101
|
Vasconcelos DM, Gonçalves RM, Almeida CR, Pereira IO, Oliveira MI, Neves N, Silva AM, Ribeiro AC, Cunha C, Almeida AR, Ribeiro CC, Gil AM, Seebach E, Kynast KL, Richter W, Lamghari M, Santos SG, Barbosa MA. Fibrinogen scaffolds with immunomodulatory properties promote in vivo bone regeneration. Biomaterials 2016; 111:163-178. [DOI: 10.1016/j.biomaterials.2016.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/30/2016] [Accepted: 10/01/2016] [Indexed: 01/27/2023]
|
102
|
Kim S, Kawai T, Wang D, Yang Y. Engineering a Dual-Layer Chitosan-Lactide Hydrogel To Create Endothelial Cell Aggregate-Induced Microvascular Networks In Vitro and Increase Blood Perfusion In Vivo. ACS APPLIED MATERIALS & INTERFACES 2016; 8:19245-19255. [PMID: 27399928 DOI: 10.1021/acsami.6b04431] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Here, we report the use of chemically cross-linked and photo-cross-linked hydrogels to engineer human umbilical vein endothelial cell (HUVEC) aggregate-induced microvascular networks to increase blood perfusion in vivo. First, we studied the effect of chemically cross-linked and photo-cross-linked chitosan-lactide hydrogels on stiffness, degradation rates, and HUVEC behaviors. The photo-cross-linked hydrogel was relatively stiff (E = ∼15 kPa) and possessed more compact networks, denser surface texture, and lower enzymatic degradation rates than the relatively soft, chemically cross-linked hydrogel (E = ∼2 kPa). While both hydrogels exhibited nontoxicity, the soft chemically cross-linked hydrogels expedited the formation of cell aggregates compared to the photo-cross-linked hydrogels. Cells on the less stiff, chemically cross-linked hydrogels expressed more matrix metalloproteinase (MMP) activity than the stiffer, photo-cross-linked hydrogel. This difference in MMP activity resulted in a more dramatic decrease in mechanical stiffness after 3 days of incubation for the chemically cross-linked hydrogel, as compared to the photo-cross-linked one. After determining the physical and biological properties of each hydrogel, we accordingly engineered a dual-layer hydrogel construct consisting of the relatively soft, chemically cross-linked hydrogel layer for HUVEC encapsulation, and the relatively stiff, acellular, photo-cross-linked hydrogel for retention of cell-laden microvasculature above. This dual-layer hydrogel construct enabled a lasting HUVEC aggregate-induced microvascular network due to the combination of stable substrate, enriched cell adhesion molecules, and extracellular matrix proteins. We tested the dual-layer hydrogel construct in a mouse model of hind-limb ischemia, where the HUVEC aggregate-induced microvascular networks significantly enhanced blood perfusion rate to ischemic legs and decreased tissue necrosis compared with both no treatment and nonaggregated HUVEC-loaded hydrogels within 2 weeks. This study suggests an effective means for regulating hydrogel properties to facilitate a stable, HUVEC aggregate-induced microvascular network for a variety of vascularized tissue applications.
Collapse
Affiliation(s)
| | - Toshiyuki Kawai
- Department of Orthopedic Surgery, Kyoto University , 54 Kawaharacho, Shogoin, Sakyo-ku Kyoto 606-8507, Japan
| | | | | |
Collapse
|
103
|
Yu W, Bien-Aime S, Mattos M, Alsadun S, Wada K, Rogado S, Fiorellini J, Graves D, Uhrich K. Sustained, localized salicylic acid delivery enhances diabetic bone regeneration via prolonged mitigation of inflammation. J Biomed Mater Res A 2016; 104:2595-603. [PMID: 27194511 DOI: 10.1002/jbm.a.35781] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/28/2016] [Accepted: 05/11/2016] [Indexed: 12/19/2022]
Abstract
Diabetes is a metabolic disorder caused by insulin resistance and/or deficiency and impairs bone quality and bone healing due to altered gene expression, reduced vascularization, and prolonged inflammation. No effective treatments for diabetic bone healing are currently available, and most existing treatments do not directly address the diabetic complications that impair bone healing. We recently demonstrated that sustained and localized delivery of salicylic acid (SA) via an SA-based polymer provides a low-cost approach to enhance diabetic bone regeneration. Herein, we report mechanistic studies that delve into the biological action and local pharmacokinetics of SA-releasing polymers shown to enhance diabetic bone regeneration. The results suggest that low SA concentrations were locally maintained at the bone defect site for more than 1 month. As a result of the sustained SA release, a significantly reduced inflammation was observed in diabetic animals, which in turn, yielded reduced osteoclast density and activity, as well as increased osteoblastogenesis. Based upon these results, localized and sustained SA delivery from the SA-based polymer effectively improved bone regeneration in diabetic animals by affecting both osteoclasts and osteoblasts, thereby providing a positive basis for clinical treatments. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2595-2603, 2016.
Collapse
Affiliation(s)
- Weiling Yu
- Department of Biomedical Engineering, Rutgers, the State University of New Jersey, 599 Taylor Road, Piscataway, New Jersey
| | - Stephan Bien-Aime
- Department of Chemistry and Chemical Biology, Rutgers, the State University of New Jersey, 610 Taylor Road, Piscataway, New Jersey
| | - Marcelo Mattos
- Department of Periodontics, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, Pennsylvania
| | - Sarah Alsadun
- Department of Periodontics, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, Pennsylvania
| | - Keisuke Wada
- Department of Periodontics, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, Pennsylvania
| | - Sarah Rogado
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 610 Taylor Road, Piscataway, New Jersey
| | - Joseph Fiorellini
- Department of Periodontics, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, Pennsylvania
| | - Dana Graves
- Department of Periodontics, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, Pennsylvania
| | - Kathryn Uhrich
- Department of Biomedical Engineering, Rutgers, the State University of New Jersey, 599 Taylor Road, Piscataway, New Jersey.,Department of Chemistry and Chemical Biology, Rutgers, the State University of New Jersey, 610 Taylor Road, Piscataway, New Jersey
| |
Collapse
|
104
|
Trombetta R, Inzana JA, Schwarz EM, Kates SL, Awad HA. 3D Printing of Calcium Phosphate Ceramics for Bone Tissue Engineering and Drug Delivery. Ann Biomed Eng 2016; 45:23-44. [PMID: 27324800 DOI: 10.1007/s10439-016-1678-3] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/08/2016] [Indexed: 01/16/2023]
Abstract
Additive manufacturing, also known as 3D printing, has emerged over the past 3 decades as a disruptive technology for rapid prototyping and manufacturing. Vat polymerization, powder bed fusion, material extrusion, and binder jetting are distinct technologies of additive manufacturing, which have been used in a wide variety of fields, including biomedical research and tissue engineering. The ability to print biocompatible, patient-specific geometries with controlled macro- and micro-pores, and to incorporate cells, drugs and proteins has made 3D-printing ideal for orthopaedic applications, such as bone grafting. Herein, we performed a systematic review examining the fabrication of calcium phosphate (CaP) ceramics by 3D printing, their biocompatibility in vitro, and their bone regenerative potential in vivo, as well as their use in localized delivery of bioactive molecules or cells. Understanding the advantages and limitations of the different 3D printing approaches, CaP materials, and bioactive additives through critical evaluation of in vitro and in vivo evidence of efficacy is essential for developing new classes of bone graft substitutes that can perform as well as autografts and allografts or even surpass the performance of these clinical standards.
Collapse
Affiliation(s)
- Ryan Trombetta
- Department of Biomedical Engineering, University of Rochester, Robert B. Goergen Hall, Rochester, NY, 14627, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
| | - Jason A Inzana
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Edward M Schwarz
- Department of Biomedical Engineering, University of Rochester, Robert B. Goergen Hall, Rochester, NY, 14627, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,Department of Orthopedics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Stephen L Kates
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.,Department of Orthopaedic Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Hani A Awad
- Department of Biomedical Engineering, University of Rochester, Robert B. Goergen Hall, Rochester, NY, 14627, USA. .,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA. .,Department of Orthopedics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
105
|
Ying H, Li Q, Zhao C. Interleukin 1β and tumor necrosis factor α promote hFOB1.19 cell viability via activating AP1. Am J Transl Res 2016; 8:2411-2418. [PMID: 27347349 PMCID: PMC4891454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/21/2016] [Indexed: 06/06/2023]
Abstract
Bone trauma healing is a complex physiological process, which may involve the function of various inflammatory cytokines. Our study aimed to explore the roles of inflammatory cytokines in bone trauma healing and reveal the potential mechanism. Concentrations of interleukin (IL)-6, IL-1β and tumor necrosis factor alpha (TNF-α) in peripheral blood serum of bone trauma patients after surgery were determined by ELISA. The human osteoblast hFOB1.19 cell line was cultured to determine the effect of these cytokines in cell viability using MTT assay. In addition, luciferase reporter assay was performed to investigate the activator protein 1 (AP1) transcriptional activity, and small interfering RNA was transfected to inhibit FOS, a component of AP1 molecule. IL-6, IL-1β and TNF-α exhibited higher level in patients with more severe bone traumas after surgery. IL-1β and TNF-α, but not IL-6, induced a significant increase of hFOB1.19 viability after three days of treatment (P < 0.05). IL-1β and TNF-α could activate AP1 transcriptional activity in hFOB1.19 cells (P < 0.001), but the activation was inhibited when cells were pretreated with inhibitor of JNKs, SP600125 (P < 0.001). Besides, the effect of IL-1β and TNF-α on promoting viability was significantly inhibited after knockdown of FOS. These findings indicated that IL-1β and TNF-α played an important role in promoting osteoblast viability via the activation of AP1 transcriptional activity, which was likely to involve the JNK/MAPK signaling pathway. Modulating inflammatory cytokines is a potential strategy for improving the outcome of bone trauma healing.
Collapse
Affiliation(s)
- Hongliang Ying
- Department of Orthopaedic Surgery, China-Japan Unite Hospital of Jilin University Changchun 130033, China
| | - Qiang Li
- Department of Orthopaedic Surgery, China-Japan Unite Hospital of Jilin University Changchun 130033, China
| | - Changfu Zhao
- Department of Orthopaedic Surgery, China-Japan Unite Hospital of Jilin University Changchun 130033, China
| |
Collapse
|
106
|
Loi F, Córdova LA, Pajarinen J, Lin TH, Yao Z, Goodman SB. Inflammation, fracture and bone repair. Bone 2016; 86:119-30. [PMID: 26946132 PMCID: PMC4833637 DOI: 10.1016/j.bone.2016.02.020] [Citation(s) in RCA: 791] [Impact Index Per Article: 87.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/30/2015] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
Abstract
The reconstitution of lost bone is a subject that is germane to many orthopedic conditions including fractures and non-unions, infection, inflammatory arthritis, osteoporosis, osteonecrosis, metabolic bone disease, tumors, and periprosthetic particle-associated osteolysis. In this regard, the processes of acute and chronic inflammation play an integral role. Acute inflammation is initiated by endogenous or exogenous adverse stimuli, and can become chronic in nature if not resolved by normal homeostatic mechanisms. Dysregulated inflammation leads to increased bone resorption and suppressed bone formation. Crosstalk among inflammatory cells (polymorphonuclear leukocytes and cells of the monocyte-macrophage-osteoclast lineage) and cells related to bone healing (cells of the mesenchymal stem cell-osteoblast lineage and vascular lineage) is essential to the formation, repair and remodeling of bone. In this review, the authors provide a comprehensive summary of the literature related to inflammation and bone repair. Special emphasis is placed on the underlying cellular and molecular mechanisms, and potential interventions that can favorably modulate the outcome of clinical conditions that involve bone repair.
