affiliation(s)
- Francesca Diomede
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | | | - Paolo Cardelli
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Ilaria Merciaro
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Valeria Ettorre
- Department of Pharmacy, University "G. d'Annunzio", Chieti, Italy
| | - Tonino Traini
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Rossella Bedini
- National Centre of Innovative Technologies in Public Health, Italian National Institute of Health, Rome, Italy
| | | | - Alessia Bramanti
- IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy.,Institute of Applied Science and Intelligent Systems "ISASI Eduardo Caianiello", CNR, Messina, Italy
| | - Antonio Nanci
- Laboratory for the study of Calcified Tissues and Biomaterials, Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec, Canada
| | - Sergio Caputi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | | | | | - Oriana Trubiani
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Via dei Vestini, 66100, Chieti, Italy.
| |
Collapse
|
77
|
Tatara AM, Kontoyiannis DP, Mikos AG. Drug delivery and tissue engineering to promote wound healing in the immunocompromised host: Current challenges and future directions. Adv Drug Deliv Rev 2018; 129:319-329. [PMID: 29221962 PMCID: PMC5988908 DOI: 10.1016/j.addr.2017.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/23/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022]
Abstract
As regenerative medicine matures as a field, more promising technologies are being translated from the benchtop to the clinic. However, many of these strategies are designed with otherwise healthy hosts in mind and validated in animal models without other co-morbidities. In reality, many of the patient populations benefiting from drug delivery and tissue engineering-based devices to enhance wound healing also have significant underlying immunodeficiency. Specifically, patients suffering from diabetes, malignancy, human immunodeficiency virus, post-organ transplantation, and other compromised states have significant pleotropic immune defects that affect wound healing. In this work, we review the role of different immune cells in the regenerative process, highlight the effect of several common immunocompromised states on wound healing, and discuss different drug delivery strategies for overcoming immunodeficiencies.
Collapse
Affiliation(s)
- Alexander M Tatara
- Medical Scientist Training Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, United States; Department of Bioengineering, Rice University, Houston, TX, United States.
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, United States.
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, United States.
| |
Collapse
|
78
|
Diomede F, D'Aurora M, Gugliandolo A, Merciaro I, Orsini T, Gatta V, Piattelli A, Trubiani O, Mazzon E. Biofunctionalized Scaffold in Bone Tissue Repair. Int J Mol Sci 2018; 19:E1022. [PMID: 29596323 PMCID: PMC5979468 DOI: 10.3390/ijms19041022] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 03/15/2018] [Accepted: 03/26/2018] [Indexed: 01/02/2023] Open
Abstract
Bone tissue engineering is based on bone grafting to repair bone defects. Bone graft substitutes can contribute to the addition of mesenchymal stem cells (MSCs) in order to enhance the rate and the quality of defect regeneration. The stem cell secretome contains many growth factors and chemokines, which could affect cellular characteristics and behavior. Conditioned medium (CM) could be used in tissue regeneration avoiding several problems linked to the direct use of MSCs. In this study, we investigated the effect of human periodontal ligament stem cells (hPDLSCs) and their CM on bone regeneration using a commercially available membrane scaffold Evolution (EVO) implanted in rat calvarias. EVO alone or EVO + hPDLSCs with or without CM were implanted in Wistar male rats subjected to calvarial defects. The in vivo results revealed that EVO membrane enriched with hPDLSCs and CM showed a better osteogenic ability to repair the calvarial defect. These results were confirmed by acquired micro-computed tomography (CT) images and the increased osteopontin levels. Moreover, RT-PCR in vitro revealed the upregulation of three genes (Collagen (COL)5A1, COL16A1 and transforming growth factor (TGF)β1) and the down regulation of 26 genes involved in bone regeneration. These results suggest a promising potential application of CM from hPDLSCs and scaffolds for bone defect restoration and in particular for calvarial repair in case of trauma.
Collapse
Affiliation(s)
- Francesca Diomede
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100 Chieti, Italy.
| | - Marco D'Aurora
- Department of Psychological, Health and Territorial Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100 Chieti, Italy.
| | | | - Ilaria Merciaro
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100 Chieti, Italy.
| | - Tiziana Orsini
- CNR-National Research Council, Institute of Cell Biology and Neurobiology (IBCN), via Ramarini 32, Monterotondo, 00015 Roma, Italy.
| | - Valentina Gatta
- Department of Psychological, Health and Territorial Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100 Chieti, Italy.
| | - Adriano Piattelli
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100 Chieti, Italy.
| | - Oriana Trubiani
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100 Chieti, Italy.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino Pulejo", 98124 Messina, Italy.
| |
Collapse
|
79
|
Abstract
Soft tissue trauma of skeletal muscle is one of the most common side effects in surgery. Muscle injuries are not only caused by accident-related injuries but can also be of an iatrogenic nature as they occur during surgical interventions when the anatomical region of interest is exposed. If the extent of trauma surpasses the intrinsic regenerative capacities, signs of fatty degeneration and formation of fibrotic scar tissue can occur, and, consequentially, muscle function deteriorates or is diminished. Despite research efforts to investigate the physiological healing cascade following trauma, our understanding of the early onset of healing and how it potentially determines success or failure is still only fragmentary. This review focuses on the initial physiological pathways following skeletal muscle trauma in comparison to bone and tendon trauma and what conclusions can be drawn from new scientific insights for the development of novel therapeutic strategies. Strategies to support regeneration of muscle tissue after injury are scarce, even though muscle trauma has a high incidence. Based on tissue specific differences, possible clinical treatment options such as local immune-modulatory and cell therapeutic approaches are suggested that aim to support the endogenous regenerative potential of injured muscle tissues.
