1
|
Chen Z, Jia M, Liu Y, Zhou H, Wang X, Wu M. Injectable Composite Hydrogel Stents for Bone Defect Management with Enhanced Osteogenesis and Angiogenesis. Int J Nanomedicine 2025; 20:4589-4606. [PMID: 40242608 PMCID: PMC12002338 DOI: 10.2147/ijn.s509686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Background The use of autologous bone grafting is considered the most successful method for managing bone defects, particularly when utilizing cancellous bone grafts for the best outcomes. Nonetheless, the scarcity of cancellous bone presents a notable obstacle in remedying these defects. Consequently, it is essential to create reliable alternatives to cancellous bone grafts to ensure effective management of bone defects. Methods In this research, we created an injectable composite hydrogel stents using gelatin methacrylate (GelMA) hydrogel to mimic the collagen properties of cancellous bone, along with the inclusion of nanohydroxyapatite (nHA) to signify the inorganic element. Furthermore, we incorporated vascular endothelial growth factor (VEGF) to improve regenerative vascular capabilities. Before being implanted into rat cranium defect models, these composite hydrogel stents were co-cultured with human umbilical vein endothelial cells (HUVEC) and bone marrow mesenchymal stem cells (BMSC). Results The composite hydrogel stents exhibited a network structure with porosity, robust mechanical properties, and beneficial degradation traits. In the degradation phase, it steadily releases Ca²⁺ and VEGF, which encourages the proliferation, migration, and osteogenic differentiation of BMSCs from rats. Moreover, this release improves the ability of HUVECs to form tubes. Collectively, these mechanisms support the regeneration of blood vessels and bone in the cranium defect region of rats. Conclusion The composite hydrogel stents demonstrated excellent cytocompatibility and biological characteristics, as evidenced by its ability to enhance both osteogenesis and angiogenesis in vivo and in vitro. Consequently, it has the potential to act as an effective alternative to natural cancellous bone.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, Bengbu, People’s Republic of China
- Department of Orthopedics, Fenjinting Hospital in Sihong, Suqian, People’s Republic of China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, People’s Republic of China
| | - Mingyu Jia
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, People’s Republic of China
| | - Yangyang Liu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, Bengbu, People’s Republic of China
| | - Huajian Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, People’s Republic of China
| | - Xiaopan Wang
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, Bengbu, People’s Republic of China
| | - Min Wu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, People’s Republic of China
| |
Collapse
|
2
|
Vanderlei BMC, Torres MC, Paredes N, Garcez AS, Pavini PTM, Suzuki SS, Moon W. Effect of photobiomodulation and corticopuncture methods on tooth displacement and gene expression: animal study. Lasers Med Sci 2024; 39:283. [PMID: 39547960 DOI: 10.1007/s10103-024-04136-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/08/2024] [Indexed: 11/17/2024]
Abstract
AIM The aim of this study was to evaluate the expression levels of vascular endothelial growth factor (VEGF), Peroxiredoxin 1 (PRX1), glucose transporter 1 (GLUT1) and type I collagen (COL1) and the rate of tooth movement comparing 3 accelerated tooth movement (ATM) methods: Corticopuncture (CP), photobiomodulation (PBM) and the combined technique (CP + PBM) on days 1, 3, 7 and 14. METHODS Orthodontic tooth movement was induced in 24 male Wistar rats. CP procedure included three perforations: two in the palate and one mesial to the molars. GaAlAs diode laser irradiation was performed on days 0, 2, 4 and 6, totaling 4 irradiations. 14 days (810 nm, 100 mW, 15 s). Gingival tissue was collected from the cervical area of both first molars and qPCR was performed to isolate and quantify mRNA levels. RESULTS All ATM groups showed increased tooth displacement compared to control after 14 days (20% for PBM; 40% for CP and 60% for CP + PBM). PBM showed higher VEGF expression on days 1,3 and 7 followed by CP and CP + PBM. PRX1 levels increased on days 1 and 3 in PBM and CP + PBM. GLUT1 increased on day 3 in all groups. No difference was found on levels of VEGF, PRX1 and GLUT1 among the groups on day 14, except for COL1 which increased significantly in PBM group. CONCLUSION All ATM methods showed higher expression of all of VEGF, PRX1, GLUT1, COL1 than control group. PBM and CP + PBM groups had more expression related to angiogenesis, glucose uptake, oxidative stress and collagen synthesis.
Collapse
Affiliation(s)
| | | | - Ney Paredes
- School of Dentistry, University of California, Los Angeles, CA, USA
| | | | | | - Selly Sayuri Suzuki
- Department of Orthodontics and Post-graduation in Orthodontics, Faculdade São Leopoldo Mandic, Campinas, SP, 52020-060, Brazil
| | - Won Moon
- Department of Orthodontics, at Institute of Oral Health Science, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
3
|
Fernandes CJC, Silva RA, Ferreira MR, Fuhler GM, Peppelenbosch MP, van der Eerden BC, Zambuzzi WF. Vascular smooth muscle cell-derived exosomes promote osteoblast-to-osteocyte transition via β-catenin signaling. Exp Cell Res 2024; 442:114211. [PMID: 39147261 DOI: 10.1016/j.yexcr.2024.114211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/17/2024]
Abstract
Blood vessel growth and osteogenesis in the skeletal system are coupled; however, fundamental aspects of vascular function in osteoblast-to-osteocyte transition remain unclear. Our study demonstrates that vascular smooth muscle cells (VSMCs), but not endothelial cells, are sufficient to drive bone marrow mesenchymal stromal cell-derived osteoblast-to-osteocyte transition via β-catenin signaling and exosome-mediated communication. We found that VSMC-derived exosomes are loaded with transcripts encoding proteins associated with the osteocyte phenotype and members of the WNT/β-catenin signaling pathway. In contrast, endothelial cell-derived exosomes facilitated mature osteoblast differentiation by reprogramming the TGFB1 gene family and osteogenic transcription factors osterix (SP7) and RUNX2. Notably, VSMCs express significant levels of tetraspanins (CD9, CD63, and CD81) and drive the intracellular trafficking of exosomes with a lower membrane zeta potential than those from other cells. Additionally, the high ATP content within these exosomes supports mineralization mechanisms, as ATP is a substrate for alkaline phosphatase. Osteocyte function was further validated by RNA sequencing, revealing activity in genes related to intermittent mineralization and sonic hedgehog signaling, alongside a significant increase in TNFSF11 levels. Our findings unveil a novel role of VSMCs in promoting osteoblast-to-osteocyte transition, thus offering new insights into bone biology and homeostasis, as well as in bone-related diseases. Clinically, these insights could pave the way for innovative therapeutic strategies targeting VSMC-derived exosome pathways to treat bone-related disorders such as osteoporosis. By manipulating these signaling pathways, it may be possible to enhance bone regeneration and improve skeletal health in patients with compromised bone structure and function.
Collapse
Affiliation(s)
- Célio J C Fernandes
- Bioassays and Cell Dynamics Lab, Dept. of Chemistry and Biochemistry, Bioscience Institute, UNESP, Botucatu, 18603-100, Sao Paulo, Brazil
| | - Rodrigo A Silva
- School of Dentistry, University of Taubaté, 12020-340, Taubaté, São Paulo, Brazil
| | - Marcel R Ferreira
- Bioassays and Cell Dynamics Lab, Dept. of Chemistry and Biochemistry, Bioscience Institute, UNESP, Botucatu, 18603-100, Sao Paulo, Brazil
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC, Erasmus University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC, Erasmus University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Bram Cj van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Willian F Zambuzzi
- Bioassays and Cell Dynamics Lab, Dept. of Chemistry and Biochemistry, Bioscience Institute, UNESP, Botucatu, 18603-100, Sao Paulo, Brazil.
| |
Collapse
|
4
|
Shineh G, Mobaraki M, Afzali E, Alakija F, Velisdeh ZJ, Mills DK. Antimicrobial Metal and Metal Oxide Nanoparticles in Bone Tissue Repair. BIOMEDICAL MATERIALS & DEVICES 2024; 2:918-941. [DOI: 10.1007/s44174-024-00159-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/06/2024] [Indexed: 01/06/2025]
|
5
|
Damiati LA, El Soury M. Bone-nerve crosstalk: a new state for neuralizing bone tissue engineering-A mini review. Front Med (Lausanne) 2024; 11:1386683. [PMID: 38690172 PMCID: PMC11059066 DOI: 10.3389/fmed.2024.1386683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Neuro bone tissue engineering is a multidisciplinary field that combines both principles of neurobiology and bone tissue engineering to develop innovative strategies for repairing and regenerating injured bone tissues. Despite the fact that regeneration and development are considered two distinct biological processes, yet regeneration can be considered the reactivation of development in later life stages to restore missing tissues. It is noteworthy that the regeneration capabilities are distinct and vary from one organism to another (teleost fishes, hydra, humans), or even in the same organism can vary dependent on the injured tissue itself (Human central nervous system vs. peripheral nervous system). The skeletal tissue is highly innervated, peripheral nervous system plays a role in conveying the signals and connecting the central nervous system with the peripheral organs, moreover it has been shown that they play an important role in tissue regeneration. Their regeneration role is conveyed by the different cells' resident in it and in its endoneurium (fibroblasts, microphages, vasculature associated cells, and Schwann cells) these cells secrete various growth factors (NGF, BDNF, GDNF, NT-3, and bFGF) that contribute to the regenerative phenotype. The peripheral nervous system and central nervous system synchronize together in regulating bone homeostasis and regeneration through neurogenic factors and neural circuits. Receptors of important central nervous system peptides such as Serotonin, Leptin, Semaphorins, and BDNF are expressed in bone tissue playing a role in bone homeostasis, metabolism and regeneration. This review will highlight the crosstalk between peripheral nerves and bone in the developmental stages as well as in regeneration and different neuro-bone tissue engineering strategies for repairing severe bone injuries.
Collapse
Affiliation(s)
- Laila A. Damiati
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Marwa El Soury
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| |
Collapse
|
6
|
Limaye A, Perumal V, Karner CM, Arinzeh TL. Plant-Derived Zein as an Alternative to Animal-Derived Gelatin for Use as a Tissue Engineering Scaffold. ADVANCED NANOBIOMED RESEARCH 2024; 4:2300104. [PMID: 38665311 PMCID: PMC11045004 DOI: 10.1002/anbr.202300104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
Natural biomaterials are commonly used as tissue engineering scaffolds due to their biocompatibility and biodegradability. Plant-derived materials have also gained significant interest due to their abundance and as a sustainable resource. This study evaluates the corn-derived protein zein as a plant-derived substitute for animal-derived gelatin, which is widely used for its favorable cell adhesion properties. Limited studies exist evaluating pure zein for tissue engineering. Herein, fibrous zein scaffolds are evaluated in vitro for cell adhesion, growth, and infiltration into the scaffold in comparison to gelatin scaffolds and are further studied in a subcutaneous model in vivo. Human mesenchymal stem cells (MSCs) on zein scaffolds express focal adhesion kinase and integrins such as αvβ3, α4, and β1 similar to gelatin scaffolds. MSCs also infiltrate zein scaffolds with a greater penetration depth than cells on gelatin scaffolds. Cells loaded onto zein scaffolds in vivo show higher cell proliferation and CD31 expression, as an indicator of blood vessel formation. Findings also demonstrate the capability of zein scaffolds to maintain the multipotent capability of MSCs. Overall, findings demonstrate plant-derived zein may be a suitable alternative to the animalderived gelatin and demonstrates zein's potential as a scaffold for tissue engineering.
