1
|
Ye P, Gu R, Zhu H, Chen J, Han F, Nie X. SOX family transcription factors as therapeutic targets in wound healing: A comprehensive review. Int J Biol Macromol 2023; 253:127243. [PMID: 37806414 DOI: 10.1016/j.ijbiomac.2023.127243] [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: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
The SOX family plays a vital role in determining the fate of cells and has garnered attention in the fields of cancer research and regenerative medicine. It also shows promise in the study of wound healing, as it actively participates in the healing processes of various tissues such as skin, fractures, tendons, and the cornea. However, our understanding of the mechanisms behind the SOX family's involvement in wound healing is limited compared to its role in cancer. Gaining insight into its role, distribution, interaction with other factors, and modifications in traumatized tissues could provide valuable new knowledge about wound healing. Based on current research, SOX2, SOX7, and SOX9 are the most promising members of the SOX family for future interventions in wound healing. SOX2 and SOX9 promote the renewal of cells, while SOX7 enhances the microvascular environment. The SOX family holds significant potential for advancing wound healing research. This article provides a comprehensive review of the latest research advancements and therapeutic tools related to the SOX family in wound healing, as well as the potential benefits and challenges of targeting the SOX family for wound treatment.
Collapse
Affiliation(s)
- Penghui Ye
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Rifang Gu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China
| | - Huan Zhu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Jitao Chen
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
2
|
Nadine S, Fernandes IJ, Correia CR, Mano JF. Close-to-native bone repair via tissue-engineered endochondral ossification approaches. iScience 2022; 25:105370. [PMID: 36339269 PMCID: PMC9626746 DOI: 10.1016/j.isci.2022.105370] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In order to solve the clinical challenges related to bone grafting, several tissue engineering (TE) strategies have been proposed to repair critical-sized defects. Generally, the classical TE approaches are designed to promote bone repair via intramembranous ossification. Although promising, strategies that direct the osteogenic differentiation of mesenchymal stem/stromal cells are usually characterized by a lack of functional vascular supply, often resulting in necrotic cores. A less explored alternative is engineering bone constructs through a cartilage-mediated approach, resembling the embryological process of endochondral ossification. The remodeling of an intermediary hypertrophic cartilaginous template triggers vascular invasion and bone tissue deposition. Thus, employing this knowledge can be a promising direction for the next generation of bone TE constructs. This review highlights the most recent biomimetic strategies for applying endochondral ossification in bone TE while discussing the plethora of cell types, culture conditions, and biomaterials essential to promote a successful bone regeneration process.
Collapse
|
3
|
Ko FC, Moran MM, Ross RD, Sumner DR. Activation of canonical Wnt signaling accelerates intramembranous bone regeneration in male mice. J Orthop Res 2022; 40:1834-1843. [PMID: 34811780 PMCID: PMC9124233 DOI: 10.1002/jor.25217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 02/04/2023]
Abstract
Canonical Wnt signaling plays an important role in skeletal development, homeostasis, and both endochondral and intramembranous repair. While studies have demonstrated that the inhibition of Wnt signaling impairs intramembranous bone regeneration, how its activation affects intramembranous bone regeneration has been underexplored. Therefore, we sought to determine the effects of activation of canonical Wnt signaling on intramembranous bone regeneration by using the well-established marrow ablation model. We hypothesized that mice with a mutation in the Wnt ligand coreceptor gene Lrp5 would have accelerated intramembranous bone regeneration. Male and female wild-type and Lrp5-mutant mice underwent unilateral femoral bone marrow ablation surgery in the right femur at 4 weeks of age. Both the left intact and right operated femurs were assessed at Days 3, 5, 7, 10, and 14. The intact femur of Lrp5 mutant mice of both sexes had higher bone mass than wild-type littermates, although to a greater degree in males than females. Overall, the regenerated bone volume in Lrp5 mutant male mice was 1.8-fold higher than that of littermate controls, whereas no changes were observed between female Lrp5 mutant and littermate control mice. In addition, the rate of intramembranous bone regeneration (from Day 3 to Day 7) was higher in Lrp5 mutant male mice compared to their same-sex littermate controls with no difference in the females. Thus, activation of canonical Wnt signaling increases bone mass in intact bones of both sexes, but accelerates intramembranous bone regeneration following an injury challenge only in male mice.
Collapse
Affiliation(s)
- Frank C. Ko
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| | - Meghan M. Moran
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| | - Ryan D. Ross
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| | - D. Rick Sumner
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| |
Collapse
|
4
|
Steppe L, Krüger BT, Tschaffon MEA, Fischer V, Tuckermann J, Ignatius A, Haffner-Luntzer M. Estrogen Receptor α Signaling in Osteoblasts is Required for Mechanotransduction in Bone Fracture Healing. Front Bioeng Biotechnol 2021; 9:782355. [PMID: 34950644 PMCID: PMC8689144 DOI: 10.3389/fbioe.2021.782355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Biomechanical stimulation by whole-body low-magnitude high-frequency vibration (LMHFV) has demonstrated to provoke anabolic effects on bone metabolism in both non-osteoporotic and osteoporotic animals and humans. However, preclinical studies reported that vibration improved fracture healing and bone formation in osteoporotic, ovariectomized (OVX) mice representing an estrogen-deficient hormonal status, but impaired bone regeneration in skeletally healthy non-OVX mice. These effects were abolished in general estrogen receptor α (ERα)-knockout (KO) mice. However, it remains to be elucidated which cell types in the fracture callus are targeted by LMHFV during bone healing. To answer this question, we generated osteoblast lineage-specific ERα-KO mice that were subjected to ovariectomy, femur osteotomy and subsequent vibration. We found that the ERα specifically on osteoblastic lineage cells facilitated the vibration-induced effects on fracture healing, because in osteoblast lineage-specific ERα-KO (ERαfl/fl; Runx2Cre) mice the negative effects in non-OVX mice were abolished, whereas the positive effects of vibration in OVX mice were reversed. To gain greater mechanistic insights, the influence of vibration on murine and human osteogenic cells was investigated in vitro by whole genome array analysis and qPCR. The results suggested that particularly canonical WNT and Cox2/PGE2 signaling is involved in the mechanotransduction of LMHFV under estrogen-deficient conditions. In conclusion, our study demonstrates a critical role of the osteoblast lineage-specific ERα in LMHFV-induced effects on fracture healing and provides further insights into the molecular mechanism behind these effects.
Collapse
Affiliation(s)
- Lena Steppe
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Benjamin Thilo Krüger
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | | | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
5
|
Haffner-Luntzer M, Fischer V, Ignatius A. Differences in Fracture Healing Between Female and Male C57BL/6J Mice. Front Physiol 2021; 12:712494. [PMID: 34434120 PMCID: PMC8381649 DOI: 10.3389/fphys.2021.712494] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Background Mice are increasingly used in fracture healing research because of the opportunity to use transgenic animals. While both, male and female mice are employed, there is no consensus in the literature whether fracture healing differs between both sexes. Therefore, the aim of the present study was to analyze diaphyseal fracture healing in female and male C57BL/6J mice, a commonly used mouse strain in bone research. Methods For that purpose, 12-week-old Female (17–20 g) and Male mice (22–26 g) received a standardized femur midshaft osteotomy stabilized by an external fixator. Mice were euthanized 10 and 21 days after fracture and bone healing was analyzed by biomechanical testing, μCT, histology, immunohistochemistry and qPCR. Results Ten days after fracture, Male mice displayed significantly more cartilage but less fibrous tissue in the fracture callus compared to Female mice, whereas the amount of bone did not differ. At day 21, Male mice showed a significantly larger fracture callus compared to Female mice. The relative amount of bone in the fracture callus did not significantly differ between both sexes, whereas its tissue mineral density was significantly higher in Male mice on day 21, indicating more mature bone and slightly more rapid fracture healing. These results were confirmed by a significantly greater absolute bending stiffness of the fractured femurs of Male mice on day 21. On the molecular level, Male mice displayed increased active β-catenin expression in the fracture callus, whereas estrogen receptor α (ERα) expression was lower. Conclusion These results suggest that Male mice display more rapid fracture healing with more prominent cartilaginous callus formation. This might be due to the higher weight of Male mice, resulting in increased mechanical loading of the fracture. Furthermore, Male mice displayed significantly greater activation of osteoanabolic Wnt/β-catenin signaling, which might also contribute to more rapid bone regeneration.