Collapse
|
80
|
Correia CR, Gaifem J, Oliveira MB, Silvestre R, Mano JF. The influence of surface modified poly(l-lactic acid) films on the differentiation of human monocytes into macrophages. Biomater Sci 2018; 5:551-560. [PMID: 28128374 DOI: 10.1039/c6bm00920d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Macrophages play a crucial role in the biological performance of biomaterials, as key factors in defining the optimal inflammation-healing balance towards tissue regeneration and implant integration. Here, we investigate how different surface modifications performed on poly(l-lactic acid) (PLLA) films would influence the differentiation of human monocytes into macrophages. We tested PLLA films without modification, surface-modified by plasma treatment (pPLLA) or by combining plasma treatment with different coating materials, namely poly(l-lysine) and a series of proteins from the extracellular matrix: collagen I, fibronectin, vitronectin, laminin and albumin. While all the tested films are non-cytotoxic, differences in cell adhesion and morphology are observed. Monocyte-derived macrophages (MDM) present a more rounded shape in non-modified films, while a more elongated phenotype is observed containing filopodia-like and podosome-like structures in all modified films. No major differences are found for the expression of HLA-DR+/CD80+ and CD206+/CD163+ surface markers, as well as for the ability of MDM to phagocytize. Interestingly, MDM differentiated on pPLLA present the highest expression of MMP9. Upon differentiation, MDM in all surface modified films present lower amounts of IL-6 and IL-10 compared to non-modified films. After stimulating MDM with the potent pro-inflammatory agent LPS, pPLLA and poly(l-lysine) and fibronectin-modified films reveal a significant reduction in IL-6 secretion, while the opposite effect is observed with IL-10. Of note, in comparison to non-modified films, all surface modified films induce a significant reduction of the IL-6/IL-10 ratio, a valuable prognosticator of the pro- versus anti-inflammatory balance. These findings provide important insights into MDM-biomaterial interactions, while strengthening the need for designing immune-informed biomaterials.
Collapse
Affiliation(s)
- Clara R Correia
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Joana Gaifem
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal. and Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | - Mariana B Oliveira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Ricardo Silvestre
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal. and Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | - João F Mano
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
81
|
Grčević D, Sironi M, Valentino S, Deban L, Cvija H, Inforzato A, Kovačić N, Katavić V, Kelava T, Kalajzić I, Mantovani A, Bottazzi B. The Long Pentraxin 3 Plays a Role in Bone Turnover and Repair. Front Immunol 2018; 9:417. [PMID: 29556234 PMCID: PMC5845433 DOI: 10.3389/fimmu.2018.00417] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 02/15/2018] [Indexed: 01/04/2023] Open
Abstract
Pentraxin 3 (PTX3) is an inflammatory mediator acting as a fluid-phase pattern recognition molecule and playing an essential role in innate immunity and matrix remodeling. Inflammatory mediators also contribute to skeletal homeostasis, operating at multiple levels in physiological and pathological conditions. This study was designed to investigate the role of PTX3 in physiological skeletal remodeling and bone healing. Micro-computed tomography (μCT) and bone histomorphometry of distal femur showed that PTX3 gene-targeted female and male mice (ptx3−/−) had lower trabecular bone volume than their wild-type (ptx3+/+) littermates (BV/TV by μCT: 3.50 ± 1.31 vs 6.09 ± 1.17 for females, p < 0.0001; BV/TV 9.06 ± 1.89 vs 10.47 ± 1.97 for males, p = 0.0435). In addition, μCT revealed lower trabecular bone volume in second lumbar vertebra of ptx3−/− mice. PTX3 was increasingly expressed during osteoblast maturation in vitro and was able to reverse the negative effect of fibroblast growth factor 2 (FGF2) on osteoblast differentiation. This effect was specific for the N-terminal domain of PTX3 that contains the FGF2-binding site. By using the closed transversal tibial fracture model, we found that ptx3−/− female mice formed significantly less mineralized callus during the anabolic phase following fracture injury compared to ptx3+/+ mice (BV/TV 17.05 ± 4.59 vs 20.47 ± 3.32, p = 0.0195). Non-hematopoietic periosteal cells highly upregulated PTX3 expression during the initial phase of fracture healing, particularly CD51+ and αSma+ osteoprogenitor subsets, and callus tissue exhibited concomitant expression of PTX3 and FGF2 around the fracture site. Thus, PTX3 supports maintenance of the bone mass possibly by inhibiting FGF2 and its negative impact on bone formation. Moreover, PTX3 enables timely occurring sequence of callus mineralization after bone fracture injury. These results indicate that PTX3 plays an important role in bone homeostasis and in proper matrix mineralization during fracture repair, a reflection of the function of this molecule in tissue homeostasis and repair.
Collapse
Affiliation(s)
- Danka Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marina Sironi
- Humanitas Clinical and Research Center, Milan, Italy
| | | | - Livija Deban
- Humanitas Clinical and Research Center, Milan, Italy.,Oxford BioTherapeutics Ltd., Abingdon, United Kingdom
| | - Hrvoje Cvija
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Nataša Kovačić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vedran Katavić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tomislav Kelava
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmingam, CT, United States
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
82
|
Abstract
Achieving satisfactory reconstruction of bone remains an important goal in orthopedic and dental conditions such as bone trauma, osteoporosis, arthritis, osteonecrosis, and periodontitis. Appropriate temporal and spatial differentiation of mesenchymal stem cells (MSCs) is essential for postnatal bone regeneration. Additionally, an acute inflammatory response is crucial at the onset of bone repair, while an adaptive immune response has important implications during late bone remodeling. Various reports have indicated bidirectional interactions between MSCs and inflammatory cells or molecules. For example, inflammatory cells can recruit MSCs, direct their migration and differentiation, so as to exert anabolic effects on bone repair. Furthermore, both pro-inflammatory and anti-inflammatory cytokines can regulate MSCs properties and subsequent bone regeneration. MSCs have demonstrated highly immunosuppressive functions, such as inhibiting the differentiation of monocytes/hematopoietic precursors and suppressing the secretion of pro-inflammatory cytokines. This review emphasizes the important interactions between inflammatory stimuli, MSCs, and bone regeneration as well as the underlying regulatory mechanisms. Better understanding of these principles will provide new opportunities for promoting bone regeneration and the treatment of bone loss associated with immunological diseases.