Collapse
Affiliation(s)
- Apurva Limaye
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
- Department of Biomedical Engineering, Columbia University, 3960 Broadway, New York, NY 10027, USA
| | - Venkatesan Perumal
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Courtney M Karner
- Department of Internal Medicine, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | |
Collapse
|
7
|
Menger MM, Bleimehl M, Bauer D, Scheuer C, Hans S, Saul D, Ehnert S, Menger MD, Histing T, Laschke MW. Cilostazol promotes blood vessel formation and bone regeneration in a murine non-union model. Biomed Pharmacother 2023; 168:115697. [PMID: 37864892 DOI: 10.1016/j.biopha.2023.115697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/23/2023] Open
Abstract
Non-unions represent a major complication in trauma and orthopedic surgery. Many factors contribute to bone regeneration, out of which an adequate vascularization has been recognized as crucial. The phosphodiesterase-3 (PDE-3) inhibitor cilostazol has been shown to exert pro-angiogenic and pro-osteogenic effects in a variety of preclinical studies. Hence, we herein investigated the effects of cilostazol on bone regeneration in an atrophic non-union model in mice. For this purpose, a 1.8 mm femoral segmental defect was stabilized by pin-clip fixation and the animals were treated daily with 30 mg/kg body weight cilostazol or saline (control) per os. At 2, 5 and 10 weeks after surgery the healing of femora was analyzed by X-ray, biomechanics, photoacoustic imaging, and micro-computed tomography (µCT). To investigate the cellular composition and the growth factor expression of the callus tissue additional histological, immunohistochemical and Western blot analyses were performed. Cilostazol-treated animals showed increased bone formation within the callus, resulting in an enhanced bending stiffness when compared to controls. This was associated with a more pronounced expression of vascular endothelial growth factor (VEGF), a higher number of CD31-positive microvessels and an increased oxygen saturation within the callus tissue. Furthermore, cilostazol induced higher numbers of tartrate-resistant acidic phosphate (TRAP)-positive osteoclasts and CD68-positive macrophages. Taken together, these findings demonstrate that cilostazol is a promising drug candidate for the adjuvant treatment of atrophic non-unions in clinical practice.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany; Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany.
| | - Michelle Bleimehl
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - David Bauer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Sandra Hans
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Dominik Saul
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| |
Collapse
|
8
|
Menger MM, Tobias AL, Bauer D, Bleimehl M, Scheuer C, Menger MD, Histing T, Laschke MW. Parathyroid hormone stimulates bone regeneration in an atrophic non-union model in aged mice. J Transl Med 2023; 21:844. [PMID: 37996876 PMCID: PMC10668449 DOI: 10.1186/s12967-023-04661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Non-union formation still represents a major burden in trauma and orthopedic surgery. Moreover, aged patients are at an increased risk for bone healing failure. Parathyroid hormone (PTH) has been shown to accelerate fracture healing in young adult animals. However, there is no information whether PTH also stimulates bone regeneration in atrophic non-unions in the aged. Therefore, the aim of the present study was to analyze the effect of PTH on bone regeneration in an atrophic non-union model in aged CD-1 mice. METHODS After creation of a 1.8 mm segmental defect, mice femora were stabilized by pin-clip fixation. The animals were treated daily with either 200 µg/kg body weight PTH 1-34 (n = 17) or saline (control; n = 17) subcutaneously. Bone regeneration was analyzed by means of X-ray, biomechanics, micro-computed tomography (µCT) imaging as well as histological, immunohistochemical and Western blot analyses. RESULTS In PTH-treated animals bone formation was markedly improved when compared to controls. This was associated with an increased bending stiffness as well as a higher number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts and CD31-positive microvessels within the callus tissue. Furthermore, PTH-treated aged animals showed a decreased inflammatory response, characterized by a lower number of MPO-positive granulocytes and CD68-positive macrophages within the bone defects when compared to controls. Additional Western blot analyses demonstrated a significantly higher expression of cyclooxygenase (COX)-2 and phosphoinositide 3-kinase (PI3K) in PTH-treated mice. CONCLUSION Taken together, these findings indicate that PTH is an effective pharmacological compound for the treatment of non-union formation in aged animals.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany.
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany.
| | - Anne L Tobias
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - David Bauer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Michelle Bleimehl
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| |
Collapse
|
9
|
Menger MM, Bauer D, Bleimehl M, Scheuer C, Braun BJ, Herath SC, Rollmann MF, Menger MD, Laschke MW, Histing T. Sildenafil, a phosphodiesterase-5 inhibitor, stimulates angiogenesis and bone regeneration in an atrophic non-union model in mice. J Transl Med 2023; 21:607. [PMID: 37684656 PMCID: PMC10486066 DOI: 10.1186/s12967-023-04441-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/14/2023] [Indexed: 09/10/2023] Open
Abstract
Non-union formation represents a major complication in trauma and orthopedic surgery. The phosphodiesterase-5 (PDE-5) inhibitor sildenafil has been shown to exert pro-angiogenic and pro-osteogenic effects in vitro and in vivo. Therefore, the aim of the present study was to analyze the impact of sildenafil in an atrophic non-union model in mice. After creation of a 1.8 mm segmental defect, mice femora were stabilized by pin-clip fixation. Bone regeneration was analyzed by means of X-ray, biomechanics, photoacoustic and micro-computed tomography (µCT) imaging as well as histological, immunohistochemical and Western blot analyses at 2, 5 and 10 weeks after surgery. The animals were treated daily with either 5 mg/kg body weight sildenafil (n = 35) or saline (control; n = 35) per os. Bone formation was markedly improved in defects of sildenafil-treated mice when compared to controls. This was associated with a higher bending stiffness as well as an increased number of CD31-positive microvessels and a higher oxygen saturation within the callus tissue. Moreover, the bone defects of sildenafil-treated animals contained more tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts and CD68-positive macrophages and exhibited a higher expression of the pro-angiogenic and pro-osteogenic markers cysteine rich protein (CYR)61 and vascular endothelial growth factor (VEGF) when compared to controls. These findings demonstrate that sildenafil acts as a potent stimulator of angiogenesis and bone regeneration in atrophic non-unions.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany.
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| | - David Bauer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Michelle Bleimehl
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Benedikt J Braun
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Steven C Herath
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Mika F Rollmann
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| |
Collapse
|
10
|
An G, Kim M, Park J, Park H, Hong T, Lim W, Song G. Embryonic exposure to chloroxylenol induces developmental defects and cardiovascular toxicity via oxidative stress, inflammation, and apoptosis in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2023; 268:109617. [PMID: 36965842 DOI: 10.1016/j.cbpc.2023.109617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
Chloroxylenol is an extensively consumed anti-microbial compound. Since its usage is on the rise due to the coronavirus pandemic and ban on other antimicrobial ingredients, recent studies have suggested the necessity of estimating its potential for ecotoxicity. The detrimental effect of chloroxylenol on zebrafish (Danio rerio) viability has been reported; however, research on the mechanisms underlying its toxicity is quite limited. Therefore, we applied the zebrafish model for elucidating responses against chloroxylenol to predict its toxicity toward human health and ecology. Zebrafish exposed to chloroxylenol (0, 0.5, 1, 2.5, 5, and 10 mg/L) at the embryonic stage (from 6 h post-fertilization (hpf) to 96 hpf) showed impaired viability and hatchability, and pathological phenotypes. To address these abnormalities, cellular responses such as oxidative stress, inflammation, and apoptosis were confirmed via in vivo imaging of a fluorescent dye or measurement of the transcriptional changes related to each response. In particular, developmental defects in the cardiovascular system of zebrafish exposed to 0, 0.5, 1, and 2.5 mg/L of chloroxylenol from 6 to 96 hpf were identified by structural analyses of the system in the flk1:eGFP transgenic line. Additional experiments were conducted using human umbilical vein endothelial cells (HUVECs) to predict the adverse impacts of chloroxylenol on the human vascular system. Chloroxylenol impairs the viability and tube formation ability of HUVECs by modulating ERK signaling. The findings obtained using the zebrafish model provide evidence of the possible risks of chloroxylenol exposure and suggest the importance of more in-depth ecotoxicological studies.
Collapse
Affiliation(s)
- Garam An
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Miji Kim
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Junho Park
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hahyun Park
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Taeyeon Hong
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
11
|
Duygu G, Yalcin-Ülker GM, Günbatan M, Soluk-Tekkesin M, Özcakir-Tomruk C. Evaluation of Preventive Role of Systemically Applied Erythropoietin after Tooth Extraction in a Bisphosphonate-Induced MRONJ Model. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1059. [PMID: 37374263 DOI: 10.3390/medicina59061059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: In this experimental study, the prophylactic effect of systemically administered erythropoietin (EPO) in medication-related osteonecrosis of the jaw (MRONJ) was evaluated. Materials and Methods: The osteonecrosis model was established using 36 Sprague Dawley rats. EPO was systemically applied before and/or after tooth extraction. Groups were formed based on the application time. All samples were evaluated histologically, histomorphometrically, and immunohistochemically. A statistically significant difference in new bone formation was observed between the groups (p < 0.001). Results: When new bone-formation rates were compared, no significant differences were observed between the control group and the EPO, ZA+PostEPO, and ZA+Pre-PostEPO groups (p = 1, 0.402, and 1, respectively); however, this rate was significantly lower in the ZA+PreEPO group (p = 0.021). No significant differences in new bone formation were observed between the ZA+PostEPO and ZA+PreEPO groups (p = 1); however, this rate was significantly higher in the ZA+Pre-PostEPO group (p = 0.009). The ZA+Pre-PostEPO group demonstrated significantly higher intensity level in VEGF protein expression than the other groups (p < 0.001). Conclusions: Administering EPO two weeks pre-extraction and continuing EPO treatment for three weeks post-extraction in ZA-treated rats optimized the inflammatory reaction, increased angiogenesis by inducing VEGF, and positively affected bone healing. Further studies are needed to determine the exact durations and doses.
Collapse
Affiliation(s)
- Gonca Duygu
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Tekirdag Namık Kemal University, Tekirdag 59030, Türkiye
| | - Gül Merve Yalcin-Ülker
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Istanbul Okan University, Istanbul 34947, Türkiye
| | - Murat Günbatan
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Istanbul Okan University, Istanbul 34947, Türkiye
| | - Merva Soluk-Tekkesin
- Department of Tumour Pathology, Institute of Oncology, Istanbul University, Istanbul 34093, Türkiye
| | - Ceyda Özcakir-Tomruk
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Yeditepe University, Istanbul 34728, Türkiye
| |
Collapse
|
12
|
Lee HY, Kim DS, Hwang GY, Lee JK, Lee HL, Jung JW, Hwang SY, Baek SW, Yoon SL, Ha Y, Kim KN, Han I, Han DK, Lee CK. Multi-modulation of immune-inflammatory response using bioactive molecule-integrated PLGA composite for spinal fusion. Mater Today Bio 2023; 19:100611. [PMID: 36969699 PMCID: PMC10034518 DOI: 10.1016/j.mtbio.2023.100611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/11/2023] [Indexed: 03/15/2023] Open
Abstract
Despite current developments in bone substitute technology for spinal fusion, there is a lack of adequate materials for bone regeneration in clinical applications. Recombinant human bone morphogenetic protein-2 (rhBMP-2) is commercially available, but a severe inflammatory response is a known side effect. Bone graft substitutes that enhance osteogenesis without adverse effects are needed. We developed a bioactive molecule-laden PLGA composite with multi-modulation for bone fusion. This bioresorbable composite scaffold was considered for bone tissue engineering. Among the main components, magnesium hydroxide (MH) aids in reduction of acute inflammation affecting disruption of new bone formation. Decellularized bone extracellular matrix (bECM) and demineralized bone matrix (DBM) composites were used for osteoconductive and osteoinductive activities. A bioactive molecule, polydeoxyribonucleotide (PDRN, PN), derived from trout was used for angiogenesis during bone regeneration. A nano-emulsion method that included Span 80 was used to fabricate bioactive PLGA-MH-bECM/DBM-PDRN (PME2/PN) composite to obtain a highly effective and safe scaffold. The synergistic effect provided by PME2/PN improved not only osteogenic and angiogenic gene expression for bone fusion but also improved immunosuppression and polarization of macrophages that were important for bone tissue repair, using a rat model of posterolateral spinal fusion (PLF). It thus had sufficient biocompatibility and bioactivity for spinal fusion.
Collapse
Affiliation(s)
- Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Gwang Yong Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Sae Yeon Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Sol lip Yoon
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bungdang Medical Center, Gyeonggi-do, 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- Corresponding author.
| | - Chang Kyu Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Corresponding author.
| |
Collapse
|
13
|
Escaloni J, Mazloomdoost D, Young I. Novel Orthobiologic Preparation and Regenerative Rehabilitation of a Complex Shoulder Injury in a Competitive Adolescent Female Athlete. Int J Sports Phys Ther 2023; 18:240-252. [PMID: 36793563 PMCID: PMC9897030 DOI: 10.26603/001c.68143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 12/23/2022] [Indexed: 02/05/2023] Open
Abstract
Background Platelet-rich plasma (PRP) and prolotherapy have resulted in promising outcomes in patients with various types of shoulder injuries. However, there is a lack of preliminary evidence supporting preparation of PRP production, timely application of these therapies and regenerative rehabilitation protocols. The purpose of this case report is to describe the distinct method including orthobiologic preparation, tissue-specific treatment and regenerative rehabilitation of an athlete with a complex shoulder injury. Case Presentation A 15y/o competitive female wrestler with a complex shoulder injury presented to the clinic after unsuccessful conservative rehabilitation. Unique methods were incorporated to optimize PRP production, specific tissue healing and regenerative rehabilitation. Multiple injuries required different orthobiologic interventions at different time frames, in order to promote optimal healing and stability of the shoulder. Outcomes The described interventions resulted in successful outcomes including pain, disability, full return to sport, and regenerative tissue healing confirmed with diagnostic imaging. Level of Evidence 5.