Collapse
Affiliation(s)
- Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Centre Ulm, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Centre Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Centre Ulm, Ulm, Germany
| |
Collapse
|
6
|
Li S, Zhou H, Hu C, Yang J, Ye J, Zhou Y, Li Z, Chen L, Zhou Q. Total Flavonoids of Rhizoma Drynariae Promotes Differentiation of Osteoblasts and Growth of Bone Graft in Induced Membrane Partly by Activating Wnt/β-Catenin Signaling Pathway. Front Pharmacol 2021; 12:675470. [PMID: 34122101 PMCID: PMC8188237 DOI: 10.3389/fphar.2021.675470] [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: 03/03/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Total flavonoids of Rhizoma drynariae (TFRD), a Chinese medicine, is widely used in the treatment of fracture, bone defect, osteoporosis and other orthopedic diseases, and has achieved good effects. Purpose of this trial was to explore efficacy of TFRD on bone graft’s mineralization and osteoblasts’ differentiation in Masquelet induced membrane technique in rats. Forty male Sprague-Dawley rats were randomly divided into high dose group (H-TFRD), middle dose group (M-TFRD), low dose group (L-TFRD) and control group (control). The critical size bone defect model of rats was established with 10 rats in each group. Polymethyl methacrylate (PMMA) spacer was implanted into the defect of right femur in rats. After the formation of the induced membrane, autogenous bone was implanted into the induced membrane. After 12 weeks of bone graft, bone tissues in the area of bone graft were examined by X-ray, Micro-CT, hematoxylin-eosin (HE) and Masson trichrome staining to evaluate the growth of the bone graft. The β-catenin, c-myc, COL1A1, BMP-2 and OPN in bone graft were quantitatively analyzed by Western blot and Immunohistostaining. Osteoblasts were cultured in the medium containing TFRD. Cell Counting Kit-8 (CCK-8) method, Alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining, Western blot, RT-PCR and other methods were used to detect the effects of TFRD on the proliferation of osteoblasts and the regulation of Wnt/β-catenin signaling pathway. In vivo experiments showed that the growth and mineralization of bone graft in TFRD group was better. Moreover, the expression of Wnt/β-catenin and osteogenesis-related proteins in bone tissue of TFRD group was more than that in other groups. In vitro experiments indicated that osteoblasts proliferated faster, activity of ALP was higher, number of mineralized nodules and proteins related to osteogenesis were more in TFRD group. But blocking Wnt/β-catenin signaling pathway could limit these effects. Therefore, TFRD could promote mineralization of bone graft and differentiation of osteoblasts in a dose-dependent manner during growing period of the bone graft of induced membrane technique, which is partly related to the activation of Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Shuyuan Li
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongliang Zhou
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cheng Hu
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiabao Yang
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinfei Ye
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuexi Zhou
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zige Li
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Leilei Chen
- Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qishi Zhou
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
7
|
Huang Y, Xu Y, Feng S, He P, Sheng B, Ni J. miR-19b enhances osteogenic differentiation of mesenchymal stem cells and promotes fracture healing through the WWP1/Smurf2-mediated KLF5/β-catenin signaling pathway. Exp Mol Med 2021; 53:973-985. [PMID: 34035464 PMCID: PMC8178348 DOI: 10.1038/s12276-021-00631-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 01/08/2023] Open
Abstract
Bone marrow mesenchymal stem cell (BMSC)-derived exosomes have been found to enhance fracture healing. In addition, microRNAs contributing to the healing of various bone fractures have attracted widespread attention in recent years, but knowledge of the mechanisms by which they act is still very limited. In this study, we clarified the function of altered microRNA-19b (miR-19b) expression in BMSCs in fracture healing. We modulated miR-19b expression via mimics/inhibitors in BMSCs and via agomirs in mice to explore the effects of these changes on osteogenic factors, bone cell mineralization and the healing status of modeled fractures. Through gain- and loss-of function assays, the binding affinity between miR-19b and WWP1/Smurf2 was identified and characterized to explain the underlying mechanism involving the KLF5/β-catenin signaling pathway. miR-19b promoted the differentiation of human BMSCs into osteoblasts by targeting WWP1 and Smurf2. Overexpression of WWP1 or Smurf2 degraded the target protein KLF5 in BMSCs through ubiquitination to inhibit fracture healing. KLF5 knockdown delayed fracture healing by modulating the Wnt/β-catenin signaling pathway. Furthermore, miR-19b enhanced fracture healing via the KLF5/β-catenin signaling pathway by targeting WWP1 or Smurf2. Moreover, miR-19b was found to be enriched in BMSC-derived exosomes, and treatment with exosomes promoted fracture healing in vivo. Collectively, these results indicate that mesenchymal stem cell-derived exosomal miR-19b represses the expression of WWP1 or Smurf2 and elevates KLF5 expression through the Wnt/β-catenin signaling pathway, thereby facilitating fracture healing. Understanding how a small regulatory RNA molecule helps to promote fracture healing could lead to new treatments for broken bones. Working with human cells and mouse models, a team led by Yongqiang Xu from the Hunan Provincial People’s Hospital in Changsha, China, showed how microRNA-19b in extracellular vesicles secreted by bone marrow stem cells (BMSCs) contributes to the healing process. The researchers found that the microRNA blocks the function of two proteins that normally restrain the activity of a third protein needed for BMSCs to home in on the site of injury and turn into new bone tissue. In mice with leg bone fractures, injections of microRNA-19b–filled vesicles derived from BMSCs accelerated healing and recovery, suggesting that similar therapies might be helpful in human patients.
Collapse
Affiliation(s)
- Yan Huang
- Department of Orthopaedics, Hunan Provincial People's Hospital, Changsha, China.,Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yongqiang Xu
- Department of Orthopaedics, Hunan Provincial People's Hospital, Changsha, China.
| | - Siyin Feng
- Department of Orthopaedics, Hunan Provincial People's Hospital, Changsha, China
| | - Pan He
- Department of Orthopaedics, Hunan Provincial People's Hospital, Changsha, China
| | - Bing Sheng
- Department of Orthopaedics, Hunan Provincial People's Hospital, Changsha, China
| | - Jiangdong Ni
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
8
|
Gao J, Fan L, Zhao L, Su Y. The interaction of Notch and Wnt signaling pathways in vertebrate regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:11. [PMID: 33791915 PMCID: PMC8012441 DOI: 10.1186/s13619-020-00072-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022]
Abstract
Regeneration is an evolutionarily conserved process in animal kingdoms, however, the regenerative capacities differ from species and organ/tissues. Mammals possess very limited regenerative potential to replace damaged organs, whereas non-mammalian species usually have impressive abilities to regenerate organs. The regeneration process requires proper spatiotemporal regulation from key signaling pathways. The canonical Notch and Wnt signaling pathways, two fundamental signals guiding animal development, have been demonstrated to play significant roles in the regeneration of vertebrates. In recent years, increasing evidence has implicated the cross-talking between Notch and Wnt signals during organ regeneration. In this review, we summarize the roles of Notch signaling and Wnt signaling during several representative organ regenerative events, emphasizing the functions and molecular bases of their interplay in these processes, shedding light on utilizing these two signaling pathways to enhance regeneration in mammals and design legitimate therapeutic strategies.
Collapse
Affiliation(s)
- Junying Gao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China.,College of Fisheries, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Lixia Fan
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China.,College of Fisheries, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Long Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China. .,College of Fisheries, Ocean University of China, Qingdao, 266003, Shandong, China.
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China. .,College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, Shandong, China.
| |
Collapse
|
9
|
Schupbach D, Comeau-Gauthier M, Harvey E, Merle G. Wnt modulation in bone healing. Bone 2020; 138:115491. [PMID: 32569871 DOI: 10.1016/j.bone.2020.115491] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
Abstract
Genetic studies have been instrumental in the field of orthopaedics for finding tools to improve the standard management of fractures and delayed unions. The Wnt signaling pathway that is crucial for development and maintenance of many organs also has a very promising pathway for enhancement of bone regeneration. The Wnt pathway has been shown to have a direct effect on stem cells during bone regeneration, making Wnt a potential target to stimulate bone repair after trauma. A more complete view of how Wnt influences animal bone regeneration has slowly come to light. This review article provides an overview of studies done investigating the modulation of the canonical Wnt pathway in animal bone regeneration models. This not only includes a summary of the recent work done elucidating the roles of Wnt and β-catenin in fracture healing, but also the results of thirty transgenic studies, and thirty-eight pharmacological studies. Finally, we discuss the discontinuation of sclerostin clinical trials, ongoing clinical trials with lithium, the results of Dkk antibody clinical trials, the shift into combination therapies and the future opportunities to enhance bone repair and regeneration through the modulation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Drew Schupbach
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Marianne Comeau-Gauthier
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Edward Harvey
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada.
| | - Geraldine Merle
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Department of Chemical Engineering, Polytechnique Montreal, 2500, chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada.
| |
Collapse
|
10
|
Deng Z, Gao X, Sun X, Cui Y, Amra S, Huard J. Gender differences in tibial fractures healing in normal and muscular dystrophic mice. Am J Transl Res 2020; 12:2640-2651. [PMID: 32655796 PMCID: PMC7344076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/25/2020] [Indexed: 06/11/2023]
Abstract
Duchenne muscular dystrophy (DMD) patients have a high fracture risk and poor fracture healing. The dystrophin-/- (mdx) mouse is a murine model of DMD and exhibits delayed bone fracture healing. Since our research team has shown that adult stem cells, such as muscle-derived stem cells, display a gender difference in their osteogenic potential with the male cells being more osteogenic, we hypothesize that a potential gender differences may exist during bone healing in normal and mdx mice. To test this hypothesis, wild-type (WT) and mdx mice underwent tibial fracture surgery and microCT live scanning biweekly. The mice were sacrificed at 6 weeks post-surgery and the calluses were collected for histological analysis. To further investigate the mechanism, another two sets of mice were sacrificed at 10 days after fracture for RNA extraction and gene expression analysis and histology. MicroCT results showed, at 6 weeks post- surgery, the calluses were larger but showed less remodeling in both normal and mdx male mice when compared to females, at the same time point. However, females had higher callus bone volume density and an increase in osteoclast (OCs) number. At 10 days after fracture surgery, male mice had formed larger calluses, whereas females formed well-remodeled calluses with more osteoblasts and a greater bone area for both WT and mdx mice. Higher IGF-1 expression was observed in male mdx mice when compared to their female counterparts, whereas female WT mice had higher BMP-9 expression when compared to WT males. In conclusion, male mice formed larger bone calluses than females during tibial fracture healing for both WT and mdx mice. This may be attributed to higher IGF-1 expression, activation of Wnt/β-catennin signaling pathway and greater OB numbers during callus formation. Female mice achieved better bone remodeling in the regenerated bone with higher bone quality due to increased OC numbers that promote faster remodeling of the fracture calluses, and higher BMP-9 expression levels. Therefore, gender is one of many factors that need to be considered for both animal and human bone research.