Collapse
|
83
|
Huang Y, Wu C, Zhang X, Chang J, Dai K. Regulation of immune response by bioactive ions released from silicate bioceramics for bone regeneration. Acta Biomater 2018; 66:81-92. [PMID: 28864248 DOI: 10.1016/j.actbio.2017.08.044] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/29/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022]
Abstract
Silicate bioceramics have been considered to possess a wide prospect of clinical application for orthopedic tissue regeneration due to their excellent osteogenesis and angiogenesis. However, the mechanism for silicate bioceramics stimulating bone formation is not fully understood. The host immune defense to implants is proved to greatly influence the osteogenesis and new bone formation, but up to now, few studies are focused on the silicate bioceramics modulated host immune responses. In our present study, two representative silicate bioceramics, akermanite (AKT) and nagelschmidtite (NAGEL) were used as model materials to investigate the inflammation responses in vitro and in vivo, and β-tricalcium phosphate (β-TCP) bioceramics were used as a control. It was found that the mouse macrophage cell RAW264.7 that cultured on AKT and NAGEL bioceramics displayed not only less viability and proliferation, but also a significant less inflammatory cytokine secretion than those on β-TCP in vitro. The formation of foreign body giant cells and fibrous capsules, the invasion of macrophages, as well as the detected inflammatory cytokines around the implanted materials were much lower in both AKT and NAGEL bioceramic groups as compared with those in the β-TCP controls in vivo. Furthermore, it was found that not just the certain concentration of extracellular Si-containing ionic products released from the silicate bioceramics, but also the separate Si, Mg and Ca ions revealed the activity to inhibit the macrophage inflammatory responses by the way of suppressing the activated inflammatory MAPK and NF-κB signaling pathway and promoting the caspase-dependent apoptosis of macrophages. In general, our study suggests that the silicate bioceramics could regulate immune responses by altering the ionic microenvironment between the implants and hosts, which may offer new insight about the mechanism of the bioactivity of silicate bioceramics in bone regeneration and provide profitable guidance for designing new biomaterials for bone tissue engineering. STATEMENT OF SIGNIFICANCE Silicate bioceramics have been widely used for orthopedic tissue regeneration because of their excellent characteristics in bone formation. However, there are few studies concerning their interrelationships with the host immune defense that has been proved to greatly influence osteogenesis. In our present study, the akermanite and nagelschmidtite were used as two representative silicate bioceramics to investigate the inflammation responses in vitro and in vivo; and for the first time, the bioactive ions released from the silicate bioceramics were discovered to regulate the macrophage immune responses through both inhibiting the inflammatory signaling and activating apoptosis of macrophages. Our findings in this study may not only increase the understanding in osteogenic activity of silicate bioceramics, but also provide profitable guidance for designing and manufacturing new biomaterials for bone tissue engineering.
Collapse
|
84
|
Abstract
Plaque calcification develops by the inflammation-dependent mechanisms involved in progression and regression of atherosclerosis. Macrophages can undergo two distinct polarization states, that is, pro-inflammatory M1 phenotype in progression and anti-inflammatory M2 phenotype in regression. In plaque progression, predominant M1 macrophages promote the initial calcium deposition within the necrotic core of the lesions, called as microcalcification, through not only vesicle-mediated mineralization as the result of apoptosis of macrophages and vascular smooth muscle cells (VSMCs), but also VSMC differentiation into early phase osteoblasts. On the other hand, in plaque regression M2 macrophages are engaged in the healing response to plaque inflammation. In association with the resolution of chronic inflammation, M2 macrophages may facilitate macroscopic calcium deposition, called as macrocalcification, through induction of osteoblastic differentiation and maturation of VSMCs. Oncostatin M, which has been shown to promote osteoblast differentiation in bone, may play a pivotal role in the development of plaque calcification. Clinically, two types of plaque calcification have distinct implications. Macrocalcification leads to plaque stability, while microcalcification is more likely to be associated with plaque rupture. Statin therapy, which reduces cardiovascular mortality, has been shown to exert its dual actions on plaque morphology, that is, regression of atheroma and increment of macroscopic calcium deposits. Statins may facilitate the healing process against plaque inflammation by enhancing M2 polarization of macrophages. Vascular calcification has pleiotropic properties as pro-inflammatory “microcalcification” and anti-inflammatory “macrocalcification”. The molecular mechanisms of this process in relation with plaque progression as well as plaque regression should be intensively elucidated.