Collapse
Affiliation(s)
- Florence Loi
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Luis A Córdova
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA; Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Sergio Livingstone Polhammer 943, Independencia, 8380000 Santiago, Chile.
| | - Jukka Pajarinen
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Tzu-hua Lin
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Zhenyu Yao
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Stuart B Goodman
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA; 300 Pasteur Drive, Edwards Building, Room R114, Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
107
|
Trauma-induced heterotopic bone formation and the role of the immune system: A review. J Trauma Acute Care Surg 2016; 80:156-65. [PMID: 26491794 DOI: 10.1097/ta.0000000000000883] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Extremity trauma, spinal cord injuries, head injuries, and burn injuries place patients at high risk of pathologic extraskeletal bone formation. This heterotopic bone causes severe pain, deformities, and joint contractures. The immune system has been increasingly implicated in this debilitating condition. This review summarizes the various roles immune cells and inflammation play in the formation of ectopic bone and highlights potential areas of future investigation and treatment. Cell types in both the innate and adaptive immune system such as neutrophils, macrophages, mast cells, B cells, and T cells have all been implicated as having a role in ectopic bone formation through various mechanisms. Many of these cell types are promising areas of therapeutic investigation for potential treatment. The immune system has also been known to also influence osteoclastogenesis, which is heavily involved in ectopic bone formation. Chronic inflammation is also known to have an inhibitory role in the formation of ectopic bone, whereas acute inflammation is necessary for ectopic bone formation.
Collapse
|
108
|
Yassin MA, Leknes KN, Sun Y, Lie SA, Finne-Wistrand A, Mustafa K. Surfactant tuning of hydrophilicity of porous degradable copolymer scaffolds promotes cellular proliferation and enhances bone formation. J Biomed Mater Res A 2016; 104:2049-59. [DOI: 10.1002/jbm.a.35741] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/09/2016] [Accepted: 04/06/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Mohammed A. Yassin
- Department of Clinical Dentistry; Faculty of Medicine and Dentistry; University of Bergen; Bergen Norway
| | - Knut N. Leknes
- Department of Clinical Dentistry; Faculty of Medicine and Dentistry; University of Bergen; Bergen Norway
| | - Yang Sun
- Department of Clinical Dentistry; Faculty of Medicine and Dentistry; University of Bergen; Bergen Norway
- Department of Fibre and Polymer Technology; Royal Institute of Technology (KTH); Stockholm Sweden
| | - Stein A. Lie
- Department of Clinical Dentistry; Faculty of Medicine and Dentistry; University of Bergen; Bergen Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology; Royal Institute of Technology (KTH); Stockholm Sweden
| | - Kamal Mustafa
- Department of Clinical Dentistry; Faculty of Medicine and Dentistry; University of Bergen; Bergen Norway
| |
Collapse
|
109
|
Vasconcelos DM, Santos SG, Lamghari M, Barbosa MA. The two faces of metal ions: From implants rejection to tissue repair/regeneration. Biomaterials 2016; 84:262-275. [DOI: 10.1016/j.biomaterials.2016.01.046] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/20/2022]
|
110
|
Jansisyanont P, Tiyapongprapan S, Chuenchompoonut V, Sangvanich P, Thunyakitpisal P. The effect of acemannan sponges in post-extraction socket healing: A randomized trial. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY MEDICINE AND PATHOLOGY 2016. [DOI: 10.1016/j.ajoms.2015.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
111
|
Croes M, Öner FC, van Neerven D, Sabir E, Kruyt MC, Blokhuis TJ, Dhert WJA, Alblas J. Proinflammatory T cells and IL-17 stimulate osteoblast differentiation. Bone 2016; 84:262-270. [PMID: 26780388 DOI: 10.1016/j.bone.2016.01.010] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 01/04/2016] [Accepted: 01/07/2016] [Indexed: 12/15/2022]
Abstract
The local immune response is important to consider when the aim is to improve bone regeneration. Recently T lymphocytes and their associated cytokines have been identified as regulators in fracture callus formation, but it is not known whether T cells affect bone progenitor cells directly. The goal of this in vitro study was to investigate the role of different T cell subsets and their secreted factors on the osteogenic differentiation of human mesenchymal stem cells (MSCs). Significant increases in the alkaline phosphatase activity and the subsequent matrix mineralization by MSCs were found after their exposure to activated T cells or activated T cell-derived conditioned medium. Blocking IFN-γ in the conditioned medium abolished its pro-osteogenic effect, while blocking TGF-β further enhanced osteogenesis. The relative contribution of an anti- or proinflammatory T cell phenotype in MSC osteogenic differentiation was studied next. Enrichment of the fraction of anti-inflammatory regulatory T cells had no beneficial osteogenic effect. In contrast, soluble factors derived from enriched T helper 17 cells upregulated the expression of osteogenic markers by MSCs. IL-17A, and IL-17F, their main proinflammatory cytokines, similarly exhibited strong osteogenic effects when exposed directly to MSCs. IL-17A in particular showed a synergistic action together with bone morphogenetic protein 2. These results indicate that individual T cell subsets, following their activation, affect osteoblast maturation in a different manner through the production of soluble factors. From all T cells, the proinflammatory T cells, including the T helper 17 cells, are most stimulatory for osteogenesis.
Collapse
Affiliation(s)
- Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands.
| | - F Cumhur Öner
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands.
| | - Danihel van Neerven
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands.
| | - Ekrem Sabir
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands.
| | - Moyo C Kruyt
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands.
| | - Taco J Blokhuis
- Department of Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands.
| | - Wouter J A Dhert
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands; Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3508 TD Utrecht, The Netherlands.
| | - Jacqueline Alblas
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, The Netherlands.
| |
Collapse
|
112
|
Suliman S, Sun Y, Pedersen TO, Xue Y, Nickel J, Waag T, Finne‐Wistrand A, Steinmüller‐Nethl D, Krueger A, Costea DE, Mustafa K. In Vivo Host Response and Degradation of Copolymer Scaffolds Functionalized with Nanodiamonds and Bone Morphogenetic Protein 2. Adv Healthc Mater 2016; 5:730-42. [PMID: 26853449 DOI: 10.1002/adhm.201500723] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/30/2015] [Indexed: 12/22/2022]
Abstract
The aim is to evaluate the effect of modifying poly[(l-lactide)-co-(ε-caprolactone)] scaffolds (PLCL) with nanodiamonds (nDP) or with nDP+physisorbed BMP-2 (nDP+BMP-2) on in vivo host tissue response and degradation. The scaffolds are implanted subcutaneously in Balb/c mice and retrieved after 1, 8, and 27 weeks. Molecular weight analysis shows that modified scaffolds degrade faster than the unmodified. Gene analysis at week 1 shows highest expression of proinflammatory markers around nDP scaffolds; although the presence of inflammatory cells and foreign body giant cells is more prominent around the PLCL. Tissue regeneration markers are highly expressed in the nDP+BMP-2 scaffolds at week 8. A fibrous capsule is detectable by week 8, thinnest around nDP scaffolds and at week 27 thickest around PLCL scaffolds. mRNA levels of ALP, COL1α2, and ANGPT1 are significantly upregulating in the nDP+BMP-2 scaffolds at week 1 with ectopic bone seen at week 8. Even when almost 90% of the scaffold is degraded at week 27, nDP are observable at implantation areas without adverse effects. In conclusion, modifying PLCL scaffolds with nDP does not aggravate the host response and physisorbed BMP-2 delivery attenuates inflammation while lowering the dose of BMP-2 to a relatively safe and economical level.
Collapse
Affiliation(s)
- Salwa Suliman
- Department of Clinical Dentistry Center for Clinical Dental Research University of Bergen 5009 Bergen Norway
- Gade Laboratory for Pathology Department of Clinical Medicine University of Bergen 5020 Bergen Norway
- Center for International Health Department of Global Public Health and Primary Care University of Bergen 5009 Bergen Norway
| | - Yang Sun
- Department of Fibre and Polymer Technology KTH Royal Institute of Technology 10044 Stockholm Sweden
| | - Torbjorn O. Pedersen
- Department of Clinical Dentistry Center for Clinical Dental Research University of Bergen 5009 Bergen Norway
| | - Ying Xue
- Department of Clinical Dentistry Center for Clinical Dental Research University of Bergen 5009 Bergen Norway
| | - Joachim Nickel
- Chair Tissue Engineering and Regenerative Medicine University Hospital of Würzburg 97070 Würzburg Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Translational Center “Regenerative Therapies for Oncology and Musculoskeletal Diseases”‐ Würzburg branch D‐97070 Würzburg Germany
| | - Thilo Waag
- Institute of Organic Chemistry University of Würzburg 97074 Würzburg Germany
| | - Anna Finne‐Wistrand
- Department of Fibre and Polymer Technology KTH Royal Institute of Technology 10044 Stockholm Sweden
| | | | - Anke Krueger
- Institute of Organic Chemistry University of Würzburg 97074 Würzburg Germany
| | - Daniela E. Costea
- Gade Laboratory for Pathology Department of Clinical Medicine University of Bergen 5020 Bergen Norway
- Center for International Health Department of Global Public Health and Primary Care University of Bergen 5009 Bergen Norway
- Department of Pathology Hauekeland University Hospital 5020 Bergen Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry Center for Clinical Dental Research University of Bergen 5009 Bergen Norway
| |
Collapse
|
113
|
Liu H, Hao W, Wang X, Su H. miR-23b targets Smad 3 and ameliorates the LPS-inhibited osteogenic differentiation in preosteoblast MC3T3-E1 cells. J Toxicol Sci 2016; 41:185-93. [PMID: 26961602 DOI: 10.2131/jts.41.185] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Hongzhi Liu
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| | - Wei Hao
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| | - Xin Wang
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| | - Hao Su
- Department of Orthopedic Trauma, Yantai Yuhuangding Hospital of Qingdao University, China
| |
Collapse
|
114
|
Allen AB, Zimmermann JA, Burnsed OA, Yakubovich DC, Stevens HY, Gazit Z, McDevitt TC, Guldberg RE. Environmental manipulation to promote stem cell survival in vivo: use of aggregation, oxygen carrier, and BMP-2 co-delivery strategies. J Mater Chem B 2016; 4:3594-3607. [DOI: 10.1039/c5tb02471d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
While mesenchymal stem cell (MSC)-based strategies for critically-sized bone defect repair hold promise, poor cell survival in vivo remains a significant barrier to the translation of these therapeutics.
Collapse
Affiliation(s)
- Ashley B. Allen
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Josh A. Zimmermann
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Olivia A. Burnsed
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Doron Cohn Yakubovich
- Skeletal Biotech Laboratory
- The Hebrew University-Hadassah Faculty of Dental Medicine
- Jerusalem
- Israel
| | - Hazel Y. Stevens
- George W. Woodruff School of Mechanical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Zulma Gazit
- Skeletal Biotech Laboratory
- The Hebrew University-Hadassah Faculty of Dental Medicine
- Jerusalem
- Israel
- Regenerative Medicine Institute
| | - Todd C. McDevitt
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Robert E. Guldberg
- George W. Woodruff School of Mechanical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| |
Collapse
|
115
|
Abstract
Fracture healing is a unique multifaceted process requiring the presence of cells, molecular mediators, and angiogenic factors. The state of inflammation dominates the initial phase, but the ideal magnitude and duration of the process for an optimal outcome remains obscure. Biological response modifiers, such as platelet-rich plasma (PRP) preparations, have been used to reconstitute the desirable early inflammatory state, but the results obtained remain inconclusive. Ongoing research to characterize and quantify the inflammatory response after bone fracture is essential in order to better understand the molecular insights of this localized reaction and to expand our armamentarium in the management of patients with an impaired fracture healing response. Non-steroidal anti-inflammatory drugs frequently administered for analgesia after trauma procedures continue to be a cause of concern for a successful bone repair response.