Collapse
Affiliation(s)
- James Escaloni
- American Academy of Manipulative Therapy
- Wellward Regenerative Medicine
| | | | - Ian Young
- American Academy of Manipulative Therapy
- Tybee Wellness & Osteopractic
| |
Collapse
|
14
|
Nurrachman AS, Azhari A, Epsilawati L, Pramanik F. Temporal Pattern of micro-CT Angiography Vascular Parameters and VEGF mRNA Expression in Fracture Healing: a Radiograph and Molecular Comparison. Eur J Dent 2023. [PMID: 36716788 DOI: 10.1055/s-0042-1757466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis plays an important role in fracture healing with vascular endothelial growth factor (VEGF) as the main protein involved. Micro-computed tomography (CT) angiography may be used to analyze this revascularization with several parameters such as number of branches, total volume, and diameter. This systematic review is aimed to assess available studies on the temporal pattern of vascular imaging on micro-CT angiographs, especially in terms of the number of branches, total volume, and diameter as well as the temporal pattern of VEGF mRNA expression as the molecular comparison during bone fracture healing. This review was conducted according to the PRISMA guidelines. Electronic database searches were performed using PubMed, ProQuest, ScienceDirect, EBSCOhost, Taylor & Francis Online, and hand searching. The search strategy and keywords were adjusted to each database using the Boolean operators and other available limit functions to identify most relevant articles based on our inclusion and exclusion criteria. Screening and filtration were done in several stages by removing the duplicates and analyzing each title, abstract, and full-text in all included entries. Data extraction was done for syntheses to summarize the temporal pattern of each parameter. A total of 28 articles were eligible and met all criteria, 11 articles were synthesized in its angiograph's analysis, 16 articles were synthesized in its VEGF mRNA expression analysis, and 1 article had both parameters analyzed. The overall temporal pattern of both three micro-CT angiographic parameters and VEGF mRNA expression was in line qualitatively. The number of branches, total volume, and diameter of the blood vessels in micro-CT angiography showed an exponential rise at week 2 and decline at week 3 of fracture healing, with the VEGF mRNA expression concurrently showing a consistent pattern in the phase.
Collapse
Affiliation(s)
- Aga Satria Nurrachman
- Department of Oral and Maxillofacial Radiology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Azhari Azhari
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| | - Lusi Epsilawati
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| | - Farina Pramanik
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| |
Collapse
|
15
|
Guo R, Zhuang H, Chen X, Ben Y, Fan M, Wang Y, Zheng P. Tissue engineering in growth plate cartilage regeneration: Mechanisms to therapeutic strategies. J Tissue Eng 2023; 14:20417314231187956. [PMID: 37483459 PMCID: PMC10359656 DOI: 10.1177/20417314231187956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023] Open
Abstract
The repair of growth plate injuries is a highly complex process that involves precise spatiotemporal regulation of multiple cell types. While significant progress has been made in understanding the pathological mechanisms underlying growth plate injuries, effectively regulating this process to regenerate the injured growth plate cartilage remains a challenge. Tissue engineering technology has emerged as a promising therapeutic approach for achieving tissue regeneration through the use of functional biological materials, seed cells and biological factors, and it is now widely applied to the regeneration of bone and cartilage. However, due to the unique structure and function of growth plate cartilage, distinct strategies are required for effective regeneration. Thus, this review provides an overview of current research on the application of tissue engineering to promote growth plate regeneration. It aims to elucidates the underlying mechanisms by which tissue engineering promotes growth plate regeneration and to provide novel insights and therapeutic strategies for future research on the regeneration of growth plate.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pengfei Zheng
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Aryani T, Budiatin AS, Samirah, Maulidina A, Firdaus AI, Gani MA, Nisak K, Khotib J, Syukriya AJ. The administration of bovine hydroxyapatite-alendronate implant accelerates bone defect healing in an osteoporotic rat. Technol Health Care 2023; 31:1747-1757. [PMID: 37092192 DOI: 10.3233/thc-220612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
BACKGROUND Bone fracture is the main consequence of osteoporosis, which may become a neglected disease. OBJECTIVE This study aims to fabricate bovine hydroxyapatite-gelatine (BHA-GEL) based bone-implant with alendronate (ALE) in vivo. METHODS Wistar rats were used for an osteoporotic animal model induced by ovariectomy. There were three groups: negative control, BHA-GEL implant, and BHA-GEL-ALE implant. Each group performed a defect by drilling the femur (diameter of 2.2 mm and depth of 2 mm). Observations on the closure of bone defects were performed by X-ray radiography at the second and sixth week after surgery. The mechanism of bone healing was observed by using hematoxylin-eosin (HE) staining and immunohistochemical technique with anti-vascular endothelial growth factor (VEGF) and anti-alkaline phosphatase (ALP) antibodies. RESULTS The radiograph examination showed the implanted group had accelerated bone growth. In addition, the osteoblast, osteoclast and osteocyte had accelerated migration to the defect area. Moreover, the immunoreactive score (IRS) of VEGF at the sixth week in the BHA-GEL-ALE group was lower than the other groups. Meanwhile, the IRS of ALP in BHA-GEL-ALE was higher compared to other groups. CONCLUSION The BHA-GEL-ALE implant accelerates the healing of bone defect in the osteoporotic rat by increasing the ALP expression and the total number of cells.
Collapse
Affiliation(s)
- Toetik Aryani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Aniek Setiya Budiatin
- Department of Clinical Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Samirah
- Department of Clinical Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Aulia Maulidina
- Department of Clinical Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Aulia Intan Firdaus
- Department of Clinical Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Maria Apriliani Gani
- Doctoral Programme in Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Khoirotin Nisak
- Department of Clinical Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Junaidi Khotib
- Department of Clinical Pharmacy, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Alvi Jauharotus Syukriya
- Master Programme in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
Shen N, Maggio M, Woods I, C. Lowry M, Almasri R, Gorgun C, Eichholz K, Stavenschi E, Hokamp K, Roche F, O’Driscoll L, Hoey D. Mechanically activated mesenchymal-derived bone cells drive vessel formation via an extracellular vesicle mediated mechanism. J Tissue Eng 2023; 14:20417314231186918. [PMID: 37654438 PMCID: PMC10467237 DOI: 10.1177/20417314231186918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/23/2023] [Indexed: 09/02/2023] Open
Abstract
Blood vessel formation is an important initial step for bone formation during development as well as during remodelling and repair in the adult skeleton. This results in a heavily vascularized tissue where endothelial cells and skeletal cells are constantly in crosstalk to facilitate homeostasis, a process that is mediated by numerous environmental signals, including mechanical loading. Breakdown in this communication can lead to disease and/or poor fracture repair. Therefore, this study aimed to determine the role of mature bone cells in regulating angiogenesis, how this is influenced by a dynamic mechanical environment, and understand the mechanism by which this could occur. Herein, we demonstrate that both osteoblasts and osteocytes coordinate endothelial cell proliferation, migration, and blood vessel formation via a mechanically dependent paracrine mechanism. Moreover, we identified that this process is mediated via the secretion of extracellular vesicles (EVs), as isolated EVs from mechanically stimulated bone cells elicited the same response as seen with the full secretome, while the EV-depleted secretome did not elicit any effect. Despite mechanically activated bone cell-derived EVs (MA-EVs) driving a similar response to VEGF treatment, MA-EVs contain minimal quantities of this angiogenic factor. Lastly, a miRNA screen identified mechanoresponsive miRNAs packaged within MA-EVs which are linked with angiogenesis. Taken together, this study has highlighted an important mechanism in osteogenic-angiogenic coupling in bone and has identified the mechanically activated bone cell-derived EVs as a therapeutic to promote angiogenesis and potentially bone repair.
Collapse
Affiliation(s)
- N. Shen
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - M. Maggio
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - I. Woods
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - M. C. Lowry
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, and Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - R. Almasri
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, and Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - C. Gorgun
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - K.F. Eichholz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - E. Stavenschi
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - K. Hokamp
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, College Green, Dublin, Ireland
| | - F.M. Roche
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, College Green, Dublin, Ireland
| | - L. O’Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, and Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - D.A. Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
18
|
Chen YC, Fu YS, Tsai SW, Wu PK, Chen CM, Chen WM, Chen CF. IL-1b in the Secretomes of MSCs Seeded on Human Decellularized Allogeneic Bone Promotes Angiogenesis. Int J Mol Sci 2022; 23:15301. [PMID: 36499629 PMCID: PMC9737155 DOI: 10.3390/ijms232315301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis plays an important role in the development of bone and bone regeneration to provide the required molecules. Mesenchymal stem cells (MSCs) are pluripotent, self-renewing, and spindle-shaped cells, which can differentiate into multiple lineages such as chondrocytes, osteocytes, and adipocytes. MSCs derived from bone marrow (BMMSCs), adipose tissue (ADMSCs), and Wharton's jelly (UCMSCs) are popular in the field of tissue regeneration. MSCs have been proposed that can promote bone regeneration by enhancing vascularization. In this study, the angiogenic potential of secretomes of undifferentiated and osteo-differentiated BMMSCs, ADMSCs, and UCMSCs seeded on human decellularized allogeneic bone were compared. Human umbilical vein endothelial cells (HUVECs) were treated with MSC secretomes. Cell growth, cell migration, and angiogenesis of HUVECs were analyzed by MTT, wound healing, and tube formation assays. Angiogenic gene expression levels of MSCs were evaluated using real-time quantitative PCR. Antibody neutralization was performed to validate the candidate target. Our study demonstrates that the angiogenic gene expression profile is tissue-dependent and the angiogenic ability of secretomes is independent of the state of differentiation. We also explore that IL-1b is important for MSC angiogenic potential. Taken together, this study proves that IL-1b in the secretomes plays a vital role in angiogenesis.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yu-Show Fu
- Department of Anatomy and Cell Biology, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Shang-Wen Tsai
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Po-Kuei Wu
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chao-Ming Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Wei-Ming Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Cheng-Fong Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
19
|
Kefir peptides promote osteogenic differentiation to enhance bone fracture healing in rats. Life Sci 2022; 310:121090. [DOI: 10.1016/j.lfs.2022.121090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022]
|
20
|
Elghareeb MM, Elshopakey GE, Elkhooly TA, Salama B, Samy A, Bazer FW, Elmetwally MA, Almutairi MH, Aleya L, Abdel-Daim MM, Rezk S. Estradiol and zinc-doped nano hydroxyapatite as therapeutic agents in the prevention of osteoporosis; oxidative stress status, inflammation, bone turnover, bone mineral density, and histological alterations in ovariectomized rats. Front Physiol 2022; 13:989487. [PMID: 36200054 PMCID: PMC9527315 DOI: 10.3389/fphys.2022.989487] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoporosis (OP) is a serious health problem, and the most popular therapeutic strategy for OP is hormone replacement (estrogen); however, it increases the risk of reproductive cancers. Hydroxyapatite (HA) nanoparticles have a similar chemical structure to the bone mineral component and can be used as a new remedy for OP. This study was designed to investigate the osteoporosis-protective potential of nano zinc hydroxyapatite (ZnHA-NPs) and/or estradiol (E2) combined therapy. A total of 35 adult female rats were assigned into five groups (n = 7): 1) control group; 2) ovariectomized group (OVX); 3) OVX received oral estradiol replacement therapy (OVX/E2); 4) OVX received ZnHA replacement therapy (OVX/ZnHA); and 5) OVX received both estradiol and ZnHA-NPs combined therapy (OVX/E2+ZnHA). After 3 months of treatment, serum bone markers and estrogen level, oxidative/antioxidant, and inflammatory cytokines were determined. Additionally, femoral expression of estrogen receptors alpha and beta (ESR1; ESR2), receptor activator of nuclear factor-kappa B (RANKL) ligand, osteoprotegerin (OPG), bone mineral density (BMD), histological alterations, and immunohistochemical expression of vascular endothelial growth factor (VEGF) and proliferating cell nuclear antigen (PCNA) were assessed. ALP, PINP, Ca, and P concentrations improved significantly (p < 0.05) in all treatment groups, especially in the OVX/E + ZnHA group. MDA and NO were higher in OVX rats, while SOD activity and GSH were lower (p < 0.05). E2 alone or with ZnHA-NPs restored the estimated antioxidant molecules and cytokines toward normal levels in OVX rats (p < 0.05). On the other hand, E2 and ZnHA increased OPG and OC expression in femurs while decreasing ESR1, ESR2, and NF-kB expression (p < 0.05). The combination treatment was superior in the restoration of normal femoral histoarchitecture and both cortical and trabecular BMD (p < 0.05). Overall, the combined therapy of OVX/E2+ZnHA was more effective than the individual treatments in attenuating excessive bone turnover and preventing osteoporosis.