Collapse
Affiliation(s)
- Atsushi Shioi
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine.,Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine
| | - Yuji Ikari
- Department of Cardiovascular Medicine, Tokai University School of Medicine
| |
Collapse
|
85
|
Barba A, Diez-Escudero A, Maazouz Y, Rappe K, Espanol M, Montufar EB, Bonany M, Sadowska JM, Guillem-Marti J, Öhman-Mägi C, Persson C, Manzanares MC, Franch J, Ginebra MP. Osteoinduction by Foamed and 3D-Printed Calcium Phosphate Scaffolds: Effect of Nanostructure and Pore Architecture. ACS APPLIED MATERIALS & INTERFACES 2017; 9:41722-41736. [PMID: 29116737 DOI: 10.1021/acsami.7b14175] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Some biomaterials are osteoinductive, that is, they are able to trigger the osteogenic process by inducing the differentiation of mesenchymal stem cells to the osteogenic lineage. Although the underlying mechanism is still unclear, microporosity and specific surface area (SSA) have been identified as critical factors in material-associated osteoinduction. However, only sintered ceramics, which have a limited range of porosities and SSA, have been analyzed so far. In this work, we were able to extend these ranges to the nanoscale, through the foaming and 3D-printing of biomimetic calcium phosphates, thereby obtaining scaffolds with controlled micro- and nanoporosity and with tailored macropore architectures. Calcium-deficient hydroxyapatite (CDHA) scaffolds were evaluated after 6 and 12 weeks in an ectopic-implantation canine model and compared with two sintered ceramics, biphasic calcium phosphate and β-tricalcium phosphate. Only foams with spherical, concave macropores and not 3D-printed scaffolds with convex, prismatic macropores induced significant ectopic bone formation. Among them, biomimetic nanostructured CDHA produced the highest incidence of ectopic bone and accelerated bone formation when compared with conventional microstructured sintered calcium phosphates with the same macropore architecture. Moreover, they exhibited different bone formation patterns; in CDHA foams, the new ectopic bone progressively replaced the scaffold, whereas in sintered biphasic calcium phosphate scaffolds, bone was deposited on the surface of the material, progressively filling the pore space. In conclusion, this study demonstrates that the high reactivity of nanostructured biomimetic CDHA combined with a spherical, concave macroporosity allows the pushing of the osteoinduction potential beyond the limits of microstructured calcium phosphate ceramics.
Collapse
Affiliation(s)
- Albert Barba
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Bone Healing Group, Small Animal Surgery Department, Veterinary School, Universitat Autònoma de Barcelona , 08193 Bellaterra, Barcelona, Spain
| | - Anna Diez-Escudero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Yassine Maazouz
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Katrin Rappe
- Bone Healing Group, Small Animal Surgery Department, Veterinary School, Universitat Autònoma de Barcelona , 08193 Bellaterra, Barcelona, Spain
| | - Montserrat Espanol
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Edgar B Montufar
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Mar Bonany
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Joanna M Sadowska
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Jordi Guillem-Marti
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Caroline Öhman-Mägi
- Materials in Medicine Group, Division of Applied Materials Science, Department of Engineering Sciences, Uppsala University , 751 21 Uppsala, Sweden
| | - Cecilia Persson
- Materials in Medicine Group, Division of Applied Materials Science, Department of Engineering Sciences, Uppsala University , 751 21 Uppsala, Sweden
| | - Maria-Cristina Manzanares
- Human Anatomy and Embryology Unit, Department of Pathology and Experimental Therapeutics, Universitat de Barcelona , 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Franch
- Bone Healing Group, Small Animal Surgery Department, Veterinary School, Universitat Autònoma de Barcelona , 08193 Bellaterra, Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya , Avinguda Eduard Maristany 10-14, 08019 Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC) , 08028 Barcelona, Spain
| |
Collapse
|
86
|
Zhang H, Zhu T, Zhang L, Wu Q. Stromal cell‑derived factor‑1 induces matrix metalloproteinase expression in human endplate chondrocytes, cartilage endplate degradation in explant culture, and the amelioration of nucleus pulposus degeneration in vivo. Int J Mol Med 2017; 41:969-976. [PMID: 29207021 DOI: 10.3892/ijmm.2017.3278] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 11/13/2017] [Indexed: 11/05/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a strong etiological factor in chronic lower back pain. Stem cell migration toward the site of IVD degeneration for regeneration is restricted by avascularity and distance. Our previous study indicated that the expression of stromal cell‑derived factor‑1 (SDF‑1) and its receptor, C-X-C chemokine receptor type 4 (CXCR4) was upregulated in degenerated cartilage endplate (CEP) and nucleus pulposus (NP). In the present study, SDF‑1 increased CXCR4 mRNA and protein expression in human endplate chondrocytes in a dose‑dependent manner. The results of reverse transcription-quantitative polymerase chain reaction, western blotting and zymography indicated that SDF‑1 increased matrix metalloproteinase (MMP)‑1, ‑3 and ‑13 mRNA and protein expression in human endplate chondrocytes in a dose‑dependent manner. The results of zymography suggested that SDF‑1 also increased MMP‑2 and ‑9 protein expression in a dose‑dependent manner. The CXCR4‑specific chemical inhibitor AMD3100 significantly decreased the levels of MMP‑1, ‑2, ‑3, ‑9 and ‑13 expression. In a human cartilage explant culture model, SDF‑1 accelerated the degradation of extracellular matrix (ECM), and AMD3100 decreased cartilage cleavage. However, in a rat tail disc degeneration model, the injection of SDF‑1 into the NP resulted in the retention of dense areas of proteoglycan matrix and enhanced NP regeneration. These results suggest that SDF‑1, as an inflammatory cytokine, induces MMP expression in human endplate chondrocytes and that ECM remodeling in the CEP may be a favorable factor of endogenous stem cell homing into the NP for regeneration in vivo.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Ting Zhu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Li Zhang
- Department of Clinical Laboratory, Hangzhou Red Cross Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| | - Qionghua Wu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
87
|
Schell H, Duda GN, Peters A, Tsitsilonis S, Johnson KA, Schmidt-Bleek K. The haematoma and its role in bone healing. J Exp Orthop 2017; 4:5. [PMID: 28176273 PMCID: PMC5296258 DOI: 10.1186/s40634-017-0079-3] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/30/2017] [Indexed: 12/28/2022] Open
Abstract
Fracture treatment is an old endeavour intended to promote bone healing and to also enable early loading and regain of function in the injured limb. However, in today's clinical routine the healing potential of the initial fracture haematoma is still not fully recognized. The Arbeitsgemeinschaft für Osteosynthesefragen (AO) formed in Switzerland in 1956 formulated four AO principles of fracture treatment which are still valid today. Fracture treatment strategies have continued to evolve further, as for example the relatively new concept of minimally invasive plate osteosynthesis (MIPO). This MIPO treatment strategy harbours the benefit of an undisturbed original fracture haematoma that supports the healing process. The extent of the supportive effect of this haematoma for the bone healing process has not been considered in clinical practice so far. The rising importance of osteoimmunological aspects in bone healing supports the essential role of the initial haematoma as a source for inflammatory cells that release the cytokine pattern that directs cell recruitment towards the injured tissue. In reviewing the potential benefits of the fracture haematoma, the early development of angiogenic and osteogenic potentials within the haematoma are striking. Removing the haematoma during surgery could negatively influence the fracture healing process. In an ovine open tibial fracture model the haematoma was removed 4 or 7 days after injury and the bone that formed during the first two weeks of healing was significantly reduced in comparison with an undisturbed control. These findings indicate that whenever possible the original haematoma formed upon injury should be conserved during clinical fracture treatment to benefit from the inherent healing potential.