Collapse
|
116
|
Molinos M, Almeida CR, Caldeira J, Cunha C, Gonçalves RM, Barbosa MA. Inflammation in intervertebral disc degeneration and regeneration. J R Soc Interface 2015; 12:20141191. [PMID: 25673296 DOI: 10.1098/rsif.2014.1191] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is one of the major causes of low back pain, a problem with a heavy economic burden, which has been increasing in prevalence as populations age. Deeper knowledge of the complex spatial and temporal orchestration of cellular interactions and extracellular matrix remodelling is critical to improve current IVD therapies, which have so far proved unsatisfactory. Inflammation has been correlated with degenerative disc disease but its role in discogenic pain and hernia regression remains controversial. The inflammatory response may be involved in the onset of disease, but it is also crucial in maintaining tissue homeostasis. Furthermore, if properly balanced it may contribute to tissue repair/regeneration as has already been demonstrated in other tissues. In this review, we focus on how inflammation has been associated with IVD degeneration by describing observational and in vitro studies as well as in vivo animal models. Finally, we provide an overview of IVD regenerative therapies that target key inflammatory players.
Collapse
Affiliation(s)
- Maria Molinos
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Universidade do Porto, Porto, Portugal
| | - Catarina R Almeida
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal
| | - Joana Caldeira
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal Instituto de Patologia e Imunologia-IPATIMUP, Universidade do Porto, Porto, Portugal
| | - Carla Cunha
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal
| | - Raquel M Gonçalves
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal
| | - Mário A Barbosa
- Instituto de Engenharia Biomédica-INEB, Universidade do Porto, Porto, Portugal Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Universidade do Porto, Porto, Portugal
| |
Collapse
|
117
|
Interleukin-1β in intervertebral disk degeneration. Clin Chim Acta 2015; 450:262-72. [PMID: 26341894 DOI: 10.1016/j.cca.2015.08.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/26/2015] [Accepted: 08/30/2015] [Indexed: 01/06/2023]
Abstract
Intervertebral disk degeneration (IDD) is the most common diagnosis in patients with low back pain, a main cause of musculoskeletal disability in the world. Interleukin-1 (IL-1) β is the most important member of the IL-1 family, and has a strong pro-inflammatory activity by stimulating the secretion of multiple pro-inflammatory mediators. IL-1β is highly expressed in degenerative intervertebral disk (IVD) tissues and cells, and it has been shown to be involved in multiple pathological processes during disk degeneration, including inflammatory responses, matrix destruction, angiogenesis and innervation, cellular apoptosis, oxidative stress and cellular senescence. However, inhibition of IL-1β is found to promote extracellular matrix (ECM) repair and protect against disk regeneration. In this review, after a brief description of IL-1β signaling, we mainly focus on the expression profiles, roles and therapeutic potential of IL-1β in IDD. A better understanding will help develop novel IL-1β-based therapeutic interventions for degenerative disk disease.
Collapse
|
118
|
Do AV, Khorsand B, Geary SM, Salem AK. 3D Printing of Scaffolds for Tissue Regeneration Applications. Adv Healthc Mater 2015; 4:1742-62. [PMID: 26097108 PMCID: PMC4597933 DOI: 10.1002/adhm.201500168] [Citation(s) in RCA: 512] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/26/2015] [Indexed: 12/21/2022]
Abstract
The current need for organ and tissue replacement, repair, and regeneration for patients is continually growing such that supply is not meeting demand primarily due to a paucity of donors as well as biocompatibility issues leading to immune rejection of the transplant. In order to overcome these drawbacks, scientists have investigated the use of scaffolds as an alternative to transplantation. These scaffolds are designed to mimic the extracellular matrix (ECM) by providing structural support as well as promoting attachment, proliferation, and differentiation with the ultimate goal of yielding functional tissues or organs. Initial attempts at developing scaffolds were problematic and subsequently inspired an interest in 3D printing as a mode for generating scaffolds. Utilizing three-dimensional printing (3DP) technologies, ECM-like scaffolds can be produced with a high degree of complexity, where fine details can be included at a micrometer level. In this Review, the criteria for printing viable and functional scaffolds, scaffolding materials, and 3DP technologies used to print scaffolds for tissue engineering are discussed. Creating biofunctional scaffolds could potentially help to meet the demand by patients for tissues and organs without having to wait or rely on donors for transplantation.
Collapse
Affiliation(s)
- Anh-Vu Do
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Behnoush Khorsand
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Sean M Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
119
|
Croes M, Oner FC, Kruyt MC, Blokhuis TJ, Bastian O, Dhert WJA, Alblas J. Proinflammatory Mediators Enhance the Osteogenesis of Human Mesenchymal Stem Cells after Lineage Commitment. PLoS One 2015; 10:e0132781. [PMID: 26176237 PMCID: PMC4503569 DOI: 10.1371/journal.pone.0132781] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 06/18/2015] [Indexed: 01/09/2023] Open
Abstract
Several inflammatory processes underlie excessive bone formation, including chronic inflammation of the spine, acute infections, or periarticular ossifications after trauma. This suggests that local factors in these conditions have osteogenic properties. Mesenchymal stem cells (MSCs) and their differentiated progeny contribute to bone healing by synthesizing extracellular matrix and inducing mineralization. Due to the variation in experimental designs used in vitro, there is controversy about the osteogenic potential of proinflammatory factors on MSCs. Our goal was to determine the specific conditions allowing the pro-osteogenic effects of distinct inflammatory stimuli. Human bone marrow MSCs were exposed to tumor necrosis factor alpha (TNF-α) and lipopolysaccharide (LPS). Cells were cultured in growth medium or osteogenic differentiation medium. Alternatively, bone morphogenetic protein 2 (BMP-2) was used as osteogenic supplement to simulate the conditions in vivo. Alkaline phosphatase activity and calcium deposition were indicators of osteogenicity. To elucidate lineage commitment-dependent effects, MSCs were pre-differentiated prior treatment. Our results show that TNF-α and LPS do not affect the expression of osteogenic markers by MSCs in the absence of an osteogenic supplement. In osteogenic differentiation medium or together with BMP-2 however, these mediators highly stimulated their alkaline phosphatase activity and subsequent matrix mineralization. In pre-osteoblasts, matrix mineralization was significantly increased by these mediators, but irrespective of the culture conditions. Our study shows that inflammatory factors potently enhance the osteogenic capacity of MSCs. These properties may be harnessed in bone regenerative strategies. Importantly, the commitment of MSCs to the osteogenic lineage greatly enhances their responsiveness to inflammatory signals.
Collapse
Affiliation(s)
- Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - F. Cumhur Oner
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Moyo C. Kruyt
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Taco J. Blokhuis
- Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Okan Bastian
- Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wouter J. A. Dhert
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jacqueline Alblas
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- * E-mail:
| |
Collapse
|
120
|
Interactions between MSCs and immune cells: implications for bone healing. J Immunol Res 2015; 2015:752510. [PMID: 26000315 PMCID: PMC4427002 DOI: 10.1155/2015/752510] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/18/2015] [Accepted: 01/19/2015] [Indexed: 02/07/2023] Open
Abstract
It is estimated that, of the 7.9 million fractures sustained in the United States each year, 5% to 20% result in delayed or impaired healing requiring therapeutic intervention. Following fracture injury, there is an initial inflammatory response that plays a crucial role in bone healing; however, prolonged inflammation is inhibitory for fracture repair. The precise spatial and temporal impact of immune cells and their cytokines on fracture healing remains obscure. Some cytokines are reported to be proosteogenic while others inhibit bone healing. Cell-based therapy utilizing mesenchymal stromal cells (MSCs) is an attractive option for augmenting the fracture repair process. Osteoprogenitor MSCs not only differentiate into bone, but they also exert modulatory effects on immune cells via a variety of mechanisms. In this paper, we review the current literature on both in vitro and in vivo studies on the role of the immune system in fracture repair, the use of MSCs in the enhancement of fracture healing, and interactions between MSCs and immune cells. Insight into this paradigm can provide valuable clues in identifying cellular and noncellular targets that can potentially be modulated to enhance both natural bone healing and bone repair augmented by the exogenous addition of MSCs.
Collapse
|
121
|
Wang C, Shen J, Yukata K, Inzana JA, O'Keefe RJ, Awad HA, Hilton MJ. Transient gamma-secretase inhibition accelerates and enhances fracture repair likely via Notch signaling modulation. Bone 2015; 73:77-89. [PMID: 25527421 PMCID: PMC4336841 DOI: 10.1016/j.bone.2014.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/05/2014] [Accepted: 12/11/2014] [Indexed: 12/21/2022]
Abstract
Approximately 10% of skeletal fractures result in healing complications and non-union, while most fractures repair with appropriate stabilization and without pharmacologic intervention. It is the latter injuries that cannot be underestimated as the expenses associated with their treatment and subsequent lost productivity are predicted to increase to over $74 billion by 2015. During fracture repair, local mesenchymal stem/progenitor cells (MSCs) differentiate to form new cartilage and bone, reminiscent of events during skeletal development. We previously demonstrated that permanent loss of gamma-secretase activity and Notch signaling accelerates bone and cartilage formation from MSC progenitors during skeletal development, leading to pathologic acquisition of bone and depletion of bone marrow derived MSCs. Here, we investigated whether transient and systemic gamma-secretase and Notch inhibition is capable of accelerating and enhancing fracture repair by promoting controlled MSC differentiation near the fracture site. Our radiographic, microCT, histological, cell and molecular analyses reveal that single and intermittent gamma-secretase inhibitor (GSI) treatments significantly enhance cartilage and bone callus formation via the promotion of MSC differentiation, resulting in only a moderate reduction of local MSCs. Biomechanical testing further demonstrates that GSI treated fractures exhibit superior strength earlier in the healing process, with single dose GSI treated fractures exhibiting bone strength approaching that of un-fractured tibiae. These data further establish that transient inhibition of gamma-secretase activity and Notch signaling temporarily increases osteoclastogenesis and accelerates bone remodeling, which coupled with the effects on MSCs likely explains the accelerated and enhanced fracture repair. Therefore, we propose that the Notch pathway serves as an important therapeutic target during skeletal fracture repair.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jie Shen
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kiminori Yukata
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jason A Inzana
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Regis J O'Keefe
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hani A Awad
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Matthew J Hilton
- Department of Orthopaedics and Rehabilitation, The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
122
|
Gonçalves RM, Pereira ACL, Pereira IO, Oliveira MJ, Barbosa MA. Macrophage response to chitosan/poly-(γ-glutamic acid) nanoparticles carrying an anti-inflammatory drug. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:167. [PMID: 25791458 DOI: 10.1007/s10856-015-5496-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/14/2015] [Indexed: 06/04/2023]
Abstract
The inflammatory response to biomaterials, traditionally viewed as detrimental, is nowadays considered essential for tissue repair/regeneration, being macrophages recognized as the key players in resolving inflammation. Here, the preparation of chitosan (Ch)/poly-(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) as vehicle for a non-steroid anti-inflammatory drug, diclofenac (Df), is described and the response of primary human macrophages to this system is evaluated. Df was incorporated in Ch/γ-PGA NPs at controlled pH (5.0) (maximum 0.05 mg/ml). The components molar ratio and order of addition revealed to be critical to obtain NPs (315 ± 50 nm with 0.36 ± 0.06 polydispersion index). Df was released at physiological pH and this drug-delivery system was proved to be non toxic to macrophages, being rapidly internalized (95 %). Importantly, efficacy of Df-NPs was confirmed by their ability of inhibit/revert PGE2 production of activated macrophages. Therefore, Df-NPs could contribute to stifle local inflammatory reactions, namely those associated with biomaterials.