Collapse
Affiliation(s)
- Zhenhan Deng
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX 77054, USA
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong, China
| | - Xueqin Gao
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX 77054, USA
- Center for Regenerative Sports Medicine, The Steadman Philippon Research InstituteVail, CO 81657, USA
| | - Xuying Sun
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX 77054, USA
| | - Yan Cui
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX 77054, USA
| | - Sarah Amra
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX 77054, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX 77054, USA
- Center for Regenerative Sports Medicine, The Steadman Philippon Research InstituteVail, CO 81657, USA
| |
Collapse
|
11
|
Zhang W, Zhao H, Chen K, Huang Y. Overexpressing of POU2F2 accelerates fracture healing via regulating HMGA1/Wnt/β-catenin signaling pathway. Biosci Biotechnol Biochem 2019; 84:491-499. [PMID: 31782345 DOI: 10.1080/09168451.2019.1695574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To elucidate the role of POU2F2 (POU class 2 homeobox 2) in fracture healing, 30 rats with femoral fracture were randomly grouped into three groups: FF group, LV-POU2F2 group and LV-scramble group. Rats were injected with PBS, lentivirus expressing POU2F2 or scramble lentivirus once a week for 4 weeks. Results showed that overexpressing of POU2F2 promoted fracture healing and callus growth. Besides, overexpressing of POU2F2 promoted protein and mRNA expression of Col10a1, Runx2, Osterix, and Osteocalcin. High Mobility Group AT-hook 1 (HMGA1) is a non-histone protein participating in chromatin remodeling of cells. Western blotting manifested HMGA1/Wnt/β-catenin pathway was activated in POU2F2 group. Moreover, in-vitro study of hMSCs cells supported the above data. In conclusion, POU2F2 promotes fracture healing via activating the HMGA1/Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hanke Zhao
- Department of Orthopedics, Changzhou Wujin People's Hospital, Changzhou, China
| | - Kun Chen
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ye Huang
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Jia HL, Zhou DS. Retracted: Downregulation of microRNA-367 promotes osteoblasts growth and proliferation of mice during fracture by activating the PANX3-mediated Wnt/β-catenin pathway. J Cell Biochem 2019; 120:8247-8258. [PMID: 30556206 DOI: 10.1002/jcb.28108] [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: 07/20/2018] [Accepted: 10/29/2018] [Indexed: 02/02/2023]
Abstract
A majority of people suffering from bone fractures fail to heal and develop a nonunion, which is a challenging orthopedic complication requiring complex and expensive treatment. Previous data showed the inhibition of some microRNAs (miRNAs or miRs) can enhance fracture healing. The objective of the present study is to explore effects of miR-367 on the osteoblasts growth and proliferation of mouse during fracture via the Wnt/β-catenin pathway by targeting PANX3. Primarily, the femur fracture model was successfully established in 66 (C57BL/6) 6-week-old male mice. To verify whether miR-367 target PANX3, we used the target prediction program and performed luciferase activity determination. Subsequently, to figure out the underlying regulatory roles of miR-367 in fracture, osteoblasts were elucidated by treatment with miR-367 mimic, miR-367 inhibitor, or siRNA against PANX3 to determine the expression of miR-367, siPANX3, β-catenin, and Wnt5b as well as cell proliferation and apoptosis. The results demonstrated that PANX3 was verified as a target gene of miR-367. MiR-367 was found to highly expressed but PANX3, β-catenin, and Wnt5b were observed poorly expressed in fracture mice. downregulated miR-367 increased the mRNA and protein expression of PANX3, β-catenin, and Wnt5b, increased cell growth, proliferation, and migration, while decreased cell apoptosis in osteoblasts. Altogether, our study demonstrates that the downregulation of miR-367 may promote osteoblasts growth and proliferation in fracture through the activation of the PANX3-dependent Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Hong-Lei Jia
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Dong-Sheng Zhou
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
13
|
Liu YB, Lin LP, Zou R, Zhao QH, Lin FQ. Silencing long non-coding RNA MEG3 accelerates tibia fraction healing by regulating the Wnt/β-catenin signalling pathway. J Cell Mol Med 2019; 23:3855-3866. [PMID: 30955246 PMCID: PMC6533481 DOI: 10.1111/jcmm.14229] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 01/21/2019] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Abstract
As fracture healing is related to gene expression, fracture healing is prospected to be implicated in long non‐coding RNAs (lncRNAs). This study focuses on the effects of epigenetic silencing of long non‐coding RNA maternally expressed gene 3 (lncRNA MEG3) on fracture healing by regulating the Wnt/β‐catenin signalling pathway. Genes expressed in fracture were screened using bioinformatics and the subcellular location of MEG3 was determined using FISH. Next, we successfully established tibia fracture (TF) models of C57BL/6J and Col2a1‐ICAT mice and the effect of silencing lncRNA MEG3 on fracture healing was detected after TF mice were treated with phosphate buffer saline (PBS), MEG3 siRNA and scramble siRNA. X‐ray imaging, Safranin‐O/fast green and haematoxylin‐eosin (HE) staining and histomorphometrical and biomechanical analysis were adopted to observe and to detect the fracture healing conditions. Additionally, the positive expression of collagen II and osteocalcin was examined using immunohistochemistry. At last, in the in vitro experiment, the relationship of MEG3 and the Wnt/β‐catenin signalling pathway in fraction healing was investigated. MEG3 was located in the cell nucleus. In addition, it was found that MEG3 and the Wnt/β‐catenin signalling pathway were associated with fraction healing. Moreover, silencing MEG3 was proved to elevate callus area and maximum bending load and to furthermore enhance the recanalization of bone marrow cavity. Finally, MEG3 knockdown elevated levels of Col10a1, Runx2, Osterix, Osteocalcin, Wnt10b and β‐catenin/β‐catenin whereas it reduced p‐GSK‐3β/GSK‐3β levels. Taken together, our data supported that epigenetic silencing of lncRNA MEG3 could promote the tibia fracture healing by activating the Wnt/β‐catenin signalling pathway.
Collapse
Affiliation(s)
- Yu-Bao Liu
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Lu-Pan Lin
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Rui Zou
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Qing-Hua Zhao
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Fu-Qing Lin
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| |
Collapse
|
14
|
Yamaguchi Y, Kumagai K, Imai S, Miyatake K, Saito T. Sclerostin is upregulated in the early stage of chondrogenic differentiation, but not required in endochondral ossification in vitro. PLoS One 2018; 13:e0201839. [PMID: 30071108 PMCID: PMC6072128 DOI: 10.1371/journal.pone.0201839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/22/2018] [Indexed: 11/18/2022] Open
Abstract
Sclerostin is a potent inhibitor of the canonical Wnt signaling pathway. Wnt signaling pathways have multiple roles in the regulation of cartilage development, growth, and maintenance. This study focused on the role of sclerostin in the process of chondrogenic differentiation. We hypothesized that sclerostin is essential to induce chondrogenic differentiation and regulate endochondral ossification. ATDC5 cells were used to investigate chondrogenic differentiation and terminal calcification. During chondrogenic differentiation, intrinsic sclerostin was upregulated in the early stage, but downregulated in the late stage. Addition of sclerostin elevated expressions of Sox9 and Col2a1 (P<0.05) and reduced expressions of Runx2, Col10a1, MMP-3, MMP-13, and ADAMTS5 (P<0.05) through inhibition of the Wnt-β-catenin signaling pathway (P<0.05). Terminal calcification was significantly inhibited by sclerostin (P<0.05). In contrast, deletion of sclerostin decreased expressions of Sox9 and Col2a1 (P<0.05), increased expressions of Runx2, Col10a1, MMP-3, and MMP-13 (P<0.05), and promoted terminal calcification (P<0.05). This study provides insights into the possible role of sclerostin in the regulation of chondrogenic differentiation. Sclerostin is upregulated in the early stage of chondrogenic differentiation, but is not required in endochondral ossification. Sclerostin is a candidate modulator for chondrogenic differentiation.