Collapse
Affiliation(s)
- H Schell
- Julius Wolff Institut and Center for Musculoskeletal Surgery Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - G N Duda
- Julius Wolff Institut and Center for Musculoskeletal Surgery Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - A Peters
- Julius Wolff Institut and Center for Musculoskeletal Surgery Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - S Tsitsilonis
- Julius Wolff Institut and Center for Musculoskeletal Surgery Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - K A Johnson
- Faculty of Veterinary Science, University of Sydney, Sydney, Australia
| | - K Schmidt-Bleek
- Julius Wolff Institut and Center for Musculoskeletal Surgery Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
88
|
Zhou L, Lin Z, Ding J, Huang W, Chen J, Wu D. Inflammatory and biocompatibility evaluation of antimicrobial peptide GL13K immobilized onto titanium by silanization. Colloids Surf B Biointerfaces 2017; 160:581-588. [DOI: 10.1016/j.colsurfb.2017.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/08/2017] [Accepted: 10/03/2017] [Indexed: 12/27/2022]
|
89
|
|
90
|
Jo YY, Kweon H, Kim DW, Baek K, Kim MK, Kim SG, Chae WS, Choi JY, Rotaru H. Bone regeneration is associated with the concentration of tumour necrosis factor-α induced by sericin released from a silk mat. Sci Rep 2017; 7:15589. [PMID: 29138464 PMCID: PMC5686134 DOI: 10.1038/s41598-017-15687-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/31/2017] [Indexed: 01/04/2023] Open
Abstract
To understand the osteogenic effect of the middle layer of the silk cocoon, sericin was examined for its cellular effects associated with tumor necrosis factor-α (TNF-α) signaling in this study. The fragmented sericin proteins in the silk mat were evaluated for the TNF-α expression level in murine macrophages. The concentration of protein released from silk mats was higher in the outermost and the innermost layers than in the middle layers, and the protein released from the silk mat was identified as sericin. The level of TNF-α in murine macrophages was dependent on the applied concentration of sericin, and the expression of genes associated with osteogenesis in osteoblast-like cells was dependent on the applied concentration of TNF-α. In animal experiments, silk mats from the middle layers led to a higher regenerated bone volume than silk mats from the innermost layer or the outermost layer. If TNF-α protein was incorporated into the silk mats from the middle layers, bone regeneration was suppressed compared with unloaded silk mats from the middle layers. Accordingly, silk mats from the silk cocoon can be considered to be a fragmented sericin-secreting carrier, and the level of sericin secretion is associated with TNF-α induction and bone regeneration.
Collapse
Affiliation(s)
- You-Young Jo
- Sericultural and Apicultural Division, National Institute of Agricultural Science, RDA, Wanju, 55365, Republic of Korea
| | - HaeYong Kweon
- Sericultural and Apicultural Division, National Institute of Agricultural Science, RDA, Wanju, 55365, Republic of Korea
| | - Dae-Won Kim
- Department of Oral Biochemistry, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea
| | - Kyunghwa Baek
- Department of Oral Pharmacology, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea
| | - Min-Keun Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea
| | - Seong-Gon Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea.
| | - Weon-Sik Chae
- Analysis Research Division, Daegu Center, Korea Basic Science Institute, Daegu, 41566, Republic of Korea
| | - Je-Yong Choi
- School of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, Skeletal Diseases Analysis Center, Korea Mouse Phenotyping Center (KMPC), Kyungpook National University, Daegu, 41944, Korea.