Collapse
|
123
|
Abstract
BACKGROUND AND PURPOSE NSAIDs are commonly used in the clinic, and there is a general perception that this does not influence healing in common types of human fractures. Still, NSAIDs impair fracture healing dramatically in animal models. These models mainly pertain to fractures of cortical bone in shafts, whereas patients more often have corticocancellous fractures in metaphyses. We therefore tested the hypothesis that the effect of an NSAID is different in shaft healing and metaphyseal healing. METHODS 26 mice were given an osteotomy of their left femur with an intramedullary nail. 13 received injections of indomethacin, 1 mg/kg twice daily. After 17 days of healing, the femurs were analyzed with 3-point bending and microCT. 24 other mice had holes drilled in both proximal tibias, to mimic a stable metaphyseal injury. A screw was inserted in the right tibial hole only. After 7 days of indomethacin injections or control injections, screw fixation was measured with mechanical pull-out testing and the side without a screw was analyzed with microCT. RESULTS In the shaft model, indomethacin led to a 35% decrease in force at failure (95% CI: 14-54). Callus size was reduced to a similar degree, as seen by microCT. Metaphyseal healing was less affected by indomethacin, as no effect on pull-out force could be seen (95% CI: -27 to 17) and there was only a small drop in new bone volume inside the drill hole. The difference in the relative effect of indomethacin between the 2 models was statistically significant (p = 0.006). INTERPRETATION Indomethacin had a minimal effect on stable metaphyseal fractures, but greatly impaired healing of unstable shaft fractures. This could explain some of the differences found between animal models and clinical experience.
Collapse
Affiliation(s)
- Olof Sandberg
- Orthopedics Section, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Per Aspenberg
- Orthopedics Section, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
124
|
Schmidt-Bleek K, Kwee BJ, Mooney DJ, Duda GN. Boon and Bane of Inflammation in Bone Tissue Regeneration and Its Link with Angiogenesis. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:354-64. [PMID: 25742724 DOI: 10.1089/ten.teb.2014.0677] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Delayed healing or nonhealing of bone is an important clinical concern. Although bone, one of the two tissues with scar-free healing capacity, heals in most cases, healing is delayed in more than 10% of clinical cases. Treatment of such delayed healing condition is often painful, risky, time consuming, and expensive. Tissue healing is a multistage regenerative process involving complex and well-orchestrated steps, which are initiated in response to injury. At best, these steps lead to scar-free tissue formation. At the onset of healing, during the inflammatory phase, stationary and attracted macrophages and other immune cells at the fracture site release cytokines in response to injury. This initial reaction to injury is followed by the recruitment, proliferation, and differentiation of mesenchymal stromal cells, synthesis of extracellular matrix proteins, angiogenesis, and finally tissue remodeling. Failure to heal is often associated with poor revascularization. Since blood vessels mediate the transport of circulating cells, oxygen, nutrients, and waste products, they appear essential for successful healing. The strategy of endogenous regeneration in a tissue such as bone is interesting to analyze since it may represent a blueprint of successful tissue formation. This review highlights the interdependency of the time cascades of inflammation, angiogenesis, and tissue regeneration. A better understanding of these inter-relations is mandatory to early identify patients at risk as well as to overcome critical clinical conditions that limit healing. Instead of purely tolerating the inflammatory phase, modulations of inflammation (immunomodulation) might represent a valid therapeutic strategy to enhance angiogenesis and foster later phases of tissue regeneration.
Collapse
Affiliation(s)
- Katharina Schmidt-Bleek
- 1 Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Brian J Kwee
- 3 The Mooney Lab: Laboratory for Cell and Tissue Engineering, Harvard-School of Engineering and Applied Sciences , Cambridge, Massachusetts
| | - David J Mooney
- 3 The Mooney Lab: Laboratory for Cell and Tissue Engineering, Harvard-School of Engineering and Applied Sciences , Cambridge, Massachusetts
| | - Georg N Duda
- 1 Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| |
Collapse
|
125
|
Aydin A, Halici Z, Albayrak A, Polat B, Karakus E, Yildirim OS, Bayir Y, Cadirci E, Ayan AK, Aksakal AM. Treatment with Carnitine Enhances Bone Fracture Healing under Osteoporotic and/or Inflammatory Conditions. Basic Clin Pharmacol Toxicol 2015; 117:173-9. [PMID: 25625309 DOI: 10.1111/bcpt.12384] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/12/2015] [Indexed: 01/20/2023]
Abstract
The aim of this study was to examine the effects of carnitine on bone healing in ovariectomy (OVX) and inflammation (INF)-induced osteoporotic rats. The rats were randomly divided into nine groups (n = 8 animals per group): sham-operated (Group 1: SHAM); sham + magnesium silicate (Mg-silicate) (Group 2: SHAM + INF); ovariectomy (Group 3: OVX); ovariectomy + femoral fracture (Group 4: OVX + FRC); ovariectomy + femoral fracture + Mg-silicate (Group 5: OVX + FRC + INF); ovariectomy + femoral fracture + carnitine 50 mg/kg (Group 6: OVX + FRC + CAR50); ovariectomy + femoral fracture + carnitine 100 mg/kg (Group 7: OVX + FRC + CAR100); ovariectomy + femoral fracture + Mg-silicate + carnitine 50 mg/kg (Group 8: OVX + FRC + INF + CAR50); and ovariectomy + femoral fracture + Mg-silicate + carnitine 100 mg/kg (Group 9: OVX + FRC + INF + CAR100). Eight weeks after OVX, which allowed for osteoporosis to develop, INF was induced with subcutaneous Mg-silicate. On day 80, all of the rats in groups 4-9 underwent fracture operation on the right femur. Bone mineral density (BMD) showed statistically significant improvements in the treatment groups. The serum markers of bone turnover (osteocalcin and osteopontin) and pro-inflammatory cytokines (tumour necrosis factor α, interleukin 1β and interleukin 6) were decreased in the treatment group. The X-ray images showed significantly increased callus formation and fracture healing in the groups treated with carnitine. The present results show that in a rat model with osteoporosis induced by ovariectomy and Mg-silicate, treatment with carnitine improves the healing of femur fractures.
Collapse
Affiliation(s)
- Ali Aydin
- Department of Orthopedics and Traumatology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Zekai Halici
- Department of Pharmacology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Abdulmecit Albayrak
- Department of Pharmacology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Beyzagul Polat
- Department of Pharmacology, Ataturk University Faculty of Pharmacy, Erzurum, Turkey
| | - Emre Karakus
- Department of Pharmacology, Ataturk University Faculty of Veterinary Medicine, Erzurum, Turkey
| | - Omer Selim Yildirim
- Department of Orthopedics and Traumatology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Yasin Bayir
- Department of Biochemistry, Ataturk University Faculty of Pharmacy, Erzurum, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Arif Kursad Ayan
- Department of Nuclear Medicine, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Ahmet Murat Aksakal
- Department of Orthopedics and Traumatology, Sevket Yilmaz Education and Research Hospital, Bursa, Turkey
| |
Collapse
|
126
|
Peric M, Dumic-Cule I, Grcevic D, Matijasic M, Verbanac D, Paul R, Grgurevic L, Trkulja V, Bagi CM, Vukicevic S. The rational use of animal models in the evaluation of novel bone regenerative therapies. Bone 2015; 70:73-86. [PMID: 25029375 DOI: 10.1016/j.bone.2014.07.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/30/2014] [Accepted: 07/05/2014] [Indexed: 12/31/2022]
Abstract
Bone has a high potential for endogenous self-repair. However, due to population aging, human diseases with impaired bone regeneration are on the rise. Current strategies to facilitate bone healing include various biomolecules, cellular therapies, biomaterials and different combinations of these. Animal models for testing novel regenerative therapies remain the gold standard in pre-clinical phases of drug discovery and development. Despite improvements in animal experimentation, excessive poorly designed animal studies with inappropriate endpoints and inaccurate conclusions are being conducted. In this review, we discuss animal models, procedures, methods and technologies used in bone repair studies with the aim to assist investigators in planning and performing scientifically sound experiments that respect the wellbeing of animals. In the process of designing an animal study for bone repair investigators should consider: skeletal characteristics of the selected animal species; a suitable animal model that mimics the intended clinical indication; an appropriate assessment plan with validated methods, markers, timing, endpoints and scoring systems; relevant dosing and statistically pre-justified sample sizes and evaluation methods; synchronization of the study with regulatory requirements and additional evaluations specific to cell-based approaches. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Mihaela Peric
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia.
| | - Ivo Dumic-Cule
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia
| | - Danka Grcevic
- University of Zagreb School of Medicine, Department of Physiology and Immunology, Salata 3, Zagreb, Croatia
| | - Mario Matijasic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia
| | - Donatella Verbanac
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia
| | - Ruth Paul
- Paul Regulatory Services Ltd, Fisher Hill Way, Cardiff CF15 8DR, UK
| | - Lovorka Grgurevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia
| | - Vladimir Trkulja
- University of Zagreb School of Medicine, Department of Pharmacology, Salata 11, Zagreb, Croatia
| | - Cedo M Bagi
- Pfizer Inc., Global Research and Development, Global Science and Technology, 100 Eastern Point Road, Groton, CT 06340, USA
| | - Slobodan Vukicevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia.
| |
Collapse
|
127
|
Abstract
A review of how the geometrical design of scaffolds influences the bone tissue regeneration process.