Collapse
Affiliation(s)
- Yasuteru Yamaguchi
- Department of Orthopaedic Surgery and Muscloskeletal Science, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Ken Kumagai
- Department of Orthopaedic Surgery and Muscloskeletal Science, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- * E-mail:
| | - Sosuke Imai
- Department of Orthopaedic Surgery and Muscloskeletal Science, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Kazuma Miyatake
- Department of Orthopaedic Surgery and Muscloskeletal Science, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Tomoyuki Saito
- Department of Orthopaedic Surgery and Muscloskeletal Science, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
15
|
Wong SA, Rivera KO, Miclau T, Alsberg E, Marcucio RS, Bahney CS. Microenvironmental Regulation of Chondrocyte Plasticity in Endochondral Repair-A New Frontier for Developmental Engineering. Front Bioeng Biotechnol 2018; 6:58. [PMID: 29868574 PMCID: PMC5962790 DOI: 10.3389/fbioe.2018.00058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022] Open
Abstract
The majority of fractures heal through the process of endochondral ossification, in which a cartilage intermediate forms between the fractured bone ends and is gradually replaced with bone. Recent studies have provided genetic evidence demonstrating that a significant portion of callus chondrocytes transform into osteoblasts that derive the new bone. This evidence has opened a new field of research aimed at identifying the regulatory mechanisms that govern chondrocyte transformation in the hope of developing improved fracture therapies. In this article, we review known and candidate molecular pathways that may stimulate chondrocyte-to-osteoblast transformation during endochondral fracture repair. We also examine additional extrinsic factors that may play a role in modulating chondrocyte and osteoblast fate during fracture healing such as angiogenesis and mineralization of the extracellular matrix. Taken together the mechanisms reviewed here demonstrate the promising potential of using developmental engineering to design therapeutic approaches that activate endogenous healing pathways to stimulate fracture repair.
Collapse
Affiliation(s)
- Sarah A Wong
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,School of Dentistry, University of California, San Francisco, San Francisco, CA, United States
| | - Kevin O Rivera
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,School of Dentistry, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Eben Alsberg
- Department of Orthopaedic Surgery and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Ralph S Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,School of Dentistry, University of California, San Francisco, San Francisco, CA, United States
| | - Chelsea S Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
16
|
Goto H, Nishio M, To Y, Oishi T, Miyachi Y, Maehama T, Nishina H, Akiyama H, Mak TW, Makii Y, Saito T, Yasoda A, Tsumaki N, Suzuki A. Loss of Mob1a/b in mice results in chondrodysplasia due to YAP1/TAZ-TEAD-dependent repression of SOX9. Development 2018; 145:dev.159244. [PMID: 29511023 DOI: 10.1242/dev.159244] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/19/2018] [Indexed: 12/30/2022]
Abstract
Hippo signaling is modulated in response to cell density, external mechanical forces, and rigidity of the extracellular matrix (ECM). The Mps one binder kinase activator (MOB) adaptor proteins are core components of Hippo signaling and influence Yes-associated protein 1 (YAP1) and transcriptional co-activator with PDZ-binding motif (TAZ), which are potent transcriptional regulators. YAP1/TAZ are key contributors to cartilage and bone development but the molecular mechanisms by which the Hippo pathway controls chondrogenesis are largely unknown. Cartilage is rich in ECM and also subject to strong external forces - two upstream factors regulating Hippo signaling. Chondrogenesis and endochondral ossification are tightly controlled by growth factors, morphogens, hormones, and transcriptional factors that engage in crosstalk with Hippo-YAP1/TAZ signaling. Here, we generated tamoxifen-inducible, chondrocyte-specific Mob1a/b-deficient mice and show that hyperactivation of endogenous YAP1/TAZ impairs chondrocyte proliferation and differentiation/maturation, leading to chondrodysplasia. These defects were linked to suppression of SOX9, a master regulator of chondrogenesis, the expression of which is mediated by TEAD transcription factors. Our data indicate that a MOB1-dependent YAP1/TAZ-TEAD complex functions as a transcriptional repressor of SOX9 and thereby negatively regulates chondrogenesis.
Collapse
Affiliation(s)
- Hiroki Goto
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.,Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Miki Nishio
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.,Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Yoko To
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Tatsuya Oishi
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yosuke Miyachi
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.,Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Tomohiko Maehama
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Hiroshi Nishina
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Haruhiko Akiyama
- Department of Orthopaedic Surgery, Gifu University School of Medicine, Gifu 501-1194, Japan
| | - Tak Wah Mak
- Campbell Family Institute for Breast Cancer Research at the Princess Margaret Cancer Centre, University Health Network, Toronto M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, University Health Network, Toronto M5G 2C1, Canada
| | - Yuma Makii
- Department of Sensory and Motor System Medicine, Faculty of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Taku Saito
- Department of Sensory and Motor System Medicine, Faculty of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Akihiro Yasoda
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Noriyuki Tsumaki
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Akira Suzuki
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan .,Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
17
|
Li Z, Pan J, Ma J, Zhang Z, Bai Y. Microarray gene expression of periosteum in spontaneous bone regeneration of mandibular segmental defects. Sci Rep 2017; 7:13535. [PMID: 29051537 PMCID: PMC5648814 DOI: 10.1038/s41598-017-13586-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/25/2017] [Indexed: 12/14/2022] Open
Abstract
Spontaneous bone regeneration could occur to reestablish mandibular bony continuity in patients who underwent partial or total mandibulectomy for tumors with periosteum-preserving. However, scarce data is available related to the precise role of periosteum in this bone regeneration. Therefore we aimed to investigate the gene expression of periosteum that were involved in the mandibular bone regeneration. Mandibular segmental defects were created in six mini-pigs with periosteum preserved. The periosteum of defects and control site were harvested at 1 and 2 weeks. Gene ontology (GO) analysis showed that the mechanisms concerning immature wound healing were clearly up-regulated at week 1. In contrast, by week-2, the GO categories of skeletal development, ossification and bone mineralization were significantly over-represented at week-2 with several genes encoding cell differentiation, extracellular matrix formation, and anatomical structure development. Furthermore, Tgfβ/Bmp, Wnt and Notch signaling were all related to the osteogenic process in this study. Besides osteogenesis, genes related to angiogenesis and neurogenesis were also prominent at week-2. These findings revealed that the gene expression profile of the periosteum’s cells participating in bone regeneration varied in different time points, and numbers of candidate genes that differentially expressed during early healing stages of intramembranous bone regeneration were suggested.
Collapse
Affiliation(s)
- Zheyi Li
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Juli Pan
- School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Jinling Ma
- School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Zhen Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| |
Collapse
|
18
|
Yuan C, Cai J. Time-series expression profile analysis of fracture healing in young and old mice. Mol Med Rep 2017; 16:4529-4536. [PMID: 28849124 PMCID: PMC5647013 DOI: 10.3892/mmr.2017.7198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/13/2017] [Indexed: 11/06/2022] Open
Abstract
Bone fracture healing is a complex process, which is associated with several factors, including age and osteoporosis. Certain genes and biological processes that may contribute to fracture healing, have been identified following developments in systems biology and molecular biology technologies, which may benefit the treatment of bone fractures. The present study identified key genes, which may be important in fracture healing through bioinformatics analysis of gene microarray datasets from the Gene Expression Omnibus. Gene clusters, which were consistently up/downregulated through time following a fracture in young (6-week-old) mice and old (8-month-old retired breeders) mice were obtained via soft clustering of differentially expressed genes (DEGs) between samples at 1 and 3 days, 1 and 5 days, and 3 and 5 days post-fracture in the two age groups, based on the Mfuzz package of R. Functional enrichment analysis of gene clusters using the Database for Annotation, Visualization and Integrated Discovery indicated that biological processes and pathways, including those associated with bone development, skeletal system development, amino sugar and nucleotide sugar metabolism, were significantly enriched in these up/downregulated genes. Of note, a total of 207 overlapped consistently upregulated genes were obtained between the two age groups, whereas no overlap was identified between the two lists of consistently downregulated genes. The overlapped genes were found to be associated with the biological processes of blood vessel development, vasculature development and skeletal system development, compared with all genes in the clusters. In addition, certain genes, including epidermal growth factor-like domain multiple 6 (EGFL6), kazal-type serine peptidase inhibitor domain 1 (KAZALD1), olfactomedin 2B (OLFM2B), collagen type III α1 (COL3A1), collagen type II α1 (COL2A1), von Willebrand factor A domain-containing 1 (VWA1), elastin microfibril interfacer 1 (EMILIN1) and aggrecan (ACAN), of the extracellular matrix organization, a process performed at the cellular level and resulting in the assembly and arrangement of constituent parts, were confirmed to be associated with fracture healing via reverse transcription-quantitative polymerase chain reaction analysis. The present study identified certain genes and biological processes/pathways, which may be associated with fracture healing and may assist in fundamental investigations and treatment in the future.