Collapse
Affiliation(s)
- Mona M. Elghareeb
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Gehad E. Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Tarek A. Elkhooly
- Nanomedicine Research Unit, Faculty of Medicine, Delta University for Science and Technology, Belqas, Egypt
- Refractories, Ceramics, and Building Materials Department, National Research Centre, Giza, Egypt
- Department of Physics, Faculty of Science, New Mansoura University, New Mansoura City, Egypt
| | - Basma Salama
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Alaa Samy
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Mohammed A Elmetwally
- Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mikhlid H. Almutairi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Lotfi Aleya
- UMR CNRS 6249, Chrono-Environnement Laboratory, Bourgogne, Franche-Comté University, Besançon, France
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Shaymaa Rezk
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
21
|
Riquelme-Gallego B, García-Molina L, Cano-Ibáñez N, Andújar-Vera F, González-Salvatierra S, García-Fontana C, Bueno-Cavanillas A, Muñoz-Torres M, García-Fontana B. Undercarboxylated Osteocalcin: A Promising Target for Early Diagnosis of Cardiovascular and Glycemic Disorders in Patients with Metabolic Syndrome: A Pilot Study. Nutrients 2022; 14:nu14142991. [PMID: 35889946 PMCID: PMC9321347 DOI: 10.3390/nu14142991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/13/2022] [Accepted: 07/16/2022] [Indexed: 12/04/2022] Open
Abstract
Lifestyle changes are causing an exponential increase in the prevalence of obesity and metabolic syndrome (MetS) worldwide. The most frequent complications of these are the development of diabetes (T2D) and cardiovascular disease (CVD). Accurate tools are needed to classify the cardiovascular risk (CVR) in the MetS population. In recent years, numerous biomarkers of bone metabolism have been associated with CVR. The aim of this study was to determine the levels of undercarboxylated osteocalcin (ucOC) in a cohort of patients with MetS and to analyse its association with MetS parameters and CVR as well as with T2D prevalence. A longitudinal study was conducted in which a MetS population was followed for one year. Weight change, adherence to the Mediterranean diet (MedDiet), ucOC levels, MetS parameters and CVR were analysed and CVR was calculated using different scores. Our results showed a decrease of CVR associated with a better adherence to the MetDiet resulting in higher HDL-C and ucOC levels though the improvement of MetS risk factors. This bone protein appeared as a potential biomarker to classify CVR in the MetS population, especially for MetS patients without prevalent T2D. Furthermore, ucOC serum levels could be good predictors of T2D prevalence.
Collapse
Affiliation(s)
- Blanca Riquelme-Gallego
- Department of Preventive Medicine and Public Health, University of Granada, 18016 Granada, Spain; (B.R.-G.); (L.G.-M.); (N.C.-I.); (A.B.-C.)
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
| | - Laura García-Molina
- Department of Preventive Medicine and Public Health, University of Granada, 18016 Granada, Spain; (B.R.-G.); (L.G.-M.); (N.C.-I.); (A.B.-C.)
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
| | - Naomi Cano-Ibáñez
- Department of Preventive Medicine and Public Health, University of Granada, 18016 Granada, Spain; (B.R.-G.); (L.G.-M.); (N.C.-I.); (A.B.-C.)
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Francisco Andújar-Vera
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain;
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), 18014 Granada, Spain
| | - Sheila González-Salvatierra
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Cristina García-Fontana
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Correspondence: (C.G.-F.); (M.M.-T.); (B.G.-F.); Tel.: +34-958023460 (C.G.-F.); +34-958246124 (M.M.-T.); +34-958023460 (B.G.-F.)
| | - Aurora Bueno-Cavanillas
- Department of Preventive Medicine and Public Health, University of Granada, 18016 Granada, Spain; (B.R.-G.); (L.G.-M.); (N.C.-I.); (A.B.-C.)
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Manuel Muñoz-Torres
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
- Department of Medicine, University of Granada, 18016 Granada, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Correspondence: (C.G.-F.); (M.M.-T.); (B.G.-F.); Tel.: +34-958023460 (C.G.-F.); +34-958246124 (M.M.-T.); +34-958023460 (B.G.-F.)
| | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria (ibs.GRANADA), 18014 Granada, Spain;
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Correspondence: (C.G.-F.); (M.M.-T.); (B.G.-F.); Tel.: +34-958023460 (C.G.-F.); +34-958246124 (M.M.-T.); +34-958023460 (B.G.-F.)
| |
Collapse
|
22
|
Naguib M, Ali N, ElSaraf N, Rashed L, Azzam H. Does Serum Osteocalcin Level Affect Carotid Atherosclerosis in Post-Menopausal Diabetic Females? A Case-Control Study. Int J Gen Med 2022; 15:4513-4523. [PMID: 35509602 PMCID: PMC9059994 DOI: 10.2147/ijgm.s353492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To determine the association between serum osteocalcin and carotid intima media thickness (CIMT) in a group of post-menopausal females with type 2 diabetes (T2DM). Patients and Methods This cross-sectional study enrolled 75 postmenopausal women with T2DM and 40 age matched postmenopausal healthy females. Age, body mass index, blood pressure were recorded for all subjects. Laboratory tests including fasting blood glucose (FBG), glycated hemoglobin (HbA1c) and lipid profile were measured. Serum osteocalcin was measured using ELISA. Bone mineral density (BMD) was measured by DEXA scan. CIMT was assessed with B-mode ultrasonography. Results Patients with T2DM had significantly lower serum osteocalcin compared to control (63.73±27.20 vs 136.16±21.96 pg/mL, P<0.001). Patients with osteoporosis had significantly lower osteocalcin level compared to those with normal BMD. Patients with T2DM had a significant negative correlation between serum osteocalcin and CIMT (r= -0.332; P=0.003), FBG (r= -0.732; P< 0.001), HbA1c (r=-0.672; P< 0.001), and HOMA-IR (r= -0.672; P< 0.001). However, multiple linear regression analysis revealed that CIMT in patients with diabetes was only significantly associated with age (P= 0.001), duration of diabetes (P< 0.001), SBP (P< 0.001), HOMA-IR (P=0.033), LDL (P=0.005), and HDL (P< 0.001). Furthermore, serum insulin (β= -0.183, P=0.033), FBG (β= -0.604, P< 0.001) and LDL (β= -0.195, P= 0.02) were independently negatively correlated with serum osteocalcin. Conclusion In this study, Postmenopausal women with diabetes had significantly lower osteocalcin compared to non-diabetic women. Although serum osteocalcin was negatively correlated with CIMT, multivariate regression analysis revealed that osteocalcin level was only independently related to worse glycemic parameters in postmenopausal women with T2DM.
Collapse
Affiliation(s)
- Mervat Naguib
- Diabetes and Endocrinology Unite, Internal Medicine Department, Faculty of Medicine Kasr Al-Ainy Hospital Cairo University, Cairo, Egypt
| | - Nadia Ali
- Internal Medicine Department, Ahmed Maher Hospital, Cairo, Egypt
| | - Nashwa ElSaraf
- Internal Medicine Department, Faculty of Medicine Kasr Al-Ainy Hospital Cairo University, Cairo, Egypt
| | - Lila Rashed
- Department of Biochemistry, Faculty of Medicine Cairo University, Cairo, Egypt
| | - Hebatoallah Azzam
- Radiology Department, Faculty of Medicine Kasr Al-Ainy Hospital Cairo University, Cairo, Egypt
| |
Collapse
|
23
|
Long non-coding RNA HCAR promotes endochondral bone repair by upregulating VEGF and MMP13 in hypertrophic chondrocyte through sponging miR-15b-5p. Genes Dis 2022; 9:456-465. [PMID: 35224160 PMCID: PMC8843884 DOI: 10.1016/j.gendis.2020.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/08/2020] [Accepted: 07/30/2020] [Indexed: 01/30/2023] Open
Abstract
Endochondral bone formation is an important route for bone repair. Although emerging evidence has revealed the functions of long non-coding RNAs (lncRNAs) in bone and cartilage development, the effect of lncRNAs in endochondral bone repair is still largely unknown. Here, we identified a lncRNA, named Hypertrophic Chondrocyte Angiogenesis-related lncRNA (HCAR), and proved it to promote the endochondral bone repair by upregulating the expression of matrix metallopeptidase 13 (Mmp13) and vascular endothelial growth factor α (Vegfa) in hypertrophic chondrocytes. Lnc-HCAR knockdown in hypertrophic chondrocytes restrained the cartilage matrix remodeling and decrease the CD31hiEmcnhi vessels number in a bone repair model. Mechanistically, we proved that lnc-HCAR was mainly enriched in the cytoplasm using fluorescence in situ hybridization (FISH) assay, and it acted as a molecular sponge for miR-15b-5p. Further, in hypertrophic chondrocytes, lnc-HCAR competitively bound to miR-15b-5p to increase Vegfa and Mmp13 expression. Our results proved that lncRNA is deeply involved in endochondral bone repair, which will provide a new theoretical basis for future strategies for promoting fracture healing.
Collapse
|
24
|
Abstract
Cachexia, a wasting syndrome that is often associated with cancer, is one of the primary causes of death in cancer patients. Cancer cachexia occurs largely due to systemic metabolic alterations stimulated by tumors. Despite the prevalence of cachexia, our understanding of how tumors interact with host tissues and how they affect metabolism is limited. Among the challenges of studying tumor-host tissue crosstalk are the complexity of cancer itself and our insufficient knowledge of the factors that tumors release into the blood. Drosophila is emerging as a powerful model in which to identify tumor-derived factors that influence systemic metabolism and tissue wasting. Strikingly, studies that are characterizing factors derived from different fly tumor cachexia models are identifying both common and distinct cachectic molecules, suggesting that cachexia is more than one disease and that fly models can help identify these differences. Here, we review what has been learned from studies of tumor-induced organ wasting in Drosophila and discuss the open questions.
Collapse
Affiliation(s)
- Ying Liu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro Saavedra
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
| |
Collapse
|
25
|
Freischmidt H, Armbruster J, Rothhaas C, Titze N, Guehring T, Nurjadi D, Sonntag R, Schmidmaier G, Grützner PA, Helbig L. Treatment of Infection-Related Non-Unions with Bioactive Glass-A Promising Approach or Just Another Method of Dead Space Management? MATERIALS 2022; 15:ma15051697. [PMID: 35268930 PMCID: PMC8911496 DOI: 10.3390/ma15051697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 12/17/2022]
Abstract
The treatment of infected and non-infected non-unions remains a major challenge in trauma surgery. Due to the limited availability of autologous bone grafts and the need for local anti-infective treatment, bone substitutes have been the focus of tissue engineering for years. In this context, bioactive glasses are promising, especially regarding their anti-infective potential, which could reduce the need for local and systemic treatment with conventional antibiotics. The aim of this study was to investigate the osteoinductive and osteoconductive effects, as well as the anti-infectious potential, of S53P4 using a standardized non-union model, which had not been investigated previously. Using an already established sequential animal model in infected and non-infected rat femora, we were able to investigate bioactive glass S53P4 under realistic non-union conditions regarding its osteoinductive, osteoconductive and anti-infective potential with the use of µCT scans, biomechanical testing and histological, as well as microbiological, analysis. Although S53P4 did not lead to a stable union in the non-infected or the infected setting, µCT analysis revealed an osteoinductive effect of S53P4 under non-infected conditions, which was diminished under infected conditions. The osteoconductive effect of S53P4 remained almost negligible in histological analysis, even 8 weeks after treatment. Additionally, the expected anti-infective effect could not be demonstrated. Our data suggested that S53P4 should not be used in infected non-unions, especially in those with large bone defects.