Collapse
Affiliation(s)
- Amir A. Zadpoor
- Department of Biomechanical Engineering
- Faculty of Mechanical
- Maritime
- and Materials Engineering
- Delft University of Technology (TU Delft)
| |
Collapse
|
128
|
Bartaula-Brevik S, Pedersen TO, Blois AL, Papadakou P, Finne-Wistrand A, Xue Y, Bolstad AI, Mustafa K. Leukocyte transmigration into tissue-engineered constructs is influenced by endothelial cells through Toll-like receptor signaling. Stem Cell Res Ther 2014; 5:143. [PMID: 25528303 PMCID: PMC4445275 DOI: 10.1186/scrt533] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Inflammation plays a crucial role in tissue regeneration, wound healing, and the success of tissue-engineered constructs. The aim of this study was to investigate the influence of human umbilical vein endothelial cells (ECs) on leukocyte transmigration when co-cultured with primary human bone marrow-derived multipotent stromal cells (MSCs). METHODS MSCs with and without ECs were cultured in poly (L-lactide-co-1, 5-dioxepan-2-one) (poly (LLA-co-DXO)) scaffolds for 1 week in vitro in a bioreactor system, after which they were implanted subcutaneously in non-obese diabetic/severe combined immunodeficient mice. After 1 and 3 weeks, scaffolds were retrieved, and the mRNA expression of interleukin 1-beta (IL-1β), IL-6, IL-10, hypoxia-inducible factor 1-alpha (HIF-1α), HIF-1β, and mammalian target of rapamycin was examined by real-time reverse transcription-polymerase chain reaction. Furthermore, immunofluorescent staining was performed for IL-1β, IL-6, neutrophils, and CD11b. In addition, Western blotting was done for IL-1β and IL-6. Leukocyte transmigration genes and genes in Toll-like receptor pathways, expressed by MSCs cultured in vitro with or without ECs, were further investigated with a microarray dataset. RESULTS In vitro, genes involved in leukocyte transmigration and Toll-like receptor pathways were clearly influenced by the addition of ECs. Platelet/endothelial cell adhesion molecule-1 (PECAM-1) and cadherin-5 (CDH5), both genes involved in leukocyte transmigration, were expressed significantly higher in the MSC/EC group. CONCLUSIONS The recruitment of leukocytes into tissue-engineered constructs with MSCs is strongly influenced by the addition of ECs via activation of leukocyte transmigration and Toll-like receptor pathways.
Collapse
Affiliation(s)
- Sushma Bartaula-Brevik
- />Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009 Bergen, Norway
| | - Torbjorn O Pedersen
- />Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009 Bergen, Norway
- />Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Anna L Blois
- />Centre for Cancer Biomarkers, Department of Clinical Medicine, Section for Pathology, University of Bergen, Jonas Lies vei 91B, 5021 Bergen, Norway
- />Children’s Hospital Boston, Vascular Biology Department, Harvard Medical School, 300 Longwood Avenue, Boston, MA USA
| | - Panagiota Papadakou
- />Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Anna Finne-Wistrand
- />Department of Fiber and Polymer Technology, KTH Royal Institute of Technology, Teknikringen 42, SE-100 44 Stockholm, Sweden
| | - Ying Xue
- />Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009 Bergen, Norway
| | - Anne Isine Bolstad
- />Department of Clinical Dentistry - Periodontics, University of Bergen, Årstadveien 19, 5009 Bergen, Norway
| | - Kamal Mustafa
- />Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009 Bergen, Norway
| |
Collapse
|
129
|
Tseng CH, Huang WS, Li TC, Chen HJ, Muo CH, Kao CH. Increased risk of end-stage renal disease among hip fracture patients. Eur J Intern Med 2014; 25:956-61. [PMID: 25468738 DOI: 10.1016/j.ejim.2014.10.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/11/2014] [Accepted: 10/21/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Inflammation-related microvasculr disease, albuminuria, and rapid deterioration of renal function can accelerate the development of end-stage renal disease (ESRD). The role of hip fracture (HFr), a disorder that involves inflammation, in the development of ESRD has not been fully investigated. This study explored whether HFr increases the risk of ESRD. METHODS Taiwan National Health Insurance inpatient claims were used to identify 83,550 patients newly diagnosed with HFr from 2000 to 2006, and 83,550 age- and sex-matched patients without HFr were randomly selected for comparison. Hazards of ESRD combined with HFr, comorbidities, including hypertension, hyperlipidemia, peripheral arterial disease, osteoporosis and asthma, and general health status, with Charlson comorbidity index (CCI), were assessed using data to the end of 2011. RESULTS ESRD risk was 1.42-fold higher (95% confidence interval [CI]:1.29-1.33) in the HFr cohort than in the control group, which was computed using the Cox proportional model. Age-specific analysis revealed that the adjusted hazard ratios (aHRs) of ESRD for HFr patients increased slightly as age increased, with an aHR of 1.56 (95% CI:1.35-1.81) for patients 65-74 years old, which gradually decreased to 0.88 (95% CI:0.66-1.18) for patients ≥ 85 years old. ESRD risk increased as HFr severity increased, with an aHR of 6.71 (95% CI:5.90-7.63) for patients with severe HFr. CONCLUSION This study is the first to report that HFr, in combination with underlying osteoporosis-related chronic illness, microvascular disease and chronic inflammation, is associated with an increased risk of ESRD, particularly among relatively younger people.
Collapse
Affiliation(s)
- Chun-Hung Tseng
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Clinical Medical Science and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Shih Huang
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Clinical Medical Science and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Tsai-Chung Li
- Graduate Institute of Biostatistics, College of Management, China Medical University, Taichung, Taiwan; Department of Healthcare Administration, College of Health Science, Asia University, Taichung, Taiwan
| | - Hsuan-Ju Chen
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Muo
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Hung Kao
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Clinical Medical Science and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
130
|
Kim S, Bedigrew K, Guda T, Maloney WJ, Park S, Wenke JC, Yang YP. Novel osteoinductive photo-cross-linkable chitosan-lactide-fibrinogen hydrogels enhance bone regeneration in critical size segmental bone defects. Acta Biomater 2014; 10:5021-5033. [PMID: 25174669 DOI: 10.1016/j.actbio.2014.08.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 02/05/2023]
Abstract
The purpose of this study was to develop and characterize a novel photo-cross-linkable chitosan-lactide-fibrinogen (CLF) hydrogel and evaluate the efficacy of bone morphogenetic protein-2 (BMP-2) containing a CLF hydrogel for osteogenesis in vitro and in vivo. We synthesized the CLF hydrogels and characterized their chemical structure, degradation rate, compressive modulus and in vitro BMP-2 release kinetics. We evaluated bioactivities of the BMP-2 containing CLF hydrogels (0, 50, 100 and 500ngml(-1)) in vitro using W-20-17 preosteoblast mouse bone marrow stromal cells and C2C12 mouse myoblast cells. The effect of BMP-2 containing CLF gels (0, 0.5, 1, 2 and 5μg) on bone formation was evaluated using rat critical size segmental bone defects for 4weeks. Fourier transform infrared spectroscopy spectra and scanning electron microscopy images showed chemical and structural changes by the addition of fibrinogen into the chitosan-lactide copolymer. The incorporation of fibrinogen molecules significantly increased the compressive modulus of the hydrogels. The in vitro BMP-2 release study showed initial burst releases from the CLF hydrogels followed by sustained releases, regardless of the concentration of the BMP-2 over 4weeks. Cells in all groups were viable in the presence of the hydrogels regardless of BMP-2 doses, indicating non-cytotoxicity of hydrogels. Alkaline phosphate activity and mineralization of cells exhibited dose dependence on BMP-2 containing CLF hydrogels. Radiography, microcomputed tomography and histology confirmed that the BMP-2 containing CLF hydrogels prompted neo-osteogenesis and accelerated healing of the defects in a dose-dependent manner. Thus the CLF hydrogel is a promising delivery system of growth factors for bone regeneration.
Collapse
|
131
|
Cruz ACC, Caon T, Menin Á, Granato R, Boabaid F, Simões CMO. Adipose-derived stem cells incorporated into platelet-rich plasma improved bone regeneration and maturation in vivo. Dent Traumatol 2014; 31:42-8. [PMID: 25336206 DOI: 10.1111/edt.12134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND/AIM Some cases of tooth loss related to dental trauma require bone-grafting procedures to improve the aesthetics before prosthetic rehabilitation or to enable the installation of dental implants. Bone regeneration is often a challenge and could be largely improved by mesenchymal stem cells therapy. However, the appropriate scaffold for these cells still a problem. This study evaluated the in vivo effect of human adipose-derived stem cells incorporated into autogenous platelet-rich plasma in bone regeneration and maturation. MATERIAL AND METHODS Adipose-derived stem cells were isolated from lipoaspirate tissues and used at passage 4. Immunophenotyping and multilineage differentiation of cells were performed and mesenchymal stem cells characteristics confirmed. Bicortical bone defects (10 mm diameter) were created in the tibia of six beagle dogs to evaluate the effect of adipose-derived stem cells incorporated into platelet-rich plasma scaffolds, platelet-rich plasma alone, autogenous bone grafts, and clot. Samples were removed 6 weeks postsurgeries and analyzed by quantification of primary and secondary bone formation and granulation tissue. RESULTS Adipose-derived stem cells incorporated into platelet-rich plasma scaffolds promoted the highest bone formation (primary + secondary bone) (P < 0.001), the highest bone maturation (secondary bone) (P < 0.001), and the lowest amount of granulation tissue (P < 0.001). CONCLUSIONS Adipose-derived stem cells incorporated into platelet-rich plasma scaffolds promote more bone formation and maturation, and less granulation tissue in bone defects created in canine tibia. Therefore, platelet-rich plasma can be considered as a candidate scaffold for adipose-derived stem cells to promote bone regeneration.
Collapse
|
132
|
Hutton DL, Kondragunta R, Moore EM, Hung BP, Jia X, Grayson WL. Tumor necrosis factor improves vascularization in osteogenic grafts engineered with human adipose-derived stem/stromal cells. PLoS One 2014; 9:e107199. [PMID: 25248109 PMCID: PMC4172477 DOI: 10.1371/journal.pone.0107199] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 08/13/2014] [Indexed: 11/19/2022] Open
Abstract
The innate immune response following bone injury plays an important role in promoting cellular recruitment, revascularization, and other repair mechanisms. Tumor necrosis factor-α (TNF) is a prominent pro-inflammatory cytokine in this cascade, and has been previously shown to improve bone formation and angiogenesis in a dose- and timing-dependent manner. This ability to positively impact both osteogenesis and vascular growth may benefit bone tissue engineering, as vasculature is essential to maintaining cell viability in large grafts after implantation. Here, we investigated the effects of exogenous TNF on the induction of adipose-derived stem/stromal cells (ASCs) to engineer pre-vascularized osteogenic tissue in vitro with respect to dose, timing, and co-stimulation with other inflammatory mediators. We found that acute (2-day), low-dose exposure to TNF promoted vascularization, whereas higher doses and continuous exposure inhibited vascular growth. Co-stimulation with platelet-derived growth factor (PDGF), another key factor released following bone injury, increased vascular network formation synergistically with TNF. ASC-seeded grafts were then cultured within polycaprolactone-fibrin composite scaffolds and implanted in nude rats for 2 weeks, resulting in further tissue maturation and increased angiogenic ingrowth in TNF-treated grafts. VEGF-A expression levels were significantly higher in TNF-treated grafts immediately prior to implantation, indicating a long-term pro-angiogenic effect. These findings demonstrate that TNF has the potential to promote vasculogenesis in engineered osteogenic grafts both in vitro and in vivo. Thus, modulation and/or recapitulation of the immune response following bone injury may be a beneficial strategy for bone tissue engineering.
Collapse
Affiliation(s)
- Daphne L. Hutton
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Renu Kondragunta
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Erika M. Moore
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ben P. Hung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Xiaofeng Jia
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Warren L. Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Material Sciences and Engineering, Johns Hopkins University School of Engineering, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
133
|
Schmidt-Bleek K, Petersen A, Dienelt A, Schwarz C, Duda GN. Initiation and early control of tissue regeneration - bone healing as a model system for tissue regeneration. Expert Opin Biol Ther 2014; 14:247-59. [PMID: 24397854 DOI: 10.1517/14712598.2014.857653] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Tissue regeneration in itself is a fascinating process that promises repeated renewal of tissue and organs. AREAS COVERED This article aims to illustrate the different strategies available to control tissue regeneration at a very early stage, using bone as an exemplary tissue. The aspects of a controlled inflammatory cascade to achieve a balanced immune response, cell therapeutic approaches for improved tissue formation and angiogenesis, guiding the organization of newly formed extracellular matrix by biomaterials, the relevance of mechanical signals for tissue regeneration processes, and the chances and limitations of growth factor treatments are discussed. EXPERT OPINION The currently available knowledge is reviewed and perspectives for potential new targets are given. This is done under the assumption that early identification of risk patients as well as the application of early intervention strategies is possible.