Collapse
Affiliation(s)
- Chun Yuan
- Department of Orthopedics, Jinan Military General Hospital, Jinan, Shandong 250031, P.R. China
| | - Jinfang Cai
- Department of Orthopedics, Jinan Military General Hospital, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
19
|
Bao Q, Chen S, Qin H, Feng J, Liu H, Liu D, Li A, Shen Y, Zhao Y, Li J, Zong Z. An appropriate Wnt/β-catenin expression level during the remodeling phase is required for improved bone fracture healing in mice. Sci Rep 2017; 7:2695. [PMID: 28578392 PMCID: PMC5457421 DOI: 10.1038/s41598-017-02705-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/18/2017] [Indexed: 02/03/2023] Open
Abstract
Accumulating evidence demonstrates that the Wnt/β-catenin signaling pathway plays a dominant role in bone repair. However, the role of Wnt/β-catenin signaling in the remodeling phase during bone fracture healing is currently unknown. In the present study, β-catenin was activated at different levels or deleted in mice at the late stage of fracture healing, and the effects on healing quality were investigated. Deletion of β-catenin disturbed bone remodeling, as confirmed by increased bone resorption and decreased bone formation, and significantly decreased bone strength compared with wildtype mice. In addition, the constitutive activation of β-catenin significantly increased the bone mass and delayed the bone remodeling process, resulting in slightly impaired bone strength. In contrast, a slight activation of β-catenin significantly increased bone formation and slightly hindered bone resorption. These effects lead to improved bone fracture healing quality compared with wildtype mice. In summary, the present study provides the first demonstration showing that Wnt/β-catenin signaling should be maintained at a slightly activated level during the late stage of fracture healing to ensure better bone fracture repair.
Collapse
Affiliation(s)
- Quanwei Bao
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Sixu Chen
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Hao Qin
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Jianquan Feng
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX, 75246, USA
| | - Huayu Liu
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Daocheng Liu
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Ang Li
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Yue Shen
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Yufeng Zhao
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Junfeng Li
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China
| | - Zhaowen Zong
- State Key Laboratory of Trauma, Burn and Combined injury, Department of Trauma Surgery, Daping Hospital, Third Military Medical University, ChongQing, 400042, China.
| |
Collapse
|
20
|
Pflug T, Huynh-Do U, Rudloff S. Reduced β-catenin expression affects patterning of bone primordia, but not bone maturation. Biol Open 2017; 6:582-588. [PMID: 28347990 PMCID: PMC5450319 DOI: 10.1242/bio.023572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Wnt/β-catenin signaling is involved in patterning of bone primordia, but also plays an important role in the differentiation of chondrocytes and osteoblasts. During these processes the level of β-catenin must be tightly regulated. Excess β-catenin leads to conditions with increased bone mass, whereas loss of β-catenin is associated with osteoporosis or, in extreme cases, the absence of limbs. In this study, we examined skeletogenesis in mice, which retain only 25% of β-catenin. These embryos showed severe morphological abnormalities of which the lack of hindlimbs and misshaped front paws were the most striking. Surprisingly however, calcification of bone primordia occurred normally. Moreover, the Wnt-dependent regulatory network of transcription factors driving the differentiation of cartilage and bone, as well as the expression of extracellular matrix components, were preserved. These findings show that 25% β-catenin is insufficient for the correct patterning of bone primordia, but sufficient for their mineralization. Our approach helps to identify bone morphogenetic processes that can proceed normally even at low β-catenin levels, in contrast to those that require high β-catenin dosages. This information could be exploited to improve the treatment of bone diseases by fine-tuning the individual β-catenin dosage requirements. Summary: The correct allocation and patterning of skeletal elements during development requires a higher dosage of β-catenin than the maturation and mineralization of these primordia.
Collapse
Affiliation(s)
- Tobias Pflug
- Department of Clinical Research, Department of Nephrology and Hypertension, Bern University Hospital, University of Bern, Freiburgstrasse 15, Bern CH-3010, Switzerland
| | - Uyen Huynh-Do
- Department of Clinical Research, Department of Nephrology and Hypertension, Bern University Hospital, University of Bern, Freiburgstrasse 15, Bern CH-3010, Switzerland
| | - Stefan Rudloff
- Department of Clinical Research, Department of Nephrology and Hypertension, Bern University Hospital, University of Bern, Freiburgstrasse 15, Bern CH-3010, Switzerland
| |
Collapse
|
21
|
Suchorska WM, Augustyniak E, Richter M, Trzeciak T. Gene expression profile in human induced pluripotent stem cells: Chondrogenic differentiation in vitro, part A. Mol Med Rep 2017; 15:2387-2401. [PMID: 28447755 PMCID: PMC5428238 DOI: 10.3892/mmr.2017.6334] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/26/2017] [Indexed: 12/20/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) offer promise in regenerative medicine, however more data are required to improve understanding of key aspects of the cell differentiation process, including how specific chondrogenic processes affect the gene expression profile of chondrocyte-like cells and the relative value of cell differentiation markers. The main aims of the present study were as follows: To determine the gene expression profile of chondrogenic-like cells derived from hiPSCs cultured in mediums conditioned with HC-402-05a cells or supplemented with transforming growth factor β3 (TGF-β3), and to assess the relative utility of the most commonly used chondrogenic markers as indicators of cell differentiation. These issues are relevant with regard to the use of human fibroblasts in the reprogramming process to obtain hiPSCs. Human fibroblasts are derived from the mesoderm and thus share a wide range of properties with chondrocytes, which also originate from the mesenchyme. Thus, the exclusion of dedifferentiation instead of chondrogenic differentiation is crucial. The hiPSCs were obtained from human primary dermal fibroblasts during a reprogramming process. Two methods, both involving embryoid bodies (EB), were used to obtain chondrocytes from the hiPSCs: EBs formed in a chondrogenic medium supplemented with TGF-β3 (10 ng/ml) and EBs formed in a medium conditioned with growth factors from HC-402-05a cells. Based on immunofluorescence and reverse transcription-quantiative polymerase chain reaction analysis, the results indicated that hiPSCs have the capacity for effective chondrogenic differentiation, in particular cells differentiated in the HC-402-05a-conditioned medium, which present morphological features and markers that are characteristic of mature human chondrocytes. By contrast, cells differentiated in the presence of TGF-β3 may demonstrate hypertrophic characteristics. Several genes [paired box 9, sex determining region Y-box (SOX) 5, SOX6, SOX9 and cartilage oligomeric matrix protein] were demonstrated to be good markers of early hiPSC chondrogenic differentiation: Insulin-like growth factor 1, Tenascin-C, and β-catenin were less valuable. These observations provide valuable data on the use of hiPSCs in cartilage tissue regeneration.
Collapse
Affiliation(s)
| | - Ewelina Augustyniak
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61‑866 Poznan, Poland
| | - Magdalena Richter
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61‑545 Poznan, Poland
| | - Tomasz Trzeciak
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61‑545 Poznan, Poland
| |
Collapse
|
22
|
Kostenuik P, Mirza FM. Fracture healing physiology and the quest for therapies for delayed healing and nonunion. J Orthop Res 2017; 35:213-223. [PMID: 27743449 PMCID: PMC6120140 DOI: 10.1002/jor.23460] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 10/07/2016] [Indexed: 02/04/2023]
Abstract
Delayed healing and nonunion of fractures represent enormous burdens to patients and healthcare systems. There are currently no approved pharmacological agents for the treatment of established nonunions, or for the acceleration of fracture healing, and no pharmacological agents are approved for promoting the healing of closed fractures. Yet several pharmacologic agents have the potential to enhance some aspects of fracture healing. In preclinical studies, various agents working across a broad spectrum of molecular pathways can produce larger, denser and stronger fracture calluses. However, untreated control animals in most of these studies also demonstrate robust structural and biomechanical healing, leaving unclear how these interventions might alter the healing of recalcitrant fractures in humans. This review describes the physiology of fracture healing, with a focus on aspects of natural repair that may be pharmacologically augmented to prevent or treat delayed or nonunion fractures (collectively referred to as DNFs). The agents covered in this review include recombinant BMPs, PTH/PTHrP receptor agonists, activators of Wnt/β-catenin signaling, and recombinant FGF-2. Agents from these therapeutic classes have undergone extensive preclinical testing and progressed to clinical fracture healing trials. Each can promote bone formation, which is important for the stability of bridged calluses, and some but not all can also promote cartilage formation, which may be critical for the initial bridging and subsequent stabilization of fractures. Appropriately timed stimulation of chondrogenesis and osteogenesis in the fracture callus may be a more effective approach for preventing or treating DNFs compared with stimulation of osteogenesis alone. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:213-223, 2017.
Collapse
Affiliation(s)
- Paul Kostenuik
- School of DentistryUniversity of MichiganPhylon Pharma ServicesNewbury ParkCalifornia
| | | |
Collapse
|
23
|
Lin HN, Cottrell J, O'Connor JP. Variation in lipid mediator and cytokine levels during mouse femur fracture healing. J Orthop Res 2016; 34:1883-1893. [PMID: 26919197 DOI: 10.1002/jor.23213] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/23/2016] [Indexed: 02/04/2023]
Abstract
Fracture healing is regulated by a variety of inflammatory mediators and growth factors which act over time to regenerate the injured tissue. This study used a mouse femur fracture model to quantify the temporal expression pattern of lipid mediators, cytokines, and related mRNAs during healing. Cyclooxygenase (COX-1 and COX-2) and 5-lipoxygenase (5-LO) derived lipid mediators, cytokines, and mRNA levels were quantified using mass spectrometry (LC-MS/MS), bead-based multiplex assays (xMAP), and quantitative PCR of cDNA (RTqPCR), respectively. Our analysis found that, the early inflammatory response (between 0 and 4 days after fracture) in the mouse femur fracture model coincided with elevated levels of COX-derived lipid mediators and inflammatory cytokines but with decreased levels of 5-LO-derived lipid mediators. Further, the COX-derived lipid mediators remained elevated for at least 7 days after fracture, suggesting that the COX-derived lipid mediators have additional functions during later phases of the fracture healing response. Differences were also found between mRNA levels and corresponding cytokines and lipid mediator levels, supporting a role for post-transcriptional regulation of gene expression. The temporal changes in fracture callus lipid mediator levels and inflammatory cytokines support a general positive role for inflammatory cytokines and COX-derived lipid mediators on fracture healing and a general negative role for 5-lipoxygenase derived lipid mediators during the initial stages of repair. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1883-1893, 2016.