Collapse
Affiliation(s)
- Holger Freischmidt
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Jonas Armbruster
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Catharina Rothhaas
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Nadine Titze
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Thorsten Guehring
- Trauma Centre, Hospital Paulinenhilfe Stuttgart at Tübingen University Hospital, Rosenbergstr. 38, 70176 Stuttgart, Germany;
| | - Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany;
| | - Robert Sonntag
- Laboratory of Biomechanics and Implant Research, Clinic for Orthopedics and Trauma Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany;
| | - Gerhard Schmidmaier
- Clinic for Orthopedics and Trauma Surgery, Center for Orthopedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany;
| | - Paul Alfred Grützner
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Lars Helbig
- Clinic for Orthopedics and Trauma Surgery, Center for Orthopedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany;
- Correspondence:
| |
Collapse
|
26
|
Orth M, Fritz T, Stutz J, Scheuer C, Ganse B, Bullinger Y, Lee JS, Murphy WL, Laschke MW, Menger MD, Pohlemann T. Local Application of Mineral-Coated Microparticles Loaded With VEGF and BMP-2 Induces the Healing of Murine Atrophic Non-Unions. Front Bioeng Biotechnol 2022; 9:809397. [PMID: 35087807 PMCID: PMC8787303 DOI: 10.3389/fbioe.2021.809397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022] Open
Abstract
Deficient angiogenesis and disturbed osteogenesis are key factors for the development of nonunions. Mineral-coated microparticles (MCM) represent a sophisticated carrier system for the delivery of vascular endothelial growth factor (VEGF) and bone morphogenetic protein (BMP)-2. In this study, we investigated whether a combination of VEGF- and BMP-2-loaded MCM (MCM + VB) with a ratio of 1:2 improves bone repair in non-unions. For this purpose, we applied MCM + VB or unloaded MCM in a murine non-union model and studied the process of bone healing by means of radiological, biomechanical, histomorphometric, immunohistochemical and Western blot techniques after 14 and 70 days. MCM-free non-unions served as controls. Bone defects treated with MCM + VB exhibited osseous bridging, an improved biomechanical stiffness, an increased bone volume within the callus including ongoing mineralization, increased vascularization, and a histologically larger total periosteal callus area consisting predominantly of osseous tissue when compared to defects of the other groups. Western blot analyses on day 14 revealed a higher expression of osteoprotegerin (OPG) and vice versa reduced expression of receptor activator of NF-κB ligand (RANKL) in bone defects treated with MCM + VB. On day 70, these defects exhibited an increased expression of erythropoietin (EPO), EPO-receptor and BMP-4. These findings indicate that the use of MCM for spatiotemporal controlled delivery of VEGF and BMP-2 shows great potential to improve bone healing in atrophic non-unions by promoting angiogenesis and osteogenesis as well as reducing early osteoclast activity.
Collapse
Affiliation(s)
- M Orth
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.,Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - T Fritz
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - J Stutz
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.,Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - C Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - B Ganse
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.,Werner Siemens Endowed Chair of Innovative Implant Development (Fracture Healing), Saarland University, Homburg, Germany
| | - Y Bullinger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - J S Lee
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - W L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - M W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - M D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - T Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
27
|
Taohong Siwu-Containing Serum Enhances Angiogenesis in Rat Aortic Endothelial Cells by Regulating the VHL/HIF-1 α/VEGF Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6610116. [PMID: 34853600 PMCID: PMC8629617 DOI: 10.1155/2021/6610116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/27/2021] [Accepted: 10/01/2021] [Indexed: 01/15/2023]
Abstract
Background The incidence of bone fracture and bone-related diseases is increasing every year. Angiogenesis plays a vital role in fracture healing and bone repair. This study assessed the benefits of Taohong Siwu (TSW) decoction on angiogenesis in isolated rat aortic endothelial cells (RAEC) treated with TSW-containing serum. Methods The components of TSW decoction were analyzed by liquid chromatography-mass spectrometry (LC-MS). TSW-containing serum was prepared by gavage of TSW decoction to Sprague-Dawley (SD) rats. The effects of TSW-containing serum on the viability, migration, wound healing, and angiogenesis of RAEC were detected by the MTT, transwell, wound healing, and Matrigel lumen formation assays, respectively. In addition, the effects of an HIF-1α inhibitor on TSW-containing serum-induced RAEC were also assessed. The effects of TSW-containing serum on the expression of the HIF-1α signaling pathway were evaluated by qRT-PCR and western blot analysis. Results LC-MS revealed that TSW decoction primarily contained isomaltulose, choline, D-gluconic acid, L-pipecolic acid, hypotaurine, albiflorin, and tryptophan. TSW-containing serum significantly increased the viability, migration, wound healing, and angiogenesis of RAEC in a dose-dependent manner. Furthermore, our results demonstrated that HIF-1α and VEGF expressions were increased in the cells of TSW-containing serum groups, whereas VHL expression was decreased. The effects of TSW-containing serum were reversed by treatment with an HIF-1α inhibitor. Conclusion These results suggested that TSW decoction enhanced angiogenesis by regulating the VHL/HIF-1α/VEGF signaling pathway.
Collapse
|
28
|
Yang Y, Lee EH, Yang Z. Hypoxia conditioned mesenchymal stem cells in tissue regeneration application. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:966-977. [PMID: 34569290 DOI: 10.1089/ten.teb.2021.0145] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have been demonstrated as promising cell sources for tissue regeneration due to their capability of self-regeneration, differentiation and immunomodulation. MSCs also exert extensive paracrine effects through release of trophic factors and extracellular vesicles. However, despite extended exploration of MSCs in pre-clinical studies, the results are far from satisfactory due to the poor engraftment and low level of survival after implantation. Hypoxia preconditioning has been proposed as an engineering approach to improve the therapeutic potential of MSCs. During in vitro culture, hypoxic conditions can promote MSC proliferation, survival and migration through various cellular responses to the reduction of oxygen tension. The multilineage differentiation potential of MSCs is altered under hypoxia, with consistent reports of enhanced chondrogenesis. Hypoxia also stimulates the paracrine activities of MSCs and increases the production of secretome both in terms of soluble factors as well as extracellular vesicles. The secretome from hypoxia preconditioned MSCs play important roles in promoting cell proliferation and migration, enhancing angiogenesis while inhibiting apoptosis and inflammation. In this review, we summarise current knowledge of hypoxia-induced changes in MSCs and discuss the application of hypoxia preconditioned MSCs as well as hypoxic secretome in different kinds of disease models.
Collapse
Affiliation(s)
- Yanmeng Yang
- National University of Singapore, 37580, Orthopaedic Surgery, 27 Medical Drive, Singapore, Singapore, 117510;
| | - Eng Hin Lee
- National University of Singapore, Department of Orthopaedic Surgery, 1E Kent Ridge Road, NUHS Tower Block, Level 11, Singapore, Singapore, 119228;
| | - Zheng Yang
- National University of Singapore, Life Sciences Institute, Singapore, Singapore;
| |
Collapse
|
29
|
Systemic Administration of PTH Supports Vascularization in Segmental Bone Defects Filled with Ceramic-Based Bone Graft Substitute. Cells 2021; 10:cells10082058. [PMID: 34440827 PMCID: PMC8392660 DOI: 10.3390/cells10082058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/29/2021] [Accepted: 08/07/2021] [Indexed: 12/19/2022] Open
Abstract
Non-unions continue to present a challenge to trauma surgeons, as current treatment options are limited, duration of treatment is long, and the outcome often unsatisfactory. Additionally, standard treatment with autologous bone grafts is associated with comorbidity at the donor site. Therefore, alternatives to autologous bone grafts and further therapeutic strategies to improve on the outcome and reduce cost for care providers are desirable. In this study in Sprague–Dawley rats we employed a recently established sequential defect model, which provides a platform to test new potential therapeutic strategies on non-unions while gaining mechanistic insight into their actions. The effects of a combinatorial treatment of a bone graft substitute (HACaS+G) implantation and systemic PTH administration was assessed by µ-CT, histological analysis, and bio-mechanical testing and compared to monotreatment and controls. Although neither PTH alone nor the combination of a bone graft substitute and PTH led to the formation of a stable union, our data demonstrate a clear osteoinductive and osteoconductive effect of the bone graft substitute. Additionally, PTH administration was shown to induce vascularization, both as a single adjuvant treatment and in combination with the bone graft substitute. Thus, systemic PTH administration is a potential synergistic co-treatment to bone graft substitutes.
Collapse
|
30
|
Bornert F, Clauss F, Hua G, Idoux-Gillet Y, Keller L, Fernandez De Grado G, Offner D, Smaida R, Wagner Q, Fioretti F, Kuchler-Bopp S, Schulz G, Wenzel W, Gentile L, Risser L, Müller B, Huck O, Benkirane-Jessel N. Mechanistic Illustration: How Newly-Formed Blood Vessels Stopped by the Mineral Blocks of Bone Substitutes Can Be Avoided by Using Innovative Combined Therapeutics. Biomedicines 2021; 9:952. [PMID: 34440156 PMCID: PMC8394928 DOI: 10.3390/biomedicines9080952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 08/01/2021] [Indexed: 12/30/2022] Open
Abstract
One major limitation for the vascularization of bone substitutes used for filling is the presence of mineral blocks. The newly-formed blood vessels are stopped or have to circumvent the mineral blocks, resulting in inefficient delivery of oxygen and nutrients to the implant. This leads to necrosis within the implant and to poor engraftment of the bone substitute. The aim of the present study is to provide a bone substitute currently used in the clinic with suitably guided vascularization properties. This therapeutic hybrid bone filling, containing a mineral and a polymeric component, is fortified with pro-angiogenic smart nano-therapeutics that allow the release of angiogenic molecules. Our data showed that the improved vasculature within the implant promoted new bone formation and that the newly-formed bone swapped the mineral blocks of the bone substitutes much more efficiently than in non-functionalized bone substitutes. Therefore, we demonstrated that our therapeutic bone substitute is an advanced therapeutical medicinal product, with great potential to recuperate and guide vascularization that is stopped by mineral blocks, and can improve the regeneration of critical-sized bone defects. We have also elucidated the mechanism to understand how the newly-formed vessels can no longer encounter mineral blocks and pursue their course of vasculature, giving our advanced therapeutical bone filling great potential to be used in many applications, by combining filling and nano-regenerative medicine that currently fall short because of problems related to the lack of oxygen and nutrients.
Collapse
Affiliation(s)
- Fabien Bornert
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - François Clauss
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Guoqiang Hua
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laetitia Keller
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Gabriel Fernandez De Grado
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Damien Offner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Rana Smaida
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Quentin Wagner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Florence Fioretti
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Georg Schulz
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Wolfgang Wenzel
- Institute of Nanotechnology, Karlsruhe Institute of Technology, Campus North, Building 640, DE-76131 Karlsruhe, Germany;
| | - Luca Gentile
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laurent Risser
- Toulouse Institute of Mathematics, UMR 5219 University of Toulouse, CNRS UPS IMT, 31062 Toulouse, France;
| | - Bert Müller
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| |
Collapse
|
31
|
Cacciottola L, Donnez J, Dolmans MM. Ovarian tissue damage after grafting: systematic review of strategies to improve follicle outcomes. Reprod Biomed Online 2021; 43:351-369. [PMID: 34384692 DOI: 10.1016/j.rbmo.2021.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/14/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022]
Abstract
Frozen-thawed human ovarian tissue endures large-scale follicle loss in the early post-grafting period, characterized by hypoxia lasting around 7 days. Tissue revascularization occurs progressively through new vessel invasion from the host and neoangiogenesis from the graft. Such reoxygenation kinetics lead to further potential damage caused by oxidative stress. The aim of the present manuscript is to provide a systematic review of proangiogenic growth factors, hormones and various antioxidants administered in the event of ovarian tissue transplantation to protect the follicle pool from depletion by boosting revascularization or decreasing oxidative stress. Although almost all investigated studies revealed an advantage in terms of revascularization and reduction in oxidative stress, far fewer demonstrated a positive impact on follicle survival. As the cascade of events driven by ischaemia after transplantation is a complex process involving numerous players, it appears that acting on specific molecular mechanisms, such as concentrations of proangiogenic growth factors, is not enough to significantly mitigate tissue damage. Strategies exploiting the activated tissue response to ischaemia for tissue healing and remodelling purposes, such as the use of antiapoptotic drugs and adult stem cells, are also discussed in the present review, since they yielded promising results in terms of follicle pool protection.