Collapse
Affiliation(s)
- Katharina Schmidt-Bleek
- Charité - Universitätsmedizin Berlin, Julius Wolff Institut and Center for Musculoskeletal Surgery , Augustenburger Platz 1, D-13353 Berlin , Germany +49 30 450 536196 ; +49 30 450 559969 ;
| | | | | | | | | |
Collapse
|
134
|
Abou-Khalil R, Colnot C. Cellular and molecular bases of skeletal regeneration: what can we learn from genetic mouse models? Bone 2014; 64:211-21. [PMID: 24709685 DOI: 10.1016/j.bone.2014.03.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 03/19/2014] [Accepted: 03/26/2014] [Indexed: 10/25/2022]
Abstract
Although bone repairs through a very efficient regenerative process in 90% of the patients, many factors can cause delayed or impaired healing. To date, there are no reliable biological parameters to predict or diagnose bone repair defects. Orthopedic surgeons mostly base their diagnoses on radiographic analyses. With the recent progress in our understanding of the bone repair process, new methods may be envisioned. Animal models have allowed us to define the key steps of bone regeneration and the biological and mechanical factors that may influence bone healing in positive or negative ways. Most importantly, small animal models such as mice have provided powerful tools to apprehend the genetic bases of normal and impaired bone healing. The current review presents a state of the art of the genetically modified mouse models that have advanced our understanding of the cellular and molecular components of bone regeneration and repair. The review illustrates the use of these models to define the role of inflammation, skeletal cell lineages, signaling pathways, the extracellular matrix, osteoclasts and angiogenesis. These genetic mouse models promise to change the field of orthopedic surgery to help establish genetic predispositions for delayed repair, develop models of non-union that mimic the human conditions and elaborate new therapeutic approaches to enhance bone regeneration.
Collapse
Affiliation(s)
- Rana Abou-Khalil
- INSERM UMR1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Céline Colnot
- INSERM UMR1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France.
| |
Collapse
|
135
|
Könnecke I, Serra A, El Khassawna T, Schlundt C, Schell H, Hauser A, Ellinghaus A, Volk HD, Radbruch A, Duda GN, Schmidt-Bleek K. T and B cells participate in bone repair by infiltrating the fracture callus in a two-wave fashion. Bone 2014; 64:155-65. [PMID: 24721700 DOI: 10.1016/j.bone.2014.03.052] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/22/2014] [Accepted: 03/30/2014] [Indexed: 12/12/2022]
Abstract
Fracture healing is a regenerative process in which bone is restored without scar tissue formation. The healing cascade initiates with a cycle of inflammation, cell migration, proliferation and differentiation. Immune cells invade the fracture site immediately upon bone damage and contribute to the initial phase of the healing process by recruiting accessory cells to the injury site. However, little is known about the role of the immune system in the later stages of fracture repair, in particular, whether lymphocytes participate in soft and hard callus formation. In order to answer this question, we analyzed femoral fracture healing in mice by confocal microscopy. Surprisingly, after the initial inflammatory phase, when soft callus developed, T and B cells withdrew from the fracture site and were detectable predominantly at the femoral neck and knee. Thereafter lymphocytes massively infiltrated the callus region (around day 14 after injury), during callus mineralization. Interestingly, lymphocytes were not found within cartilaginous areas of the callus but only nearby the newly forming bone. During healing B cell numbers seemed to exceed those of T cells and B cells progressively underwent effector maturation. Both, osteoblasts and osteoclasts were found to have direct cell-cell contact with lymphocytes, strongly suggesting a regulatory role of the immune cells specifically in the later stages of fracture healing.
Collapse
Affiliation(s)
- Ireen Könnecke
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin - Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Alessandro Serra
- German Arthritis Research Center (DRFZ), Charitéplatz 1, 10117 Berlin, Germany.
| | - Thaqif El Khassawna
- Laboratory of Experimental Trauma Surgery, Justus-Liebig University, Kerkraderstr. 9, 35394 Giessen, Germany.
| | - Claudia Schlundt
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin - Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Hanna Schell
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin - Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Anja Hauser
- German Arthritis Research Center (DRFZ), Charitéplatz 1, 10117 Berlin, Germany.
| | - Agnes Ellinghaus
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Hans-Dieter Volk
- Berlin - Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Andreas Radbruch
- Berlin - Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; German Arthritis Research Center (DRFZ), Charitéplatz 1, 10117 Berlin, Germany.
| | - Georg N Duda
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin - Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Katharina Schmidt-Bleek
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin - Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
136
|
Abstract
OBJECTIVE To demonstrate the pro-osteogenic effect of burn injury on heterotopic bone formation using a novel burn ossicle in vivo model. BACKGROUND Heterotopic ossification (HO), or the abnormal formation of bone in soft tissue, is a troubling sequela of burn and trauma injuries. The exact mechanism by which burn injury influences bone formation is unknown. The aim of this study was to develop a mouse model to study the effect of burn injury on heterotopic bone formation. We hypothesized that burn injury would enhance early vascularization and subsequent bone formation of subcutaneously implanted mesenchymal stem cells. METHODS Mouse adipose-derived stem cells were harvested from C57/BL6 mice, transfected with a BMP-2 adenovirus, seeded on collagen scaffolds (ossicles), and implanted subcutaneously in the flank region of 8 adult mice. Burn and sham groups were created with exposure of 30% surface area on the dorsum to 60°C water or 30°C water for 18 seconds, respectively (n = 4/group). Heterotopic bone volume was analyzed in vivo by micro-computed tomography for 3 months. Histological analysis of vasculogenesis was performed with platelet endothelial cell adhesion molecule staining. Osteogenic histological analysis was performed by Safranin O, Picrosirius red, and aniline blue staining. Qualitative analysis of heterotopic bone composition was completed with ex vivo Raman spectroscopy. RESULTS Subcutaneously implanted ossicles formed heterotopic bone. Ossicles from mice with burn injuries developed significantly more bone than sham control mice, analyzed by micro-computed tomography at 1, 2, and 3 months (P < 0.05), and had enhanced early and late endochondral ossification as demonstrated by Safranin O, Picrosirius red, and aniline blue staining. In addition, burn injury enhanced vascularization of the ossicles (P < 0.05). All ossicles demonstrated chemical composition characteristic of bone as demonstrated by Raman spectroscopy. CONCLUSIONS Burn injury increases the predilection to osteogenic differentiation of ectopically implanted ossicles. Early differences in vascularity correlated with later bone development. Understanding the role of burn injury on heterotopic bone formation is an important first step toward the development of treatment strategies aimed to prevent unwanted and detrimental heterotopic bone formation.
Collapse
|
137
|
O′Neill DG, Church DB, McGreevy PD, Thomson PC, Brodbelt DC. Prevalence of disorders recorded in dogs attending primary-care veterinary practices in England. PLoS One 2014; 9:e90501. [PMID: 24594665 PMCID: PMC3942437 DOI: 10.1371/journal.pone.0090501] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/03/2014] [Indexed: 01/17/2023] Open
Abstract
Purebred dog health is thought to be compromised by an increasing occurence of inherited diseases but inadequate prevalence data on common disorders have hampered efforts to prioritise health reforms. Analysis of primary veterinary practice clinical data has been proposed for reliable estimation of disorder prevalence in dogs. Electronic patient record (EPR) data were collected on 148,741 dogs attending 93 clinics across central and south-eastern England. Analysis in detail of a random sample of EPRs relating to 3,884 dogs from 89 clinics identified the most frequently recorded disorders as otitis externa (prevalence 10.2%, 95% CI: 9.1-11.3), periodontal disease (9.3%, 95% CI: 8.3-10.3) and anal sac impaction (7.1%, 95% CI: 6.1-8.1). Using syndromic classification, the most prevalent body location affected was the head-and-neck (32.8%, 95% CI: 30.7-34.9), the most prevalent organ system affected was the integument (36.3%, 95% CI: 33.9-38.6) and the most prevalent pathophysiologic process diagnosed was inflammation (32.1%, 95% CI: 29.8-34.3). Among the twenty most-frequently recorded disorders, purebred dogs had a significantly higher prevalence compared with crossbreds for three: otitis externa (P = 0.001), obesity (P = 0.006) and skin mass lesion (P = 0.033), and popular breeds differed significantly from each other in their prevalence for five: periodontal disease (P = 0.002), overgrown nails (P = 0.004), degenerative joint disease (P = 0.005), obesity (P = 0.001) and lipoma (P = 0.003). These results fill a crucial data gap in disorder prevalence information and assist with disorder prioritisation. The results suggest that, for maximal impact, breeding reforms should target commonly-diagnosed complex disorders that are amenable to genetic improvement and should place special focus on at-risk breeds. Future studies evaluating disorder severity and duration will augment the usefulness of the disorder prevalence information reported herein.
Collapse
Affiliation(s)
- Dan G. O′Neill
- Veterinary Epidemiology, Economics and Public Health, Royal Veterinary College, London, United Kingdom
| | - David B. Church
- Small Animal Medicine and Surgery Group, Royal Veterinary College, London, United Kingdom
| | - Paul D. McGreevy
- Faculty of Veterinary Science, University of Sydney, Sydney, New South Wales, Australia
| | - Peter C. Thomson
- Faculty of Veterinary Science, University of Sydney, Sydney, New South Wales, Australia
| | - Dave C. Brodbelt
- Veterinary Epidemiology, Economics and Public Health, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
138
|
Almeida CR, Serra T, Oliveira MI, Planell JA, Barbosa MA, Navarro M. Impact of 3-D printed PLA- and chitosan-based scaffolds on human monocyte/macrophage responses: unraveling the effect of 3-D structures on inflammation. Acta Biomater 2014; 10:613-22. [PMID: 24211731 DOI: 10.1016/j.actbio.2013.10.035] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/24/2013] [Accepted: 10/29/2013] [Indexed: 01/08/2023]
Abstract
Recent studies have pointed towards a decisive role of inflammation in triggering tissue repair and regeneration, while at the same time it is accepted that an exacerbated inflammatory response may lead to rejection of an implant. Within this context, understanding and having the capacity to regulate the inflammatory response elicited by 3-D scaffolds aimed for tissue regeneration is crucial. This work reports on the analysis of the cytokine profile of human monocytes/macrophages in contact with biodegradable 3-D scaffolds with different surface properties, architecture and controlled pore geometry, fabricated by 3-D printing technology. Fabrication processes were optimized to create four different 3-D platforms based on polylactic acid (PLA), PLA/calcium phosphate glass or chitosan. Cytokine secretion and cell morphology of human peripheral blood monocytes allowed to differentiate on the different matrices were analyzed. While all scaffolds supported monocyte/macrophage adhesion and stimulated cytokine production, striking differences between PLA-based and chitosan scaffolds were found, with chitosan eliciting increased secretion of tumor necrosis factor (TNF)-α, while PLA-based scaffolds induced higher production of interleukin (IL)-6, IL-12/23 and IL-10. Even though the material itself induced the biggest differences, the scaffold geometry also impacted on TNF-α and IL-12/23 production, with chitosan scaffolds having larger pores and wider angles leading to a higher secretion of these pro-inflammatory cytokines. These findings strengthen the appropriateness of these 3-D platforms to study modulation of macrophage responses by specific parameters (chemistry, topography, scaffold architecture).