Collapse
Affiliation(s)
- Hsuan-Ni Lin
- Department of Biochemistry and Molecular Biology, Rutgers, the State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, New Jersey 07103.,Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, 185 South Orange Avenue, Newark, New Jersey 07103
| | - Jessica Cottrell
- Department of Biochemistry and Molecular Biology, Rutgers, the State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, New Jersey 07103.,Department of Biological Sciences, Seton Hall University, 400 South Orange Avenue, South Orange, New Jersey 07079
| | - J Patrick O'Connor
- Department of Biochemistry and Molecular Biology, Rutgers, the State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, New Jersey 07103.,Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, 185 South Orange Avenue, Newark, New Jersey 07103
| |
Collapse
|
24
|
Lau KHW, Rundle CH, Zhou XD, Baylink DJ, Sheng MHC. Conditional deletion of IGF-I in osteocytes unexpectedly accelerates bony union of the fracture gap in mice. Bone 2016; 92:18-28. [PMID: 27519969 DOI: 10.1016/j.bone.2016.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/03/2016] [Accepted: 08/08/2016] [Indexed: 01/14/2023]
Abstract
This study evaluated the effects of deficient IGF-I expression in osteocytes on fracture healing. Transgenic mice with conditional knockout (cKO) of Igf1 in osteocytes were generated by crossing Dmp1-Cre mice with Igf1 flox mice. Fractures were created on the mid-shaft of tibia of 12-week-old male cKO mice and wild-type (WT) littermates by three-point bending. At 21 and 28days post-fracture healing, the increases in cortical bone mineral density, mineral content, bone area, and thickness, as well as sub-cortical bone mineral content at the fracture site were each greater in cKO calluses than in WT calluses. There were 85% decrease in the cartilage area and >2-fold increase in the number of osteoclasts in cKO calluses at 14days post-fracture, suggesting a more rapid remodeling of endochondral bone. The upregulation of mRNA levels of osteoblast marker genes (cbfa1, alp, Opn, and Ocn) was greater in cKO calluses than in WT calluses. μ-CT analysis suggested an accelerated bony union of the fracture gap in cKO mice. The Sost mRNA level was reduced by 50% and the Bmp2 mRNA level was increased 3-fold in cKO fractures at 14days post-fracture, but the levels of these two mRNAs in WT fractures were unchanged, suggesting that the accelerated fracture repair may in part act through the Wnt and/or BMP signaling. In conclusion, conditional deletion of Igf1 in osteocytes not only did not impair, but unexpectedly enhanced, bony union of the fracture gap. The accelerated bony union was due in part to upregulation of the Wnt and BMP2 signaling in response to deficient osteocyte-derived IGF-I expression, which in turn favors intramembranous over endochondral bone repair.
Collapse
Affiliation(s)
- Kin-Hing W Lau
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA; Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA
| | - Charles H Rundle
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA
| | - Xiao-Dong Zhou
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Matilda H-C Sheng
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
25
|
Yap1 Regulates Multiple Steps of Chondrocyte Differentiation during Skeletal Development and Bone Repair. Cell Rep 2016; 14:2224-2237. [DOI: 10.1016/j.celrep.2016.02.021] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 01/03/2016] [Accepted: 01/29/2016] [Indexed: 01/06/2023] Open
|
26
|
The ameloblastin extracellular matrix molecule enhances bone fracture resistance and promotes rapid bone fracture healing. Matrix Biol 2016; 52-54:113-126. [PMID: 26899203 DOI: 10.1016/j.matbio.2016.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) provides structural support, cell migration anchorage, cell differentiation cues, and fine-tuned cell proliferation signals during all stages of bone fracture healing, including cartilaginous callus formation, callus remodeling, and bony bridging of the fracture gap. In the present study we have defined the role of the extracellular matrix protein ameloblastin (AMBN) in fracture resistance and fracture healing of mouse long bones. To this end, long bones from WT and AMBN(Δ5-6) truncation model mice were subjected to biomechanical analysis, fracture healing assays, and stem cell colony formation comparisons. The effect of exogenous AMBN addition to fracture sites was also determined. Our data indicate that lack of a functional AMBN in the bone matrix resulted in 31% decreased femur bone mass and 40% reduced energy to failure. On a cellular level, AMBN function inhibition diminished the proliferative capacity of fracture repair callus cells, as evidenced by a 58% reduction in PCNA and a 40% reduction in Cyclin D1 gene expression, as well as PCNA immunohistochemistry. In terms of fracture healing, AMBN truncation was associated with an enhanced and prolonged chondrogenic phase, resulting in delayed mineralized tissue gene expression and delayed ossification of the fracture repair callus. Underscoring a role of AMBN in fracture healing, there was a 6.9-fold increase in AMBN expression at the fracture site one week after fracture, and distinct AMBN immunolabeling in the fracture gap. Finally, application of exogenous AMBN protein to bone fracture sites accelerated callus formation and bone fracture healing (33% increase in bone volume and 19% increase in bone mineral density), validating the findings of our AMBN loss of function studies. Together, these data demonstrate the functional importance of the AMBN extracellular matrix protein in bone fracture prevention and rapid fracture healing.
Collapse
|
27
|
Troib A, Guterman M, Rabkin R, Landau D, Segev Y. Endurance exercise and growth hormone improve bone formation in young and growth-retarded chronic kidney disease rats. Nephrol Dial Transplant 2015; 31:1270-9. [PMID: 26560811 DOI: 10.1093/ndt/gfv373] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/05/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Childhood chronic kidney disease (CKD) is associated with both short stature and abnormal bone mineralization. Normal longitudinal growth depends on proper maturation of epiphyseal growth plate (EGP) chondrocytes, leading to the formation of trabecular bone in the primary ossification centre. We have recently shown that linear growth impairment in CKD is associated with impaired EGP growth hormone (GH) receptor signalling and that exercise improved insulin-like growth factor I (IGF-I) signalling in CKD-related muscle atrophy. METHODS In this study, 20-day-old rats underwent 5/6 nephrectomy (CKD) or sham surgery (C) and were exercised with treadmill, with or without GH supplementation. RESULTS CKD-related growth retardation was associated with a widened EGP hypertrophic zone. This was not fully corrected by exercise (except for tibial length). Exercise in CKD improved the expression of EGP key factors of endochondral ossification such as IGF-I, vascular endothelial growth factor (VEGF), receptor activator of nuclear factor kappa-B ligand (RANKL) and osteocalcin. Combining GH treatment with treadmill exercise for 2 weeks improved the decreased trabecular bone volume in CKD, as well as the expression of growth plate runt-related transcription factor 2, RANKL, metalloproteinase 13 and VEGF, while GH treatment alone could not do that. CONCLUSIONS Treadmill exercise improves tibial bone linear growth, as well as growth plate local IGF-I. When combined with GH treatment, running exercise shows beneficial effects on trabecular bone formation, suggesting the potential benefit of this combination for CKD-related short stature and bone disease.
Collapse
Affiliation(s)
- Ariel Troib
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Mayan Guterman
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Ralph Rabkin
- Research Service, Veterans Affairs Health Care Palo Alto, Stanford University, Stanford, CA, USA Medicine Department/Renal Division, Stanford University, Stanford, CA, USA
| | - Daniel Landau
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel Department of Pediatrics, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Yael Segev
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
28
|
Continuous infusion of angiotensin II modulates hypertrophic differentiation and apoptosis of chondrocytes in cartilage formation in a fracture model mouse. Hypertens Res 2015; 38:382-93. [PMID: 25693858 DOI: 10.1038/hr.2015.18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/08/2014] [Accepted: 12/15/2014] [Indexed: 12/16/2022]
Abstract
Although components of the renin-angiotensin system (RAS) are reported to be expressed in cultured chondrocytes and cartilage, little is known about the precise function of Angiotensin II (Ang II) in chondrocytes. In this study, we employed a rib fracture model mouse to investigate the effect of Ang II on chondrocytes. Ang II type 1 receptor (AT1R) was expressed in chondrocytes in the growth plate of mouse tibia. Continuous infusion of Ang II to rib-fractured mice resulted in a significant increase in the volume of cartilage, suggesting Ang II-induced hypertrophic differentiation of chondrocytes. It was also confirmed by a significant increase in the mRNA expression of Sox9 and runt-related transcription factor 2 (Runx2), which are genes related to chondrocyte differentiation, and type X collagen, matrix metalloproteinase (MMP)-13 and Indian hedgehog (Ihh), which are hypertrophic chondrocyte-specific molecular markers. Chondrocyte hypertrophy with upregulation of these genes was attenuated by administration of olmesartan, an AT1R blocker, but not by hydralazine. Moreover, Ang II infusion significantly suppressed apoptosis of chondrocytes, accompanied by significant induction of mRNA expression of bcl-2 and bcl-xL. Olmesartan, but not hydralazine, significantly attenuated the reduction of apoptotic cells and the increase in anti-apoptotic genes induced by Ang II infusion. Overall, the present study demonstrated that Ang II promoted hypertrophic differentiation of chondrocytes and reduced apoptosis of hypertrophic chondrocytes independently of high blood pressure. The present data indicate the role of Ang II in cartilage, and might provide a new concept for treatment of cartilage diseases.