Collapse
Affiliation(s)
- Luciana Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques Donnez
- Prof. Emeritus, Université Catholique de Louvain, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Department of Gynecology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
32
|
Bhatti FUR, Dadwal UC, Valuch CR, Tewari NP, Awosanya OD, de Andrade Staut C, Sun S, Mendenhall SK, Perugini AJ, Nagaraj RU, Battina HL, Nazzal MK, Blosser RJ, Maupin KA, Childress PJ, Li J, Kacena MA. The effects of high fat diet, bone healing, and BMP-2 treatment on endothelial cell growth and function. Bone 2021; 146:115883. [PMID: 33581374 PMCID: PMC8009863 DOI: 10.1016/j.bone.2021.115883] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023]
Abstract
Angiogenesis is a vital process during the regeneration of bone tissue. The aim of this study was to investigate angiogenesis at the fracture site as well as at distal locations from obesity-induced type 2 diabetic mice that were treated with bone morphogenetic protein-2 (BMP-2, local administration at the time of surgery) to heal a femoral critical sized defect (CSD) or saline as a control. Mice were fed a high fat diet (HFD) to induce a type 2 diabetic-like phenotype while low fat diet (LFD) animals served as controls. Endothelial cells (ECs) were isolated from the lungs (LECs) and bone marrow (BMECs) 3 weeks post-surgery, and the fractured femurs were also examined. Our studies demonstrate that local administration of BMP-2 at the fracture site in a CSD model results in complete bone healing within 3 weeks for all HFD mice and 66.7% of LFD mice, whereas those treated with saline remain unhealed. At the fracture site, vessel parameters and adipocyte numbers were significantly increased in BMP-2 treated femurs, irrespective of diet. At distal sites, LEC and BMEC proliferation was not altered by diet or BMP-2 treatment. HFD increased the tube formation ability of both LECs and BMECs. Interestingly, BMP-2 treatment at the time of surgery reduced tube formation in LECs and humeri BMECs. However, migration of BMECs from HFD mice treated with BMP-2 was increased compared to BMECs from HFD mice treated with saline. BMP-2 treatment significantly increased the expression of CD31, FLT-1, and ANGPT2 in LECs and BMECs in LFD mice, but reduced the expression of these same genes in HFD mice. To date, this is the first study that depicts the systemic influence of fracture surgery and local BMP-2 treatment on the proliferation and angiogenic potential of ECs derived from the bone marrow and lungs.
Collapse
Affiliation(s)
- Fazal Ur Rehman Bhatti
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Ushashi C Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Conner R Valuch
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Nikhil P Tewari
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Olatundun D Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | | | - Seungyup Sun
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Stephen K Mendenhall
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Anthony J Perugini
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rohit U Nagaraj
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Hanisha L Battina
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rachel J Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Kevin A Maupin
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Paul J Childress
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA.
| |
Collapse
|
33
|
Kim J, Lee G, Chang WS, Ki SH, Park JC. Comparison and Contrast of Bone and Dentin in Genetic Disorder, Morphology and Regeneration: A Review. J Bone Metab 2021; 28:1-10. [PMID: 33730779 PMCID: PMC7973397 DOI: 10.11005/jbm.2021.28.1.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/18/2020] [Indexed: 01/08/2023] Open
Abstract
The bone and dentin have distinct healing processes. The healing process of bones is regenerative, as newly formed tissues are morphologically and functionally similar to the original bone structures. In contrast, the healing process of dentin is reparative due to its failure to replicate some of its key morphological features. In this review, we compare and contrast the healing processes of bone and dentin. We describe how distinct morphological and physiological structures of the 2 tissues translate into different signaling molecules, growth factors, and matrix protein secretion.
Collapse
Affiliation(s)
- Jaehyun Kim
- College of Dental Medicine, Columbia University, New York, USA
| | - Gayeong Lee
- College of Dental Medicine, Columbia University, New York, USA
| | - Woo Sung Chang
- College of Dental Medicine, Columbia University, New York, USA
| | - Si Hyoung Ki
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Joo-Cheol Park
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| |
Collapse
|
34
|
Tanner MC, Boxriker S, Haubruck P, Child C, Westhauser F, Fischer C, Schmidmaier G, Moghaddam A. Expression of VEGF in Peripheral Serum Is a Possible Prognostic Factor in Bone-Regeneration via Masquelet-Technique-A Pilot Study. J Clin Med 2021; 10:jcm10040776. [PMID: 33672081 PMCID: PMC7919640 DOI: 10.3390/jcm10040776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 11/26/2022] Open
Abstract
Two-step Masquelet-technique established a new procedure in the treatment of osseous defects, addressing prerequisites postulated by the “diamond concept”. Increase in blood perfusion and growth factors are enhanced by the “Masquelet-membrane”. To describe this, we measured serum levels of Vascular Endothelial Growth Factor (VEGF) of patients with atrophic non-unions of long bones undergoing Masquelet-technique. From over 500 non-union patients undergoing Masquelet-technique with prospective follow-up we randomly selected 30 patients. 23 were included, 7 lost to follow-up or excluded because of incomplete data. Serum was drawn at specified intervals before and after surgery. Patients were followed for at least 6 months after step 2. Classification into both groups was performed according to radiological results and clinical outcome 6 months after step 2. Concentration of VEGF in patients’ serum was performed via ELISA. 14 achieved osseous consolidation (responder group), 9 cases did not (non-responder). Responders showed a significant increase of serum-VEGF in the first and second week when compared to the preoperative values of step 1. Non-responders showed a significant increase of VEGF in the second week after Steps 1 and 2. Comparison of groups showed significantly higher increase of serum-VEGF week2 after step 1 and preoperative to step 2 for responders. Results show one possibility of illustrating therapeutic progress by monitoring growth factors and possibly allowing prognostic conclusions thereof. This might lead to a more targeted treatment protocol.
Collapse
Affiliation(s)
- Michael C. Tanner
- Center for Orthopedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, 69118 Heidelberg, Germany; (P.H.); (C.C.); (F.W.); (C.F.); (G.S.)
- Correspondence: ; Tel.: +49-6221-562-6398
| | - Sonja Boxriker
- Center of Orthopedics, Trauma & Sports medicine, Aschaffenburg-Alzenau Hospital, 63739 Aschaffenburg, Germany; (S.B.); (A.M.)
| | - Patrick Haubruck
- Center for Orthopedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, 69118 Heidelberg, Germany; (P.H.); (C.C.); (F.W.); (C.F.); (G.S.)
| | - Christopher Child
- Center for Orthopedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, 69118 Heidelberg, Germany; (P.H.); (C.C.); (F.W.); (C.F.); (G.S.)
| | - Fabian Westhauser
- Center for Orthopedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, 69118 Heidelberg, Germany; (P.H.); (C.C.); (F.W.); (C.F.); (G.S.)
| | - Christian Fischer
- Center for Orthopedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, 69118 Heidelberg, Germany; (P.H.); (C.C.); (F.W.); (C.F.); (G.S.)
| | - Gerhard Schmidmaier
- Center for Orthopedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, 69118 Heidelberg, Germany; (P.H.); (C.C.); (F.W.); (C.F.); (G.S.)
| | - Arash Moghaddam
- Center of Orthopedics, Trauma & Sports medicine, Aschaffenburg-Alzenau Hospital, 63739 Aschaffenburg, Germany; (S.B.); (A.M.)
| |
Collapse
|
35
|
Johnstone MR, Brady RD, Church JE, Orr D, McDonald SJ, Grills BL. The TrkB agonist, 7,8-dihydroxyflavone, impairs fracture healing in mice. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2021; 21:263-271. [PMID: 34059571 PMCID: PMC8185262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To study the effects of the selective TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF), on fracture healing in mice and on an osteoprogenitor cell line, Kusa4b10, in vitro. METHODS Mice received unilateral closed mid-shaft tibial fractures and treated for two weeks with vehicle or 5 mg/kg/day DHF and euthanised at 28 days post-fracture. Calluses were analysed by micro-computed tomography (μCT) and three-point bending biomechanical test. Kusa4b10 cells were cultured with 50nM of 7,8-DHF or vehicle for 3-, 7-, 14-days for RT-PCR, and 21 days for mineralization. RESULTS μCT found 7,8-DHF calluses had decreased tissue volume (p=0.042), mean polar moment of inertia (p = 0.004), and mean cross-sectional area (p=0.042) compared to controls. At 28 days biomechanical analyses showed 7,8-DHF treatment decreased peak force (p=0.011) and stiffness per unit area (p=0.012). 7,8-DHF treatment did not change Kusa4b10 gene expression of Runx2 and alkaline phosphatase at all time points, nor mineralization. CONCLUSIONS 7,8-DHF treatment had a negative impact on fracture healing at 28 days post-fracture via an unknown mechanism. 7,8-DHF may have had a central role in impairing fracture healing.
Collapse
Affiliation(s)
- Maddison R. Johnstone
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Australia
| | - Rhys D. Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Jarrod E. Church
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Australia
| | - David Orr
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Australia
| | - Stuart J. McDonald
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Australia,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Brian L. Grills
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Australia,Corresponding author: Brian L. Grills, Department of Physiology, Anatomy, and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Australia, 3086 E-mail:
| |
Collapse
|
36
|
|
37
|
Abbasi N, Lee RSB, Ivanovski S, Love RM, Hamlet S. In vivo bone regeneration assessment of offset and gradient melt electrowritten (MEW) PCL scaffolds. Biomater Res 2020; 24:17. [PMID: 33014414 PMCID: PMC7529514 DOI: 10.1186/s40824-020-00196-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/21/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Biomaterial-based bone tissue engineering represents a promising solution to overcome reduced residual bone volume. It has been previously demonstrated that gradient and offset architectures of three-dimensional melt electrowritten poly-caprolactone (PCL) scaffolds could successfully direct osteoblast cells differentiation toward an osteogenic lineage, resulting in mineralization. The aim of this study was therefore to evaluate the in vivo osteoconductive capacity of PCL scaffolds with these different architectures. METHODS Five different calcium phosphate (CaP) coated melt electrowritten PCL pore sized scaffolds: 250 μm and 500 μm, 500 μm with 50% fibre offset (offset.50.50), tri layer gradient 250-500-750 μm (grad.250top) and 750-500-250 μm (grad.750top) were implanted into rodent critical-sized calvarial defects. Empty defects were used as a control. After 4 and 8 weeks of healing, the new bone was assessed by micro-computed tomography and immunohistochemistry. RESULTS Significantly more newly formed bone was shown in the grad.250top scaffold 8 weeks post-implantation. Histological investigation also showed that soft tissue was replaced with newly formed bone and fully covered the grad.250top scaffold. While, the bone healing did not happen completely in the 250 μm, offset.50.50 scaffolds and blank calvaria defects following 8 weeks of implantation. Immunohistochemical analysis showed the expression of osteogenic markers was present in all scaffold groups at both time points. The mineralization marker Osteocalcin was detected with the highest intensity in the grad.250top and 500 μm scaffolds. Moreover, the expression of the endothelial markers showed that robust angiogenesis was involved in the repair process. CONCLUSIONS These results suggest that the gradient pore size structure provides superior conditions for bone regeneration.