| | - Horatiu Rotaru
- Department of Cranio-Maxillofacial Surgery, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, 400001, Romania
| |
Collapse
|
91
|
Croes M, Boot W, Kruyt MC, Weinans H, Pouran B, van der Helm YJ, Gawlitta D, Vogely HC, Alblas J, Dhert WJ, Öner FC. Inflammation-Induced Osteogenesis in a Rabbit Tibia Model. Tissue Eng Part C Methods 2017. [DOI: 10.1089/ten.tec.2017.0151] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Michiel Croes
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Willemijn Boot
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Moyo C. Kruyt
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Delft, The Netherlands
- Department of Rheumatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Behdad Pouran
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Delft, The Netherlands
| | | | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H. Charles Vogely
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wouter J.A. Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - F. Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
92
|
Graney PL, Roohani-Esfahani SI, Zreiqat H, Spiller KL. In vitro response of macrophages to ceramic scaffolds used for bone regeneration. J R Soc Interface 2017; 13:rsif.2016.0346. [PMID: 27466438 DOI: 10.1098/rsif.2016.0346] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/04/2016] [Indexed: 01/09/2023] Open
Abstract
Macrophages, the primary cells of the inflammatory response, are major regulators of healing, and mediate both bone fracture healing and the inflammatory response to implanted biomaterials. However, their phenotypic contributions to biomaterial-mediated bone repair are incompletely understood. Therefore, we used gene expression and protein secretion analysis to investigate the interactions in vitro between primary human monocyte-derived macrophages and ceramic scaffolds that have been shown to have varying degrees of success in promoting bone regeneration in vivo Specifically, baghdadite (Ca3ZrSi2O9) and strontium-hardystonite-gahnite (Sr-Ca2ZnSi2O7-ZnAl2O4) scaffolds were chosen as two materials that enhanced bone regeneration in vivo in large defects under load compared with clinically used tricalcium phosphate-hydroxyapatite (TCP-HA). Principal component analysis revealed that the scaffolds differentially regulated macrophage phenotype. Temporal changes in gene expression included shifts in markers of pro-inflammatory M1, anti-inflammatory M2a and pro-remodelling M2c macrophage phenotypes. Of note, TCP-HA scaffolds promoted upregulation of many M1-related genes and downregulation of many M2a- and M2c-related genes. Effects of the scaffolds on macrophages were attributed primarily to direct cell-scaffold interactions because of only minor changes observed in transwell culture. Ultimately, elucidating macrophage-biomaterial interactions will facilitate the design of immunomodulatory biomaterials for bone repair.
Collapse
Affiliation(s)
- Pamela L Graney
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Seyed-Iman Roohani-Esfahani
- Biomaterials and Tissue Engineering Research Unit, School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, New South Wales 2026, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, New South Wales 2026, Australia
| | - Kara L Spiller
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
93
|
TLR4 Inactivation in Myeloid Cells Accelerates Bone Healing of a Calvarial Defect Model in Mice. Plast Reconstr Surg 2017; 140:296e-306e. [PMID: 28746278 PMCID: PMC5542792 DOI: 10.1097/prs.0000000000003541] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Toll-like receptor 4 (TLR4) has been implicated in inflammation-induced bone destruction in various chronic bone diseases; however, its direct influence on bone healing is not well understood. The authors’ previous study showed accelerated bone healing with higher osteoclastogenesis gene expression in toll-like receptor 4 knockout mice (TLR4-/-). This study aimed to further elucidate the underlying cellular mechanisms during fracture healing by generating a myeloid cell-specific toll-like receptor 4 knockout model (Lyz-TLR4-/- mice). Methods: Calvarial defects, 1.8 mm in diameter, were created in wild-type, TLR4-/-, and Lyz-TLR4-/- mice. Bone healing was investigated using micro–computed tomography and histologic, histomorphometric, and immunohistochemistry analyses. Primary bone marrow–derived cells were also isolated from wild-type, TLR4-/-, and Lyz-TLR4-/- mice to measure their osteoclast differentiation and resorption properties. Results: A similar faster bone healing response, with active intramembranous bone formation, intense osteopontin staining, and more osteoblast infiltration, was observed in TLR4-/- and Lyz-TLR4-/- mice. Tartrate-resistant acid phosphatase staining showed more osteoclast infiltration in Lyz-TLR4-/- mice than in wild-type mice at day 7. Primary bone marrow–derived cells isolated from TLR4-/- and Lyz-TLR4-/- mice presented enhanced osteoclastogenesis and resorption activity compared with those from wild-type mice. Comparable M0, M1, and M2 macrophage infiltration was found among all groups at days 1, 4, and 7. Conclusions: This study revealed that inactivation of toll-like receptor 4 in myeloid cells enhanced osteoclastogenesis and accelerated healing response during skull repair. Together with the role of toll-like receptor 4 in inflammation-mediated bone destruction, it suggests that toll-like receptor 4 might regulate inflammation-induced osteoclastogenesis under different clinical settings.
Collapse
|
94
|
Liu SP, Wang GD, Du XJ, Wan G, Wu JT, Miao LB, Liang QD. Triptolide inhibits the function of TNF-α in osteoblast differentiation by inhibiting the NF-κB signaling pathway. Exp Ther Med 2017; 14:2235-2240. [PMID: 28962148 DOI: 10.3892/etm.2017.4749] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/20/2017] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation often delays fracture healing or leads to bone nonunion. Effectively suppressing pathological inflammation is crucial for fracture healing or bone remodeling. Triptolide, which is a diterpenoid epoxide, is the major active component of the Thunder God Vine, Tripterygium wilfordii. The aim of the present study was to investigate the role of triptolide in osteoblast differentiation and explore the molecular mechanisms of triptolide in fracture healing. Alkaline phosphatase (ALP) activity was used to evaluate osteoblast differentiation. ALP activity was measured via histochemical staining and western blotting was used to determine the expression of factors associated with inflammation. C2C12 cells were initially treated with 200 ng/ml bone morphogenetic protein (BMP)-2 alone for 3 days, which caused a significant increase in ALP activity (P<0.01). However, treatment with tumor necrosis factor (TNF)-α significantly decreased the ALP activity (P<0.05). Notably, treatment with the chronic inflammatory cytokine TNF-α significantly decreased the effect of BMP-2 in C2C12 cells compared with BMP-2 treatment alone (P<0.01). C2C12 cells were treated with increasing concentrations of BMP-2 or TNF-α for 3 days. The results demonstrated that TNF-α treatment significantly inhibited BMP-2-induced osteoblast differentiation in a dose-dependent manner (P<0.01). The role of triptolide in BMP-2-induced osteoblast differentiation was also examined. Cells were treated with BMP-2, BMP-2 + TNF-α alone, or BMP2 + TNF-α with increasing concentrations of triptolide (4, 8 or 16 ng/ml). After 3 days, the results of ALP activity revealed that triptolide significantly reversed the TNF-α-associated inhibition of osteoblast differentiation (P<0.01). Western blotting analysis demonstrated that triptolide markedly inhibited the phosphorylation of nuclear factor-κB, therefore suppressing the effects of TNF-α. In summary, triptolide is able to reverse the TNF-α-associated suppression of osteoblast differentiation, suggesting that triptolide treatment may have a positive effect on bone remodeling and fracture repairing.