Collapse
|
29
|
Haffner-Luntzer M, Heilmann A, Rapp AE, Beie S, Schinke T, Amling M, Ignatius A, Liedert A. Midkine-deficiency delays chondrogenesis during the early phase of fracture healing in mice. PLoS One 2014; 9:e116282. [PMID: 25551381 PMCID: PMC4281158 DOI: 10.1371/journal.pone.0116282] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/05/2014] [Indexed: 01/02/2023] Open
Abstract
The growth and differentiation factor midkine (Mdk) plays an important role in bone development and remodeling. Mdk-deficient mice display a high bone mass phenotype when aged 12 and 18 months. Furthermore, Mdk has been identified as a negative regulator of mechanically induced bone formation and it induces pro-chondrogenic, pro-angiogenic and pro-inflammatory effects. Together with the finding that Mdk is expressed in chondrocytes during fracture healing, we hypothesized that Mdk could play a complex role in endochondral ossification during the bone healing process. Femoral osteotomies stabilized using an external fixator were created in wildtype and Mdk-deficient mice. Fracture healing was evaluated 4, 10, 21 and 28 days after surgery using 3-point-bending, micro-computed tomography, histology and immunohistology. We demonstrated that Mdk-deficient mice displayed delayed chondrogenesis during the early phase of fracture healing as well as significantly decreased flexural rigidity and moment of inertia of the fracture callus 21 days after fracture. Mdk-deficiency diminished beta-catenin expression in chondrocytes and delayed presence of macrophages during early fracture healing. We also investigated the impact of Mdk knockdown using siRNA on ATDC5 chondroprogenitor cells in vitro. Knockdown of Mdk expression resulted in a decrease of beta-catenin and chondrogenic differentiation-related matrix proteins, suggesting that delayed chondrogenesis during fracture healing in Mdk-deficient mice may be due to a cell-autonomous mechanism involving reduced beta-catenin signaling. Our results demonstrated that Mdk plays a crucial role in the early inflammation phase and during the development of cartilaginous callus in the fracture healing process.
Collapse
Affiliation(s)
- Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Aline Heilmann
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anna Elise Rapp
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Simon Beie
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
30
|
Hadjiargyrou M, O'Keefe RJ. The convergence of fracture repair and stem cells: interplay of genes, aging, environmental factors and disease. J Bone Miner Res 2014; 29:2307-22. [PMID: 25264148 PMCID: PMC4455538 DOI: 10.1002/jbmr.2373] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 08/11/2014] [Accepted: 09/10/2014] [Indexed: 01/07/2023]
Abstract
The complexity of fracture repair makes it an ideal process for studying the interplay between the molecular, cellular, tissue, and organ level events involved in tissue regeneration. Additionally, as fracture repair recapitulates many of the processes that occur during embryonic development, investigations of fracture repair provide insights regarding skeletal embryogenesis. Specifically, inflammation, signaling, gene expression, cellular proliferation and differentiation, osteogenesis, chondrogenesis, angiogenesis, and remodeling represent the complex array of interdependent biological events that occur during fracture repair. Here we review studies of bone regeneration in genetically modified mouse models, during aging, following environmental exposure, and in the setting of disease that provide insights regarding the role of multipotent cells and their regulation during fracture repair. Complementary animal models and ongoing scientific discoveries define an increasing number of molecular and cellular targets to reduce the morbidity and complications associated with fracture repair. Last, some new and exciting areas of stem cell research such as the contribution of mitochondria function, limb regeneration signaling, and microRNA (miRNA) posttranscriptional regulation are all likely to further contribute to our understanding of fracture repair as an active branch of regenerative medicine.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | | |
Collapse
|
31
|
Beier EE, Sheu TJ, Buckley T, Yukata K, O'Keefe R, Zuscik MJ, Puzas JE. Inhibition of beta-catenin signaling by Pb leads to incomplete fracture healing. J Orthop Res 2014; 32:1397-405. [PMID: 25044211 PMCID: PMC4314692 DOI: 10.1002/jor.22677] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 06/10/2014] [Indexed: 02/04/2023]
Abstract
There is strong evidence in the clinical literature to suggest that elevated lead (Pb) exposure impairs fracture healing. Since Pb has been demonstrated to inhibit bone formation, and Wnt signaling is an important anabolic pathway in chondrocyte maturation and endochondral ossification, we investigated the impact of Wnt therapy on Pb-exposed mice undergoing bone repair in a mouse tibial fracture model. We established that tibial fracture calluses from Pb-treated mice were smaller and contained less mineralized tissue than vehicle controls. This resulted in the persistence of immature cartilage in the callus and decreased β-catenin levels. Reduction of β-catenin protein was concurrent with systemic elevation of LRP5/6 antagonists DKK1 and sclerostin in Pb-exposed mice throughout fracture healing. β-catenin stimulation by the GSK3 inhibitor BIO reversed these molecular changes and restored the amount of mineralized callus. Overall, Pb is identified as a potent inhibitor of endochondral ossification in vivo with correlated effects on bone healing with noted deficits in β-catenin signaling, suggesting the Wnt/β-catenin as a pivotal pathway in the influence of Pb on fracture repair.
Collapse
Affiliation(s)
- Eric E Beier
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, New York, 14642; Department of Environmental Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, New York, 14642
| | | | | | | | | | | | | |
Collapse
|
32
|
Abou-Khalil R, Colnot C. Cellular and molecular bases of skeletal regeneration: what can we learn from genetic mouse models? Bone 2014; 64:211-21. [PMID: 24709685 DOI: 10.1016/j.bone.2014.03.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 03/19/2014] [Accepted: 03/26/2014] [Indexed: 10/25/2022]
Abstract
Although bone repairs through a very efficient regenerative process in 90% of the patients, many factors can cause delayed or impaired healing. To date, there are no reliable biological parameters to predict or diagnose bone repair defects. Orthopedic surgeons mostly base their diagnoses on radiographic analyses. With the recent progress in our understanding of the bone repair process, new methods may be envisioned. Animal models have allowed us to define the key steps of bone regeneration and the biological and mechanical factors that may influence bone healing in positive or negative ways. Most importantly, small animal models such as mice have provided powerful tools to apprehend the genetic bases of normal and impaired bone healing. The current review presents a state of the art of the genetically modified mouse models that have advanced our understanding of the cellular and molecular components of bone regeneration and repair. The review illustrates the use of these models to define the role of inflammation, skeletal cell lineages, signaling pathways, the extracellular matrix, osteoclasts and angiogenesis. These genetic mouse models promise to change the field of orthopedic surgery to help establish genetic predispositions for delayed repair, develop models of non-union that mimic the human conditions and elaborate new therapeutic approaches to enhance bone regeneration.
Collapse
Affiliation(s)
- Rana Abou-Khalil
- INSERM UMR1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Céline Colnot
- INSERM UMR1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France.
| |
Collapse
|
33
|
Baht GS, Silkstone D, Nadesan P, Whetstone H, Alman BA. Activation of hedgehog signaling during fracture repair enhances osteoblastic-dependent matrix formation. J Orthop Res 2014; 32:581-6. [PMID: 24347536 DOI: 10.1002/jor.22562] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 11/19/2013] [Indexed: 02/04/2023]
Abstract
Fracture repair is a well orchestrated process involving various cell types and signaling molecules. The hedgehog signaling pathway is activated in chondrocytes during fracture repair and is known to regulate chondrogenesis however, its role in osteoblasts during injury is yet unknown. In this study we observed tibial fracture repair in mice in which hedgehog signaling was modulated through genetic alterations of the pathway activator, smoothened. Levels of the hedgehog target genes Gli1 and Ptch1 in wildtype mice were upregulated in fracture calluses throughout the repair process. Forced activation of the hedgehog pathway in ubiquitous fashion resulted in increased matrix deposition in the fracture callus. Interestingly, inhibition in chondrocytes did not alter the fracture repair phenotype, while activation of hedgehog in osteoblasts was a requirement for normal fracture repair. In vitro, transcript levels of Gli1 and Ptch1 were elevated during osteoblastogenesis. Activation of hedgehog signaling positively affected osteoblastic differentiation and mineralization as detected using alkaline phosphatase and Von Kossa staining and Alp and Col1 expression. Here we show that the hedgehog signaling pathway plays a critical role in osteoblasts during fracture repair: inhibition of the pathway in osteoblasts leads to decreased matrix at the fracture site while activation increased matrix deposition.