Collapse
|
139
|
Uskoković V, Desai TA. In vitro analysis of nanoparticulate hydroxyapatite/chitosan composites as potential drug delivery platforms for the sustained release of antibiotics in the treatment of osteomyelitis. J Pharm Sci 2013; 103:567-79. [PMID: 24382825 DOI: 10.1002/jps.23824] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/06/2013] [Accepted: 12/03/2013] [Indexed: 12/17/2022]
Abstract
Nanoparticulate composites of hydroxyapatite (HAp) and chitosan were synthesized by ultrasound-assisted sequential precipitation and characterized for their microstructure at the atomic scale, surface charge, drug release properties, and combined antibacterial and osteogenic response. Crystallinity of HAp nanoparticles was reduced because of the interference of the surface layers of chitosan with the dissolution/reprecipitation-mediated recrystallization mechanism that conditions the transition from the as-precipitated amorphous calcium phosphate phase to the most thermodynamically stable one--HAp. Embedment of 5-10 nm sized, narrowly dispersed HAp nanoparticles within the polymeric matrix mitigated the burst release of the small molecule model drug, fluorescein, bound to HAp by physisorption, and promoted sustained-release kinetics throughout the 3 weeks of release time. The addition of chitosan to the particulate drug carrier formulation, however, reduced the antibacterial efficacy against S aureus. Excellent cell spreading and proliferation of osteoblastic MC3T3-E1 cells evidenced on microscopic conglomerates of HAp nanoparticles in vitro also markedly diminished on HAp/chitosan composites. Mitochondrial dehydrogenase activity exhibited normal values only for HAp/chitosan particle concentrations of up to 2 mg/cm(2) and significantly dropped, by about 50%, at higher particle concentrations (4 and 8 mg/cm(2)). The gene expression of osteocalcin, a mineralization inductor, and the transcription factor Runx2 was downregulated in cells incubated in the presence of 3 mg/cm(2) HAp/chitosan composite particles, whereas the expression of osteopontin, a potent mineralization inhibitor, was upregulated, further demonstrating the partially unfavorable osteoblastic cell response to the given particles. The peak in the expression of osteogenic markers paralleling the osteoblastic differentiation was also delayed most for the cell population incubated with HAp/chitosan particles. Overall, the positive effect of chitosan coating on the drug elution profile of HAp nanoparticles as carriers for the controlled delivery of antibiotics in the treatment of osteomyelitis was compensated for by the lower bacteriostatic efficiency and the comparatively unviable cell response to the composite material, especially at higher dosages.
Collapse
Affiliation(s)
- Vuk Uskoković
- Therapeutic Micro and Nanotechnology Laboratory, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, 94158-2330
| | | |
Collapse
|
140
|
Ruggiu A, Ulivi V, Sanguineti F, Cancedda R, Descalzi F. The effect of Platelet Lysate on osteoblast proliferation associated with a transient increase of the inflammatory response in bone regeneration. Biomaterials 2013; 34:9318-30. [DOI: 10.1016/j.biomaterials.2013.08.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/07/2013] [Indexed: 12/19/2022]
|
141
|
Kim YH, Furuya H, Tabata Y. Enhancement of bone regeneration by dual release of a macrophage recruitment agent and platelet-rich plasma from gelatin hydrogels. Biomaterials 2013; 35:214-24. [PMID: 24125774 DOI: 10.1016/j.biomaterials.2013.09.103] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 09/26/2013] [Indexed: 12/25/2022]
Abstract
Macrophages play an important role in regulating inflammatory responses and tissue regeneration. In the present study, their effect on bone remodeling is investigated by the simultaneous application of a macrophage recruiting agent, SEW2871 of a sphingosine-1 phosphate agonist, and platelet-rich plasma (PRP). The non-water soluble SEW2871 was solubilized in water through micelles formation with l-lactic acid grafted gelatin, and the resulting micelles with PRP were incorporated into gelatin hydrogels. Mixed SEW2871-micelles and PRP were released from gelatin hydrogels in a controlled fashion both in vitro and in vivo. In vitro migration assay revealed that the presence of PRP synergistically promoted SEW2871-induced macrophages migration. When applied to a bone defect of rats, the hydrogels incorporating mixed SEW2871-micelles and PRP recruited a higher number of macrophages than those hydrogels incorporating either SEW2871-micelles or PRP. The hydrogels incorporating mixed SEW2871-micelles and PRP enhanced the level of tumor necrosis factor (TNF)-α of pro-inflammatory cytokine, 3 days after application, while pro-inflammatory responses coupled with a significant increase in the expression level of osteoprotegerin (OPG) and interleukin (IL)-10 and transforming growth factor (TGF)-β1 of anti-inflammatory cytokine were observed 10 days postoperatively. The hydrogels incorporating mixed SEW2871-micelles and PRP promoted bone regeneration to a significant great extent compared with those incorporating PBS and either SEW2871-micelles or PRP. It is concluded that macrophages recruitment contributed to PRP-induced bone regeneration.
Collapse
Affiliation(s)
- Yang-Hee Kim
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
142
|
Philippart P, Meuleman N, Stamatopoulos B, Najar M, Pieters K, De Bruyn C, Bron D, Lagneaux L. In vivo production of mesenchymal stromal cells after injection of autologous platelet-rich plasma activated by recombinant human soluble tissue factor in the bone marrow of healthy volunteers. Tissue Eng Part A 2013; 20:160-70. [PMID: 23924315 DOI: 10.1089/ten.tea.2013.0244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Autologous mesenchymal stromal cell (MSC)-based therapies offer one of the most promising and safe methods for regeneration or reconstruction of tissues and organs. Routine procedures to obtain adequate amount of autologous stem cells need their expansion through culture, with risks of contamination and cell differentiation, leading to the loss of cell ability for therapies. We suggest the use of human bone marrow (BM) as a physiological bioreactor to produce autologous MSC by injection of autologous platelet-rich plasma activated by recombinant human soluble tissue factor (rhsTF) in iliac crest. A trial on 13 healthy volunteers showed the feasibility and harmlessness of the procedure. The phenotype and cellularity of BM cells were not modified, on day 3 after injection. Endothelial progenitor cells (EPC) were mobilized to the bloodstream, without stimulation of hematopoietic stem cells (HSC). MSC level in BM increased with a specific commitment to preosteoblastic cell population both in vivo and in vitro. This self-stimulation system of BM seems thus to be a promising feasible process 3 days before clinical cell therapy applications.
Collapse
Affiliation(s)
- Pierre Philippart
- 1 Department of Stomatology and Maxillo-Facial Surgery, HIS Site Bracops , Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
143
|
Overman JR, Helder MN, ten Bruggenkate CM, Schulten EAJM, Klein-Nulend J, Bakker AD. Growth factor gene expression profiles of bone morphogenetic protein-2-treated human adipose stem cells seeded on calcium phosphate scaffolds in vitro. Biochimie 2013; 95:2304-13. [PMID: 24028822 DOI: 10.1016/j.biochi.2013.08.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/27/2013] [Indexed: 12/17/2022]
Abstract
The secretome of stem cells strongly determines the outcome of tissue engineering strategies. We investigated how the secretome of human adipose stem cells (hASCs) can be affected by substrate, BMP-2 treatment, and degree of differentiation. We hypothesized that as differentiation progresses, hASCs produce increasingly more gene products associated with processes such as angiogenesis and bone remodeling. Human ASCs were treated for 15 min with BMP-2 (10 ng/ml) to enhance osteogenic differentiation, or with vehicle. Subsequently, hASCs were seeded on plastic or on biphasic calcium phosphate (BCP) consisting of 60% hydroxyapatite and 40% β-tricalcium phosphate. A PCR array for ~150 trophic factors and differentiation-related genes was performed at day 21 of culture. A limited set of factors was quantified by qPCR at days 0, 4, 14 and 21, and/or ELISA at day 21. Compared to plastic, BCP-cultured hASCs showed ≥2-fold higher expression of ~20 factors, e.g. cytokines such as IL-6, growth factors such as FGF7 and adhesion molecules such as VCAM1. Expression of another ~50 genes was decreased ≥2-fold on BCP vs. plastic, even though hASCs differentiate better on BCP than on plastic. BMP-2-treatment increased the expression of ~30 factors by hASCs seeded on BCP, while it decreased the expression of only PGF, PPARG and PTN. Substrate affected hASC secretion of Activin A and seemed to affect P1NP release. No clear association between hASC osteogenic differentiation and growth factor expression pattern was observed. Considering our observed lack of association between the degree of differentiation and the expression of factors associated with angiogenesis and bone remodeling by hASCs, future bone regeneration studies should focus more on systematically orchestrating the secretome of stem cells, rather than on inducing osteogenic differentiation of stem cells only. Short incubation with BMP-2 may be a promising treatment to enhance both osteogenic differentiation and environmental modulation.
Collapse
Affiliation(s)
- J R Overman
- Dept. Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands; Dept. Oral and Maxillofacial Surgery, Academic Centre for Dentistry Amsterdam/VU University Medical Center, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
144
|
Santo VE, Gomes ME, Mano JF, Reis RL. Controlled release strategies for bone, cartilage, and osteochondral engineering--Part I: recapitulation of native tissue healing and variables for the design of delivery systems. TISSUE ENGINEERING. PART B, REVIEWS 2013; 19:308-26. [PMID: 23268651 PMCID: PMC3690094 DOI: 10.1089/ten.teb.2012.0138] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 12/11/2012] [Indexed: 12/12/2022]
Abstract
The potential of growth factors to stimulate tissue healing through the enhancement of cell proliferation, migration, and differentiation is undeniable. However, critical parameters on the design of adequate carriers, such as uncontrolled spatiotemporal presence of bioactive factors, inadequate release profiles, and supraphysiological dosages of growth factors, have impaired the translation of these systems onto clinical practice. This review describes the healing cascades for bone, cartilage, and osteochondral interface, highlighting the role of specific growth factors for triggering the reactions leading to tissue regeneration. Critical criteria on the design of carriers for controlled release of bioactive factors are also reported, focusing on the need to provide a spatiotemporal control over the delivery and presentation of these molecules.