Collapse
Affiliation(s)
- Shen-Peng Liu
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Guo-Dong Wang
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Xue-Jun Du
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Guang Wan
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Jun-Tao Wu
- Department of Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Lian-Bao Miao
- Department of Orthopedics, Huaxian People's Hospital of Henan Province, Anyang, Henan 456400, P.R. China
| | - Qiu-Dong Liang
- Department of Orthopedics, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
95
|
Dixit M, Singh KB, Prakash R, Singh D. Functional block of IL-17 cytokine promotes bone healing by augmenting FOXO1 and ATF4 activity in cortical bone defect model. Osteoporos Int 2017; 28:2207-2220. [PMID: 28341898 DOI: 10.1007/s00198-017-4012-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/15/2017] [Indexed: 01/08/2023]
Abstract
UNLABELLED We determine the effect of interleukin (IL)-17 neutralizing antibody on new bone regeneration. Anti-IL-17 antibody promoted new bone regeneration in cortical bone defect model by augmenting FOXO1 and ATF4 activity thereby decreasing oxidative stress. Our study demonstrates the bone healing and regeneration potential of neutralizing IL-17antibody in osteoporotic fractures. INTRODUCTION The immune system plays important role in the fracture healing process. However, fracture healing is prolonged in disorders associated with systemic inflammation. Fracture healing is decelerated in osteoporosis, condition linked with systemic inflammation. Bone regeneration therapies like recombinant human BMP2 are associated with serious side effects. Studies have been carried out where agents like denosumab and infliximab enhance bone regeneration in osteoporotic conditions. Our previous studies show the osteoprotective and immunoprotective effects of neutralizing IL-17 antibody. Here, we determine the effect of IL-17 neutralizing antibody on new bone regeneration and compare its efficacy with known osteoporotic therapies. METHODS For the study, female BALB/c mice were ovariectomized or sham operated and left for a month followed by a 0.6-mm drill-hole injury in femur mid-diaphysis. The treatment was commenced next day onwards with anti-IL-17, anti-RANKL (Receptor activator of nuclear factor kappa-B ligand), parathyroid hormone (PTH), or alendronate for a period of 3, 10, or 21 days. Animals were then autopsied, and femur bones were dissected out for micro-CT scanning, confocal microscopy, and gene and protein expression studies. RESULTS Micro-CT analysis showed that anti-IL-17 antibody promoted bone healing at days 10 and 21, and the healing effect observed was significantly better than Ovx, anti-RANKL antibody, and ALN, and equal to PTH. Anti-IL-17 also enhanced new bone regeneration as assessed by calcein-labeling studies. Additionally, anti-IL-17 therapy enhanced expression of osteogenic markers and decreased oxidative stress at the injury site. CONCLUSION Overall, our study demonstrates bone healing and regeneration potential of neutralizing IL-17 antibody in osteoporotic fractures.
Collapse
Affiliation(s)
- M Dixit
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, India
| | - K B Singh
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, India
| | - R Prakash
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, India
| | - D Singh
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, India.
| |
Collapse
|
96
|
Manufacturing of Human Extracellular Vesicle-Based Therapeutics for Clinical Use. Int J Mol Sci 2017; 18:ijms18061190. [PMID: 28587212 PMCID: PMC5486013 DOI: 10.3390/ijms18061190] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 05/29/2017] [Accepted: 05/31/2017] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) derived from stem and progenitor cells may have therapeutic effects comparable to their parental cells and are considered promising agents for the treatment of a variety of diseases. To this end, strategies must be designed to successfully translate EV research and to develop safe and efficacious therapies, whilst taking into account the applicable regulations. Here, we discuss the requirements for manufacturing, safety, and efficacy testing of EVs along their path from the laboratory to the patient. Development of EV-therapeutics is influenced by the source cell types and the target diseases. In this article, we express our view based on our experience in manufacturing biological therapeutics for routine use or clinical testing, and focus on strategies for advancing mesenchymal stromal cell (MSC)-derived EV-based therapies. We also discuss the rationale for testing MSC-EVs in selected diseases with an unmet clinical need such as critical size bone defects, epidermolysis bullosa and spinal cord injury. While the scientific community, pharmaceutical companies and clinicians are at the point of entering into clinical trials for testing the therapeutic potential of various EV-based products, the identification of the mode of action underlying the suggested potency in each therapeutic approach remains a major challenge to the translational path.