Collapse
Affiliation(s)
- Naghmeh Abbasi
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| | - Ryan S. B. Lee
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- School of Dentistry, University of Queensland, Herston Campus, Herston, Queensland 4006 Australia
| | - Saso Ivanovski
- School of Dentistry, University of Queensland, Herston Campus, Herston, Queensland 4006 Australia
| | - Robert M. Love
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| | - Stephen Hamlet
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport, Queensland 4215 Australia
| |
Collapse
|
38
|
Zhang J, Shi H, Zhang N, Hu L, Jing W, Pan J. Interleukin-4-loaded hydrogel scaffold regulates macrophages polarization to promote bone mesenchymal stem cells osteogenic differentiation via TGF-β1/Smad pathway for repair of bone defect. Cell Prolif 2020; 53:e12907. [PMID: 32951298 PMCID: PMC7574882 DOI: 10.1111/cpr.12907] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Objective Tissue engineering is a promising strategy for repair of large bone defect. However, the immune system reactions to biological scaffold are increasingly being recognized as a crucial factor influencing regeneration efficacy. In this study, a bone‐bioactive hydrogel bead loaded with interleukin‐4 (IL‐4) was used to regulate macrophages polarization and accelerate bone regeneration. Methods IL‐4‐loaded calcium‐enriched gellan gum (Ca‐GG + IL‐4) hydrogel beads were synthesised. And the effect on cell behaviour was detected. Furthermore, the effect of the Ca‐GG + IL‐4 hydrogel bead on macrophage polarization and the effect of macrophage polarization on bone mesenchymal stem cells (BMSCs) apoptosis and osteogenic differentiation were evaluated in vitro and in vivo. Results BMSCs were able to survive in the hydrogel regardless of whether IL‐4 was incorporated. Immunofluorescence staining and qPCR results revealed that Ca‐GG + IL‐4 hydrogel bead could promote M2 macrophage polarization and increase transforming growth factor (TGF)‐β1 expression level, which activates the TGF‐β1/Smad signalling pathway in BMSCs and promotes osteogenic differentiation. Moreover, immunohistochemical analysis demonstrated Ca‐GG + IL‐4 hydrogel bead could promote M2 macrophage polarization and reduce cell apoptosis in vivo. In addition, micro‐CT and immunohistochemical analysis at 12 weeks post‐surgery showed that Ca‐GG + IL‐4 hydrogel bead could achieve superior bone defect repair efficacy in vivo. Conclusions The Ca‐GG + IL‐4 hydrogel bead effectively promoted bone defect regeneration via regulating macrophage polarization, reducing cell apoptosis and promoting BMSCs osteogenesis through TGF‐β1/Smad pathway. Therefore, it is a promising strategy for repair of bone defect.
Collapse
Affiliation(s)
- Jiankang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haitao Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Nian Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liru Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Godoy-Gallardo M, Portolés-Gil N, López-Periago AM, Domingo C, Hosta-Rigau L. Immobilization of BMP-2 and VEGF within Multilayered Polydopamine-Coated Scaffolds and the Resulting Osteogenic and Angiogenic Synergy of Co-Cultured Human Mesenchymal Stem Cells and Human Endothelial Progenitor Cells. Int J Mol Sci 2020; 21:E6418. [PMID: 32899269 PMCID: PMC7503899 DOI: 10.3390/ijms21176418] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/31/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
We have previously reported the fabrication of a polycaprolactone and hydroxyapatite composite scaffold incorporating growth factors to be used for bone regeneration. Two growth factors were incorporated employing a multilayered coating based on polydopamine (PDA). In particular, Bone morphogenetic protein-2 (BMP-2) was bound onto the inner PDA layer while vascular endothelial growth factor (VEGF) was immobilized onto the outer one. Herein, the in vitro release of both growth factors is evaluated. A fastest VEGF delivery followed by a slow and more sustained release of BMP-2 was demonstrated, thus fitting the needs for bone tissue engineering applications. Due to the relevance of the crosstalk between bone-promoting and vessel-forming cells during bone healing, the functionalized scaffolds are further assessed on a co-culture setup of human mesenchymal stem cells and human endothelial progenitor cells. Osteogenic and angiogenic gene expression analysis indicates a synergistic effect between the growth factor-loaded scaffolds and the co-culture conditions. Taken together, these results indicate that the developed scaffolds hold great potential as an efficient platform for bone-tissue applications.
Collapse
Affiliation(s)
- Maria Godoy-Gallardo
- Department of Health Technology, Centre for Nanomedicine and Theranostics, DTU Health Tech, Technical University of Denmark, Produktionstorvet, Building 423, 2800 Kgs. Lyngby, Denmark;
| | - Núria Portolés-Gil
- Materials Science Institute of Barcelona (ICMAB-CSIC), Campus de la UAB s/n, 08193 Bellaterra, Spain; (N.P.-G.); (A.M.L.-P.); (C.D.)
| | - Ana M. López-Periago
- Materials Science Institute of Barcelona (ICMAB-CSIC), Campus de la UAB s/n, 08193 Bellaterra, Spain; (N.P.-G.); (A.M.L.-P.); (C.D.)
| | - Concepción Domingo
- Materials Science Institute of Barcelona (ICMAB-CSIC), Campus de la UAB s/n, 08193 Bellaterra, Spain; (N.P.-G.); (A.M.L.-P.); (C.D.)
| | - Leticia Hosta-Rigau
- Department of Health Technology, Centre for Nanomedicine and Theranostics, DTU Health Tech, Technical University of Denmark, Produktionstorvet, Building 423, 2800 Kgs. Lyngby, Denmark;
| |
Collapse
|
40
|
Palomino-Durand C, Lopez M, Marchandise P, Martel B, Blanchemain N, Chai F. Chitosan/Polycyclodextrin (CHT/PCD)-Based Sponges Delivering VEGF to Enhance Angiogenesis for Bone Regeneration. Pharmaceutics 2020; 12:pharmaceutics12090784. [PMID: 32825081 PMCID: PMC7557476 DOI: 10.3390/pharmaceutics12090784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Vascularization is one of the main challenges in bone tissue engineering (BTE). In this study, vascular endothelial growth factor (VEGF), known for its angiogenic effect, was delivered by our developed sponge, derived from a polyelectrolyte complexes hydrogel between chitosan (CHT) and anionic cyclodextrin polymer (PCD). This sponge, as a scaffold for growth factor delivery, was formed by freeze-drying a homogeneous CHT/PCD hydrogel, and thereafter stabilized by a thermal treatment. Microstructure, water-uptake, biodegradation, mechanical properties, and cytocompatibility of sponges were assessed. VEGF-delivery following incubation in medium was then evaluated by monitoring the VEGF-release profile and its bioactivity. CHT/PCD sponge showed a porous (open porosity of 87.5%) interconnected microstructure with pores of different sizes (an average pore size of 153 μm), a slow biodegradation (12% till 21 days), a high water-uptake capacity (~600% in 2 h), an elastic property under compression (elastic modulus of compression 256 ± 4 kPa), and a good cytocompatibility in contact with osteoblast and endothelial cells. The kinetic release of VEGF was found to exert a pro-proliferation and a pro-migration effect on endothelial cells, which are two important processes during scaffold vascularization. Hence, CHT/PCD sponges were promising vehicles for the delivery of growth factors in BTE.
Collapse
Affiliation(s)
- Carla Palomino-Durand
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Marco Lopez
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Pierre Marchandise
- ULR 4490–MABLab–Adiposité Médullaire et Os, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France;
- ULR 4490–MABLab–Adiposité Médullaire et Os, Univ. Littoral Côte d’Opale, 62200 Boulogne-sur-Mer, France
| | - Bernard Martel
- UMR 8207, UMET—Unité Matériaux et Transformations, Centre National de la Recherche Scientifique (CNRS), Institut National de la Recherche Agronomique (INRA), Ecole Nationale Supérieure de Chimie de Lille (ENSCL), University of Lille, 59655 Lille, France;
| | - Nicolas Blanchemain
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Feng Chai
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
- Correspondence: ; Tel.: +33-320-626-997
| |
Collapse
|
41
|
Ding MH, Lozoya EG, Rico RN, Chew SA. The Role of Angiogenesis-Inducing microRNAs in Vascular Tissue Engineering. Tissue Eng Part A 2020; 26:1283-1302. [PMID: 32762306 DOI: 10.1089/ten.tea.2020.0170] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is an important process in tissue repair and regeneration as blood vessels are integral to supply nutrients to a functioning tissue. In this review, the application of microRNAs (miRNAs) or anti-miRNAs that can induce angiogenesis to aid in blood vessel formation for vascular tissue engineering in ischemic diseases such as peripheral arterial disease and stroke, cardiac diseases, and skin and bone tissue engineering is discussed. Endothelial cells (ECs) form the endothelium of the blood vessel and are recognized as the primary cell type that drives angiogenesis and studied in the applications that were reviewed. Besides ECs, mesenchymal stem cells can also play a pivotal role in these applications, specifically, by secreting growth factors or cytokines for paracrine signaling and/or as constituent cells in the new blood vessel formed. In addition to delivering miRNAs or cells transfected/transduced with miRNAs for angiogenesis and vascular tissue engineering, the utilization of extracellular vesicles (EVs), such as exosomes, microvesicles, and EVs collectively, has been more recently explored. Proangiogenic miRNAs and anti-miRNAs contribute to angiogenesis by targeting the 3'-untranslated region of targets to upregulate proangiogenic factors such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor, and hypoxia-inducible factor-1 and increase the transduction of VEGF signaling through the PI3K/AKT and Ras/Raf/MEK/ERK signaling pathways such as phosphatase and tensin homolog or regulating the signaling of other pathways important for angiogenesis such as the Notch signaling pathway and the pathway to produce nitric oxide. In conclusion, angiogenesis-inducing miRNAs and anti-miRNAs are promising tools for vascular tissue engineering for several applications; however, future work should emphasize optimizing the delivery and usage of these therapies as miRNAs can also be associated with the negative implications of cancer.
Collapse
Affiliation(s)
- May-Hui Ding
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eloy G Lozoya
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Rene N Rico
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Sue Anne Chew
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
42
|
Zhang J, Pan J, Jing W. Motivating role of type H vessels in bone regeneration. Cell Prolif 2020; 53:e12874. [PMID: 33448495 PMCID: PMC7507571 DOI: 10.1111/cpr.12874] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/03/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
Coupling between angiogenesis and osteogenesis has an important role in both normal bone injury repair and successful application of tissue‐engineered bone for bone defect repair. Type H blood vessels are specialized microvascular components that are closely related to the speed of bone healing. Interactions between type H endothelial cells and osteoblasts, and high expression of CD31 and EMCN render the environment surrounding these blood vessels rich in factors conducive to osteogenesis and promote the coupling of angiogenesis and osteogenesis. Type H vessels are mainly distributed in the metaphysis of bone and densely surrounded by Runx2+ and Osterix+ osteoprogenitors. Several other factors, including hypoxia‐inducible factor‐1α, Notch, platelet‐derived growth factor type BB, and slit guidance ligand 3 are involved in the coupling of type H vessel formation and osteogenesis. In this review, we summarize the identification and distribution of type H vessels and describe the mechanism for type H vessel‐mediated modulation of osteogenesis. Type H vessels provide new insights for detection of the molecular and cellular mechanisms that underlie the crosstalk between angiogenesis and osteogenesis. As a result, more feasible therapeutic approaches for treatment of bone defects by targeting type H vessels may be applied in the future.
Collapse
Affiliation(s)
- Jiankang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Dashtimoghadam E, Fahimipour F, Tongas N, Tayebi L. Microfluidic fabrication of microcarriers with sequential delivery of VEGF and BMP-2 for bone regeneration. Sci Rep 2020; 10:11764. [PMID: 32678204 PMCID: PMC7366644 DOI: 10.1038/s41598-020-68221-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Wound instability and poor functional vascularization in bone tissue engineering lead to lack of tissue integration and ultimate failure of engineered grafts. In order to harness the regenerative potential of growth factors and stimulate bone healing, present study aims to design multifunctional cell therapy microcarriers with the capability of sequential delivery of essential growth factors, bone morphogenetic protein 2 (BMP-2) and vascular endothelial growth factor (VEGF). An on-chip double emulsion method was implemented to generate monodisperse VEGF encapsulated microcarriers. Bio-inspired poly(3,4-dihydroxyphenethylamine) (PDA) was then functionalized to the microcarriers surface for BMP-2 conjugation. The microcarriers were seeded with mesenchymal stem cells (MSCs) using a dynamic culture technique for cells expansion. Finally, the microcarriers were incorporated into an injectable alginate-RGD hydrogel laden with endothelial cells (ECs) for further analysis. The DNA and calcium content, as well as ALP activity of the construct were analyzed. The confocal fluorescent microscopy was employed to monitor the MSCs and tunneling structure of ECs. Eventually, the capability of developed microcarriers for bone tissue formation was examined in vivo. Microfluidic platform generated monodisperse VEGF-loaded PLGA microcarriers with size-dependent release patterns. Microcarriers generated with the on-chip technique showed more sustained VEGF release profiles compared to the conventional bulk mixing method. The PDA functionalization of microcarriers surface not only provided immobilization of BMP-2 with prolonged bioavailability, but also enhanced the attachment and proliferation of MSCs. Dynamic culturing of microcarriers showcased their great potential to boost MSCs population required for stem cell therapy of bone defects. ALP activity and calcium content analysis of MSCs-laden microcarriers loaded into injectable hydrogels revealed their capability of tunneling formation, vascular cell growth and osteogenic differentiation. The in vivo histology and real-time polymerase chain reaction analysis revealed that transplantation of MSC-laden microcarriers supports ectopic bone formation in the rat model. The presented approach to design bioactive microcarriers offer sustained sequential delivery of bone ECM chemical cues and offer an ideal stabilized 3D microenvironment for patient-specific cell therapy applications. The proposed methodology is readily expandable to integrate other cells and cytokines in a tuned spatiotemporal manner for personalized regenerative medicine.