Collapse
Affiliation(s)
- Vítor E. Santo
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuela E. Gomes
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João F. Mano
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
145
|
Dishowitz MI, Mutyaba PL, Takacs JD, Barr AM, Engiles JB, Ahn J, Hankenson KD. Systemic inhibition of canonical Notch signaling results in sustained callus inflammation and alters multiple phases of fracture healing. PLoS One 2013; 8:e68726. [PMID: 23844237 PMCID: PMC3701065 DOI: 10.1371/journal.pone.0068726] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 06/03/2013] [Indexed: 11/18/2022] Open
Abstract
The Notch signaling pathway is an important regulator of embryological bone development, and many aspects of development are recapitulated during bone repair. We have previously reported that Notch signaling components are upregulated during bone fracture healing. However, the significance of the Notch pathway in bone regeneration has not been described. Therefore, the objective of this study was to determine the importance of Notch signaling in regulating bone fracture healing by using a temporally controlled inducible transgenic mouse model (Mx1-Cre;dnMAMLf/-) to impair RBPjκ-mediated canonical Notch signaling. The Mx1 promoter was synthetically activated resulting in temporally regulated systemic dnMAML expression just prior to creation of bilateral tibial fractures. This allowed for mice to undergo unaltered embryological and post-natal skeletal development. Results showed that systemic Notch inhibition prolonged expression of inflammatory cytokines and neutrophil cell inflammation, and reduced the proportion of cartilage formation within the callus at 10 days-post-fracture (dpf) Notch inhibition did not affect early bone formation at 10dpf, but significantly altered bone maturation and remodeling at 20dpf. Increased bone volume fraction in dnMAML fractures, which was due to a moderate decrease in callus size with no change in bone mass, coincided with increased trabecular thickness but decreased connectivity density, indicating that patterning of bone was altered. Notch inhibition decreased total osteogenic cell density, which was comprised of more osteocytes rather than osteoblasts. dnMAML also decreased osteoclast density, suggesting that osteoclast activity may also be important for altered fracture healing. It is likely that systemic Notch inhibition had both direct effects within cell types as well as indirect effects initiated by temporally upstream events in the fracture healing cascade. Surprisingly, Notch inhibition did not alter cell proliferation. In conclusion, our results demonstrate that the Notch signaling pathway is required for the proper temporal progression of events required for successful bone fracture healing.
Collapse
Affiliation(s)
- Michael I. Dishowitz
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Patricia L. Mutyaba
- Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Joel D. Takacs
- Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrew M. Barr
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Julie B. Engiles
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kurt D. Hankenson
- Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
146
|
Santos SG, Lamghari M, Almeida CR, Oliveira MI, Neves N, Ribeiro AC, Barbosa JN, Barros R, Maciel J, Martins MCL, Gonçalves RM, Barbosa MA. Adsorbed fibrinogen leads to improved bone regeneration and correlates with differences in the systemic immune response. Acta Biomater 2013; 9:7209-17. [PMID: 23571000 DOI: 10.1016/j.actbio.2013.04.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/21/2013] [Accepted: 04/02/2013] [Indexed: 12/25/2022]
Abstract
Designing new biomaterials that can modulate the inflammatory response instead of attempting just to reduce it constitutes a paradigm change in regenerative medicine. This work aimed to investigate the capacity of an immunomodulatory biomaterial to enhance bone regeneration. For that purpose we incorporated a molecule with well-established pro-inflammatory and pro-healing roles, fibrinogen, in chitosan scaffolds. Two different incorporation strategies were tested, leading to concentrations of 0.54±0.10mg fibrinogen g(-1) scaffold immediately upon adsorption (Fg-Sol), and 0.34±0.04mg fibrinogen g(-1) scaffold after washing (Fg-Ads). These materials were implanted in a critical size bone defect in rats. At two months post-implantation the extent of bone regeneration was examined by histology and the systemic immune response triggered was evaluated by determining the percentages of myeloid cells, T and B lymphocytes in the draining lymph nodes. The results obtained indicate that the fibrinogen incorporation strategy conditioned the osteogenic capacity of biomaterials. Fg-Ads scaffolds led to more bone formation, and the presence of Fg stimulated angiogenesis. Furthermore, animals implanted with Fg-Ads scaffolds showed significant increases in the percentages of B lymphocytes and myeloid cells in the draining lymph nodes, while levels of T lymphocytes were not significantly different. Finally, a significant increase in TGF-β1 was detected in the plasma of animals implanted with Fg-Ads. Taken together the results presented suggest a potential correlation between the elicited immune response and biomaterial osteogenic performance.
Collapse
Affiliation(s)
- S G Santos
- Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Reside J, Everett E, Padilla R, Arce R, Miguez P, Brodala N, De Kok I, Nares S. In vivo assessment of bone healing following Piezotome® ultrasonic instrumentation. Clin Implant Dent Relat Res 2013; 17:384-94. [PMID: 23763591 DOI: 10.1111/cid.12094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE This pilot study evaluated the molecular, histologic, and radiographic healing of bone to instrumentation with piezoelectric or high speed rotary (R) devices over a 3-week healing period. MATERIAL AND METHODS Fourteen Sprague-Dawley rats (Charles River Laboratories International, Inc., Wilmington, MA, USA) underwent bilateral tibial osteotomies prepared in a randomized split-leg design using Piezotome® (P1) (Satelec Acteon, Merignac, France), Piezotome 2® (P2) (Satelec Acteon), High-speed R instrumentation, or sham surgery (S). At 1 week, an osteogenesis array was used to evaluate differences in gene expression while quantitative analysis assessed percentage bone fill (PBF) and bone mineral density (BMD) in the defect, peripheral, and distant regions at 3 weeks. Qualitative histologic evaluation of healing osteotomies was also performed at 3 weeks. RESULTS At 1 week, expression of 11 and 18 genes involved in bone healing was significantly (p < .05) lower following P1 and P2 instrumentation, respectively, relative to S whereas 16 and 4 genes were lower relative to R. No differences in PBF or BMD were detected between groups within the osteotomy defect. However, significant differences in PBF (p = .020) and BMD (p = .008) were noted along the peripheral region between P2 and R groups, being R the group with the lowest values. Histologically, smooth osteotomy margins were present following instrumentation using P1 or P2 relative to R. CONCLUSIONS Piezoelectric instrumentation favors preservation of bone adjacent to osteotomies while variations in gene expression suggest differences in healing rates due to surgical modality. Bone instrumented by piezoelectric surgery appears less detrimental to bone healing than high-speed R device.
Collapse
Affiliation(s)
- Jonathan Reside
- Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Mountziaris PM, Dennis Lehman E, Mountziaris I, Sing DC, Kasper FK, Mikos AG. Effect of temporally patterned TNF-α delivery on in vitro osteogenic differentiation of mesenchymal stem cells cultured on biodegradable polymer scaffolds. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2013; 24:1794-813. [PMID: 23746285 DOI: 10.1080/09205063.2013.803455] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent insight into the critical role of pro-inflammatory cytokines, particularly tumor necrosis factor-α (TNF-α), in bone regeneration has heralded a new direction in the design of tissue engineering constructs. Previous studies have demonstrated that continuous delivery of 50 ng/ml TNF-α to mesenchymal stem cells (MSCs) cultured on three-dimensional (3D) biodegradable electrospun poly(ϵ-caprolactone) (PCL) microfiber meshes stimulates mineralized matrix deposition, a marker of osteogenic differentiation. Since TNF-α exhibits a biphasic pattern of expression following bone fracture in vivo, this study aimed to investigate the effects of temporal patterns of TNF-α delivery on in vitro osteogenic differentiation of MSCs cultured on 3D electrospun PCL scaffolds. MSCs were cultured for 16 days and exposed to continuous, early, intermediate, or late TNF-α delivery. To further elucidate the effects of TNF-α on osteogenic differentiation, the study design included MSCs precultured both in the presence and absence of typically required osteogenic supplement dexamethasone. Mineralized matrix deposition was not observed in constructs with dexamethasone-naïve MSCs, suggesting that TNF-α is not sufficient to trigger in vitro osteogenic differentiation of MSCs. For MSCs precultured with dexamethasone, TNF-α suppressed alkaline phosphatase activity, an early marker of osteogenic differentiation, and stimulated mineralized matrix deposition, a late stage marker of MSC osteogenic differentiation. By elucidating the impact of temporal variations in TNF-α delivery on MSC osteogenic differentiation, our results offer insight into the regenerative mechanism of TNF-α and provide the design parameters for a novel tissue engineering strategy that rationally controls TNF-α signaling to stimulate bone regeneration.
Collapse
Affiliation(s)
- Paschalia M Mountziaris
- a Department of Bioengineering , Rice University , P.O. Box 1892, MS 142 , Houston , TX , 77251-1892 , USA
| | | | | | | | | | | |
Collapse
|
149
|
Grassi F, Cattini L, Gambari L, Manferdini C, Piacentini A, Gabusi E, Facchini A, Lisignoli G. T cell subsets differently regulate osteogenic differentiation of human mesenchymal stromal cells in vitro. J Tissue Eng Regen Med 2013; 10:305-14. [PMID: 23653421 DOI: 10.1002/term.1727] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 10/18/2012] [Accepted: 01/24/2013] [Indexed: 01/03/2023]
Abstract
T lymphocytes play a key role in the regulation of bone homeostasis and bone healing. The inflammatory response at the site of bone injury is essential to the initiation of the bone repair program; however, an uncontrolled exposure to inflammatory environment has a negative effect on tissue regeneration - indeed, activated T cells were shown to inhibit osteogenic differentiation on human mesenchymal stromal cells (MSCs). Whether resting T cells can induce osteogenic differentiation of MSCs and what role specific T cells subset play in this process is still elusive. In this study, we sought to analyse the osteogenic gene expression profile of whole T cells, CD4 and CD8 T cells isolated from healthy donors and investigated whether secreted factors from each group modulate osteogenic differentiation of human MSCs. Gene expression profiling identified a pool of 51 genes involved at various stages in bone growth which are expressed above detectable levels in CD4 and CD8 T cells. Most genes of this pool were expressed at higher levels in the CD4 subset. In vitro mineralization assays revealed that conditioned medium from CD4 T cells, but not from CD8 cells, significantly increased mineralization in osteogenic cultures of human MSCs; furthermore, mRNA expression of Runt-related transcription factor 2 (RUNX-2), osteocalcin (OC), bone sialoprotein (BSP) and alkaline phosphatase (ALP) in MSCs was significantly upregulated in the presence of CD4-conditioned medium but not with that obtained from CD8. The results show a differential role for CD4 and CD8 T cells in supporting bone formation and identify an osteogenic gene signature of each subset.
Collapse
Affiliation(s)
| | - Luca Cattini
- S. C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Laura Gambari
- S. C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Cristina Manferdini
- S. C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Anna Piacentini
- S. C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elena Gabusi
- Laboratorio RAMSES, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Andrea Facchini
- S. C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gina Lisignoli
- S. C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
150
|
Amini AR, Laurencin CT, Nukavarapu SP. Bone tissue engineering: recent advances and challenges. Crit Rev Biomed Eng 2013; 40:363-408. [PMID: 23339648 DOI: 10.1615/critrevbiomedeng.v40.i5.10] [Citation(s) in RCA: 1409] [Impact Index Per Article: 117.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The worldwide incidence of bone disorders and conditions has trended steeply upward and is expected to double by 2020, especially in populations where aging is coupled with increased obesity and poor physical activity. Engineered bone tissue has been viewed as a potential alternative to the conventional use of bone grafts, due to their limitless supply and no disease transmission. However, bone tissue engineering practices have not proceeded to clinical practice due to several limitations or challenges. Bone tissue engineering aims to induce new functional bone regeneration via the synergistic combination of biomaterials, cells, and factor therapy. In this review, we discuss the fundamentals of bone tissue engineering, highlighting the current state of this field. Further, we review the recent advances of biomaterial and cell-based research, as well as approaches used to enhance bone regeneration. Specifically, we discuss widely investigated biomaterial scaffolds, micro- and nano-structural properties of these scaffolds, and the incorporation of biomimetic properties and/or growth factors. In addition, we examine various cellular approaches, including the use of mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), adult stem cells, induced pluripotent stem cells (iPSCs), and platelet-rich plasma (PRP), and their clinical application strengths and limitations. We conclude by overviewing the challenges that face the bone tissue engineering field, such as the lack of sufficient vascularization at the defect site, and the research aimed at functional bone tissue engineering. These challenges will drive future research in the field.
Collapse
Affiliation(s)
- Ami R Amini
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|