Collapse
|
97
|
Abstract
BACKGROUND Inflammation is integral to the injury response. The inflammatory response is essential to the host defense against infection and also to tissue regeneration and repair. Toll-like receptors (TLRs) are critical activators of the innate immune response and present attractive therapeutic targets for inflammation-modulated tissue regeneration. The authors' previous study showed that depletion of TLR4 resulted in accelerated skull bone healing concurrent with increased expression of osteoclastogenic genes. As such, in the present study, the authors used various knockout mouse models for TLR4 and its associated signaling mediators as tools to further understand the role of Toll-like receptor-mediated inflammation in calvarial bone healing. METHODS Calvarial defects (1.8-mm diameter) were created in wild-type, TLR4 knockout (TLR4), TLR2, MyD88, TRIF, TLR4 knockout in myeloid cell (Lyz-TLR4), and TLR4 knockout in dendritic-lineage cell (CD11c-TLR4) mice. Bone healing was examined using micro-computed tomographic, histologic, and histomorphometric analyses. RESULTS Micro-computed tomographic and histomorphometric analyses revealed that TLR4-deficient mice (TLR4, Lyz-TLR4, and CD11c-TLR4) exhibited a faster intramembraneous healing response at postoperative day 7, whereas MyD88 and CD11c-TLR4 mice showed enhanced bone healing at day 28. CONCLUSIONS The authors' data suggest a detrimental role for TLR4 in CD11c cells, mediated by Myd88 signaling, during calvarial bone healing. The authors have demonstrated that Toll-like receptor signaling components affect calvarial bone healing, establishing a link between the skeletal and immune systems during craniofacial bone healing. Toll-like receptor signaling components might be used to initiate enhanced healing in bone defects to improve clinical outcomes.
Collapse
|
98
|
Lu Z, Kleine-Nulend J, Li B. Bone Microenvironment, Stem Cells, and Bone Tissue Regeneration. Stem Cells Int 2017; 2017:1315243. [PMID: 28194182 PMCID: PMC5282439 DOI: 10.1155/2017/1315243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 12/26/2016] [Indexed: 01/23/2023] Open
Affiliation(s)
- ZuFu Lu
- Biomaterials and Tissue Engineering Research Unit, School of AMME, University of Sydney, Sydney, NSW 2006, Australia
| | - Jenneke Kleine-Nulend
- Department of Oral Cell Biology, ACTA, University of Amsterdam and VU Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, Netherlands
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, 188 Shizi St., Suzhou, Jiangsu 215006, China
| |
Collapse
|
99
|
Yanez M, Blanchette J, Jabbarzadeh E. Modulation of Inflammatory Response to Implanted Biomaterials Using Natural Compounds. Curr Pharm Des 2017; 23:6347-6357. [PMID: 28521709 PMCID: PMC5681444 DOI: 10.2174/1381612823666170510124348] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/17/2017] [Accepted: 04/28/2017] [Indexed: 02/06/2023]
Abstract
Tissue engineering offers a promising strategy to restore injuries resulting from trauma, infection, tumor resection, or other diseases. In spite of significant progress, the field faces a significant bottleneck; the critical need to understand and exploit the interdependencies of tissue healing, angiogenesis, and inflammation. Inherently, the balance of these interacting processes is affected by a number of injury site conditions that represent a departure from physiological environment, including reduced pH, increased concentration of free radicals, hypoglycemia, and hypoxia. Efforts to harness the potential of immune response as a therapeutic strategy to promote tissue repair have led to identification of natural compounds with significant anti-inflammatory properties. This article provides a concise review of the body's inflammatory response to biomaterials and describes the role of oxygen as a physiological cue in this process. We proceed to highlight the potential of natural compounds to mediate inflammatory response and improve host-graft integration. Herein, we discuss the use of natural compounds to map signaling molecules and checkpoints that regulate the cross-linkage of immune response and skeletal repair.
Collapse
Affiliation(s)
- Maria Yanez
- Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - James Blanchette
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
| | - Ehsan Jabbarzadeh
- Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
- Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208, USA
- Department of Orthopedic Surgery, University of South Carolina School of Medicine, Columbia SC, 29209, USA
| |
Collapse
|
100
|
Smith JT, Schneider AD, Katchko KM, Yun C, Hsu EL. Environmental Factors Impacting Bone-Relevant Chemokines. Front Endocrinol (Lausanne) 2017; 8:22. [PMID: 28261155 PMCID: PMC5306137 DOI: 10.3389/fendo.2017.00022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/25/2017] [Indexed: 01/07/2023] Open
Abstract
Chemokines play an important role in normal bone physiology and the pathophysiology of many bone diseases. The recent increased focus on the individual roles of this class of proteins in the context of bone has shown that members of the two major chemokine subfamilies-CC and CXC-support or promote the formation of new bone and the remodeling of existing bone in response to a myriad of stimuli. These chemotactic molecules are crucial in orchestrating appropriate cellular homing, osteoblastogenesis, and osteoclastogenesis during normal bone repair. Bone healing is a complex cascade of carefully regulated processes, including inflammation, progenitor cell recruitment, differentiation, and remodeling. The extensive role of chemokines in these processes and the known links between environmental contaminants and chemokine expression/activity leaves ample opportunity for disruption of bone healing by environmental factors. However, despite increased clinical awareness, the potential impact of many of these environmental factors on bone-related chemokines is still ill defined. A great deal of focus has been placed on environmental exposure to various endocrine disruptors (bisphenol A, phthalate esters, etc.), volatile organic compounds, dioxins, and heavy metals, though mainly in other tissues. Awareness of the impact of other less well-studied bone toxicants, such as fluoride, mold and fungal toxins, asbestos, and chlorine, is also reviewed. In many cases, the literature on these toxins in osteogenic models is lacking. However, research focused on their effects in other tissues and cell lines provides clues for where future resources could be best utilized. This review aims to serve as a current and exhaustive resource detailing the known links between several classes of high-interest environmental pollutants and their interaction with the chemokines relevant to bone healing.
Collapse
Affiliation(s)
- Justin T. Smith
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - Andrew D. Schneider
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - Karina M. Katchko
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - Chawon Yun
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - Erin L. Hsu
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
- *Correspondence: Erin L. Hsu,
| |
Collapse
|