Collapse
Affiliation(s)
| | - Farahnaz Fahimipour
- Marquette University School of Dentistry, Milwaukee, WI, USA
- Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nikita Tongas
- Marquette University School of Dentistry, Milwaukee, WI, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA.
| |
Collapse
|
44
|
Arjmand B, Sarvari M, Alavi-Moghadam S, Payab M, Goodarzi P, Gilany K, Mehrdad N, Larijani B. Prospect of Stem Cell Therapy and Regenerative Medicine in Osteoporosis. Front Endocrinol (Lausanne) 2020; 11:430. [PMID: 32719657 PMCID: PMC7347755 DOI: 10.3389/fendo.2020.00430] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
The field of cell therapy and regenerative medicine can hold the promise of restoring normal tissues structure and function. Additionally, the main targets of stem cell-based therapies are chronic diseases and lifelong disabilities without definite cures such as osteoporosis. Osteoporosis as one of the important causes of morbidity in older men and post-menopausal women is characterized by reduced bone quantity or skeletal tissue atrophy that leads to an increased risk of osteoporotic fractures. The common therapeutic methods for osteoporosis only can prevent the loss of bone mass and recover the bone partially. Nevertheless, stem cell-based therapy is considered as a new approach to regenerate the bone tissue. Herein, mesenchymal stem cells as pivotal candidates for regenerative medicine purposes especially bone regeneration are the most common type of cells with anti-inflammatory, immune-privileged potential, and less ethical concerns than other types of stem cells which are investigated in osteoporosis. Based on several findings, the mesenchymal stem cells effectiveness near to a great extent depends on their secretory function. Indeed, they can be involved in the establishment of normal bone remodeling via initiation of specific molecular signaling pathways. Accordingly, the aim herein was to review the effects of stem cell-based therapies in osteoporosis.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sarvari
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Gilany
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACER), Tehran, Iran
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACER), Tehran, Iran
| | - Neda Mehrdad
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Wang S, Qiu J, Guo A, Ren R, He W, Liu S, Liu Y. Nanoscale perfluorocarbon expediates bone fracture healing through selectively activating osteoblastic differentiation and functions. J Nanobiotechnology 2020; 18:84. [PMID: 32493334 PMCID: PMC7271395 DOI: 10.1186/s12951-020-00641-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND RATIONALE Fracture incidence increases with ageing and other contingencies. However, the strategy of accelerating fracture repair in clinical therapeutics remain a huge challenge due to its complexity and a long-lasting period. The emergence of nano-based drug delivery systems provides a highly efficient, targeted and controllable drug release at the diseased site. Thus far, fairly limited studies have been carried out using nanomedicines for the bone repair applications. Perfluorocarbon (PFC), FDA-approved clinical drug, is received increasing attention in nanomedicine due to its favorable chemical and biologic inertness, great biocompatibility, high oxygen affinity and serum-resistant capability. In the premise, the purpose of the current study is to prepare nano-sized PFC materials and to evaluate their advisable effects on promoting bone fracture repair. RESULTS Our data unveiled that nano-PFC significantly enhanced the fracture repair in the rabbit model with radial fractures, as evidenced by increased soft callus formation, collagen synthesis and accumulation of beneficial cytokines (e.g., vascular endothelial growth factor (VEGF), matrix metalloprotein 9 (MMP-9) and osteocalcin). Mechanistic studies unraveled that nano-PFC functioned to target osteoblasts by stimulating their differentiation and activities in bone formation, leading to accelerated bone remodeling in the fractured zones. Otherwise, osteoclasts were not affected upon nano-PFC treatment, ruling out the potential target of nano-PFC on osteoclasts and their progenitors. CONCLUSIONS These results suggest that nano-PFC provides a potential perspective for selectively targeting osteoblast cell and facilitating callus generation. This study opens up a new avenue for nano-PFC as a promising agent in therapeutics to shorten healing time in treating bone fracture.
Collapse
Affiliation(s)
- Shunhao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiahuang Qiu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Anyi Guo
- Beijing Jishuitan Hospital, The 4th Clinical Hospital of Peking University Health Science Center, No. 31 East Street, Xinjiekou, Xicheng District, Beijing, 100035, China
| | - Ruanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei He
- Beijing Jishuitan Hospital, The 4th Clinical Hospital of Peking University Health Science Center, No. 31 East Street, Xinjiekou, Xicheng District, Beijing, 100035, China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yajun Liu
- Beijing Jishuitan Hospital, The 4th Clinical Hospital of Peking University Health Science Center, No. 31 East Street, Xinjiekou, Xicheng District, Beijing, 100035, China.
| |
Collapse
|
46
|
Gu RD, Xiao F, Wang L, Sun KJ, Chen LL. Biocompatibility of polyetheretherketone for the treatment of orbital bone defects. Int J Ophthalmol 2020; 13:725-730. [PMID: 32420218 DOI: 10.18240/ijo.2020.05.05] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/19/2020] [Indexed: 12/12/2022] Open
Abstract
AIM To investigate the biocompatibility and therapeutic effects of polyetheretherketone (PEEK) on recovery of a rabbit orbital defect. METHODS Totally 16 New Zealand rabbits were used to establish an orbital bone defect model and then randomly divided into two groups. PEEK was implanted in the experimental group. The control group was blank, and no substance was implanted. The model rabbits were sacrificed at 4 and 8wk, and examined by general observations, histology, electron microscopy, Western blotting, and real-time polymerase chain reaction. RESULTS No infection or rejection occurred after PEEK implantation, and biocompatibility was good. The relative expression of vascular endothelial growth factor (VEGF) protein in the experimental group was significantly higher than that in the control group postoperatively (P<0.05). Bone defect repair in the experimental group was significantly better than that in the control group in the same period and some osteogenesis was observed. CONCLUSION PEEK has good biocompatibility and efficacy for the treatment of orbital bone defects in a rabbit model.
Collapse
Affiliation(s)
- Rui-Dong Gu
- Department of Ophthalmology, the Fourth People's Hospital of Shenyang, Shenyang 110031, Liaoning Province, China
| | - Fan Xiao
- Department of Ophthalmology, the Fourth People's Hospital of Shenyang, Shenyang 110031, Liaoning Province, China
| | - Lin Wang
- Department of Ophthalmology, the Fourth People's Hospital of Shenyang, Shenyang 110031, Liaoning Province, China
| | - Kai-Jian Sun
- Department of Ophthalmology, the Fourth People's Hospital of Shenyang, Shenyang 110031, Liaoning Province, China
| | - Lin-Lin Chen
- Department of Ophthalmology, the Fourth People's Hospital of Shenyang, Shenyang 110031, Liaoning Province, China
| |
Collapse
|
47
|
Suyasa IK, Wiradewi Lestari AA. Low expression of vascular endothelial growth factor and high serum level of cyclic guanine monophosphate as the risk factors of femoral head osteonecrosis in alcohol-exposed Wistar rat. Chin J Traumatol 2020; 23:107-112. [PMID: 31980236 PMCID: PMC7156883 DOI: 10.1016/j.cjtee.2019.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Severe damage to the femoral head in patients with osteonecrosis has a high impact on morbidity. Despite early diagnosis, the treatment outcome is still unsatisfactory. This study aimed to explore the expression of vascular endothelial growth factor (VEGF) and cyclic guanine monophosphate (cGMP) serum level as the risk factors of femoral head osteonecrosis in alcohol-exposed Wistar rats. METHODS This was an experimental study using randomized post-test only control group design, with samples using 10-14 weeks Wistar male rats. Rats were then divided into 6 groups: 3 groups without intervention, and 3 groups with intervention using 40% alcohol given perorally. Each one group from intervention and control group was euthanized by the end of the week for 3 consecutive weeks. Proximal femurs were examined under microscope for osteonecrosis, immunohistochemically for VEGF, and blood serum for cGMP levels. RESULTS VEGF expression in the femoral head of alcohol-exposed Wistar rats was lower than those not exposed to alcohol (p < 0.005). Blood serum cGMP levels of alcohol-exposed Wistar rats were higher than those not exposed to alcohol (p < 0.005). The number of necrotic osteocytes in the femoral head of Wistar rats exposed to alcohol was greater than those not exposed to alcohol (p < 0.005). There are significant differences between VEGF, cGMP levels, and number of necrotic osteocytes in the control group and treatment at 1st, 2nd, and 3rd week (p < 0.005). CONCLUSIONS Based on the result of this study, VEGF and cGMP may be considered as diagnostic biomarkers for alcohol-induced femoral head osteonecrosis.
Collapse
Affiliation(s)
- I Ketut Suyasa
- Department of Orthopedic and Traumatology, Faculty of Medicine, Udayana University, Bali, Indonesia
| | - Anak Agung Wiradewi Lestari
- Department of Clinical Pathology, Faculty of Medicine, Udayana University, Bali, Indonesia,Corresponding author.
| |
Collapse
|
48
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part II - Modulation of angiogenesis. Clin Hemorheol Microcirc 2020; 73:409-438. [PMID: 31177206 DOI: 10.3233/ch-199103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The treatment of critical-size bone defects following complicated fractures, infections or tumor resections is a major challenge. The same applies to fractures in patients with impaired bone healing due to systemic inflammatory and metabolic diseases. Despite considerable progress in development and establishment of new surgical techniques, design of bone graft substitutes and imaging techniques, these scenarios still represent unresolved clinical problems. However, the development of new active substances offers novel potential solutions for these issues. This work discusses therapeutic approaches that influence angiogenesis or hypoxic situations in healing bone and surrounding tissue. In particular, literature on sphingosine-1-phosphate receptor modulators and nitric oxide (NO•) donors, including bi-functional (hybrid) compounds like NO•-releasing cyclooxygenase-2 inhibitors, was critically reviewed with regard to their local and systemic mode of action.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
49
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part III - Further strategies for local and systemic modulation. Clin Hemorheol Microcirc 2020; 73:439-488. [PMID: 31177207 DOI: 10.3233/ch-199104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this third in a series of reviews on adjuvant drug-assisted bone healing, further approaches aiming at influencing the healing process are discussed. Local and systemic modulation of bone metabolism is pursued with use of a number of drugs with completely different indications, which are characterized by a pleiotropic spectrum of action. These include drugs used to treat lipid disorders (HMG-CoA reductase inhibitors), hypertension (ACE inhibitors), osteoporosis (bisphosphonates), cancer (proteasome inhibitors) and others. Potential applications to enhance bone healing are discussed.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
50
|
Anti-Vascular Endothelial Growth Factors as a Potential Risk for Implant Failure: A Clinical Report. Case Rep Med 2020; 2020:6141493. [PMID: 32089703 PMCID: PMC7021462 DOI: 10.1155/2020/6141493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 10/29/2019] [Accepted: 01/14/2020] [Indexed: 11/18/2022] Open
Abstract
Knowledge of the risk factors for implant osseointegration is essential for clinical decision-making and optimizing treatment success. This clinical report presents a rare case of implant failure in a patient who received intravitreal injections of a vascular endothelial growth factor (VEGF) inhibitor for the treatment of age-related macular degeneration. Following CARE guidelines, the report presents a case rehabilitated with a mandibular 2-implant overdenture using the immediate-loading protocol and standard procedures. The implants failed within six weeks of immediate loading although primary stability (≥50 Ncm) was achieved during surgery and clinical follow-ups did not show any deviance from standard implant care or patient-related complications. Further investigation suggested that the intake of a VEGF inhibitor may be the cause of failure. This clinical report highlights the importance of systemic risk factors in implant success and their consideration during planning for implant-assisted treatment.
Collapse
|