Collapse
Affiliation(s)
- Gurpreet S Baht
- Department of Developmental & Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | | | | | | | | |
Collapse
|
34
|
Lauing KL, Sundaramurthy S, Nauer RK, Callaci JJ. Exogenous activation of Wnt/β-catenin signaling attenuates binge alcohol-induced deficient bone fracture healing. Alcohol Alcohol 2014; 49:399-408. [PMID: 24627571 DOI: 10.1093/alcalc/agu006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Excessive alcohol consumption is associated with fracture non-union. Canonical Wnt pathway signaling activity regulates normal fracture healing. We previously demonstrated that binge alcohol exposure modulates β-catenin levels in the fracture callus of mice. Here, we sought to determine whether exogenous enhancement β-catenin signaling activity could restore normal fracture healing to binge-exposed mice. METHODS C57BL/6 male mice were exposed to episodic alcohol or saline for 6 total days of alcohol exposure over a 2-week period. Following alcohol exposure, mice were subjected to a stabilized mid-shaft tibia fracture. Beginning 4 days post-injury, mice received daily injections of either lithium chloride or saline subcutaneously. Protein levels of activated, inactivated, and total β-catenin and GSK-3β in fracture calluses were measured at post-injury day 9. Biomechanical strength testing and histology of callus tissue was assessed at post fracture day 14. RESULTS Binge alcohol was associated with decreased callus biomechanical strength, and reduced cartilaginous callus formation. Alcohol decreased levels of callus-associated activated β-catenin while concomitantly increasing the levels of inactive β-catenin at post-injury day 9. Alcohol also increased callus associated activated GSK-3β at post-injury day 9. Lithium chloride (an inhibitor of GSK-3β) treatment increased activated β-catenin protein levels, significantly decreased activated GSK-3β and restored cartilaginous callus formation and endochondral ossification. CONCLUSION These data link alcohol-impaired fracture healing with deregulation of Canonical Wnt signaling activity in the fracture callus. Exogenous activation of the Wnt pathway using LiCl attenuated the damaging effects of binge alcohol exposure on the fracture healing process by modulating canonical Wnt signaling activity.
Collapse
Affiliation(s)
- Kristen L Lauing
- Alcohol Research Program, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA Burn and Shock Trauma Institute, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA
| | - Sumana Sundaramurthy
- Alcohol Research Program, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA Department of Orthopaedic Surgery and Rehabilitation, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA
| | - Rachel K Nauer
- Burn and Shock Trauma Institute, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA
| | - John J Callaci
- Alcohol Research Program, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA Burn and Shock Trauma Institute, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA Department of Orthopaedic Surgery and Rehabilitation, Loyola University Stritch School of Medicine, 2160 S. 1st Ave, Maywood, IL 60546, USA
| |
Collapse
|
35
|
Heilmann A, Schinke T, Bindl R, Wehner T, Rapp A, Haffner-Luntzer M, Nemitz C, Liedert A, Amling M, Ignatius A. The Wnt serpentine receptor Frizzled-9 regulates new bone formation in fracture healing. PLoS One 2013; 8:e84232. [PMID: 24391920 PMCID: PMC3877253 DOI: 10.1371/journal.pone.0084232] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/21/2013] [Indexed: 12/18/2022] Open
Abstract
Wnt signaling is a key regulator of bone metabolism and fracture healing. The canonical Wnt/β-catenin pathway is regarded as the dominant mechanism, and targeting this pathway has emerged as a promising strategy for the treatment of osteoporosis and poorly healing fractures. In contrast, little is known about the role of non-canonical Wnt signaling in bone. Recently, it was demonstrated that the serpentine receptor Fzd9, a Wnt receptor of the Frizzled family, is essential for osteoblast function and positively regulates bone remodeling via the non-canonical Wnt pathway without involving β-catenin-dependent signaling. Here we investigated whether the Fzd9 receptor is essential for fracture healing using a femur osteotomy model in Fzd9−/− mice. After 10, 24 and 32 days the fracture calli were analyzed using biomechanical testing, histomorphometry, immunohistochemistry, and micro-computed tomography. Our results demonstrated significantly reduced amounts of newly formed bone at all investigated healing time points in the absence of Fzd9 and, accordingly, a decreased mechanical competence of the callus tissue in the late phase of fracture healing. In contrast, cartilage formation and numbers of osteoclasts degrading mineralized matrix were unaltered. β-Catenin immunolocalization showed that canonical Wnt-signaling was not affected in the absence of Fzd9 in osteoblasts as well as in proliferating and mature chondrocytes within the fracture callus. The expression of established differentiation markers was not altered in the absence of Fzd9, whereas chemokines Ccl2 and Cxcl5 seemed to be reduced. Collectively, our results suggest that non-canonical signaling via the Fzd9 receptor positively regulates intramembranous and endochondral bone formation during fracture healing, whereas it does not participate in the formation of cartilage or in the osteoclastic degradation of mineralized matrix. The finding that Fzd9, in addition to its role in physiological bone remodeling, regulates bone repair may have implications for the development of treatments for poorly or non-healing fractures.
Collapse
Affiliation(s)
- Aline Heilmann
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronny Bindl
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Tim Wehner
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Anna Rapp
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Claudia Nemitz
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
36
|
Pountos I, Georgouli T, Pneumaticos S, Giannoudis PV. Fracture non-union: Can biomarkers predict outcome? Injury 2013; 44:1725-32. [PMID: 24075219 DOI: 10.1016/j.injury.2013.09.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/09/2013] [Indexed: 02/02/2023]
Abstract
Delayed bone healing and non-union occurs in approximately 10-15% of long bone fractures. Both pathologies may result in prolonged period of pain, disability and repetitive operative interventions. Despite intense investigations and progress done in understanding the pathophysiologic processes governing bone healing, the diagnostic tools have not been altered. The clinical findings and radiographic features remain the two important landmarks of diagnosing non-union and even when the diagnosis is established there is debate on the ideal timing and mode of intervention. Emerging evidence suggest that there are certain molecules and genes that can serve as predictors of potentially unsuccessful fracture union. This article summarises the current evidence on the available 'bio-markers'to predict fracture non-union.
Collapse
Affiliation(s)
- I Pountos
- Academic Department of Trauma and Orthopaedic Surgery, School of Medicine, University of Leeds, UK; Leeds Biomedical Research Unit, Leeds, UK
| | | | | | | |
Collapse
|
37
|
Lin Z, Fateh A, Salem DM, Intini G. Periosteum: biology and applications in craniofacial bone regeneration. J Dent Res 2013; 93:109-16. [PMID: 24088412 DOI: 10.1177/0022034513506445] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The bone-regenerative potentials of the periosteum have been explored as early as the 17th century. Over the past few years, however, much has been discovered in terms of the molecular and cellular mechanisms that control the periosteal contribution to bone regeneration. Lineage tracing analyses and knock-in transgenic mice have helped define the relative contributions of the periosteum and endosteum to bone regeneration. Additional studies have shed light on the critical roles that BMP, FGF, Hedgehog, Notch, PDGF, Wnt, and inflammation signaling have or may have in periosteal-mediated bone regeneration, fostering the path to novel approaches in bone-regenerative therapy. Thus, by examining the role that each pathway has in periosteal-mediated bone regeneration, in this review we analyze the status of the current research on the regenerative potential of the periosteum. The provided analysis aims to inform both clinician-scientists who may have interest in the current studies about the biology of the periosteum as well as dental surgeons who may find this review useful to perform periosteal-harnessing bone-regenerative procedures.
Collapse
Affiliation(s)
- Z Lin
- Harvard School of Dental Medicine, 188 Longwood Avenue, REB 403, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
38
|
Wang X, Yu YY, Lieu S, Yang F, Lang J, Lu C, Werb Z, Hu D, Miclau T, Marcucio R, Colnot C. MMP9 regulates the cellular response to inflammation after skeletal injury. Bone 2013; 52:111-9. [PMID: 23010105 PMCID: PMC3513654 DOI: 10.1016/j.bone.2012.09.018] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 09/14/2012] [Accepted: 09/15/2012] [Indexed: 11/19/2022]
Abstract
Like other tissue injuries, bone fracture triggers an inflammatory response, which plays an important role in skeletal repair. Inflammation is believed to have both positive and negative effects on bone repair, but the underlying cellular mechanisms are not well understood. To assess the role of inflammation on skeletal cell differentiation, we used mouse models of fracture repair that stimulate either intramembranous or endochondral ossification. In the first model, fractures are rigidly stabilized leading to direct bone formation, while in the second model, fracture instability causes cartilage and bone formation. We compared the inflammatory response in these two mechanical environments and found changes in the expression patterns of inflammatory genes and in the recruitment of inflammatory cells and osteoclasts. These results suggested that the inflammatory response could influence skeletal cell differentiation after fracture. We then exploited matrix metalloproteinase 9 (MMP9) that is expressed in inflammatory cells and osteoclasts, and which we previously showed is a potential regulator of cell fate decisions during fracture repair. Mmp9(-/-) mice heal stabilized fractures via endochondral ossification, while wild type mice heal via intramembranous ossification. In parallel, we observed increases in macrophages and T cells in the callus of Mmp9(-/-) compared to wild type mice. To assess the link between the profile of inflammatory cells and skeletal cell fate functionally, we transplanted Mmp9(-/-) mice with wild type bone marrow, to reconstitute a wild type hematopoietic lineage in interaction with the Mmp9(-/-) stroma and periosteum. Following transplantation, Mmp9(-/-) mice healed stabilized fractures via intramembranous ossification and exhibited a normal profile of inflammatory cells. Moreover, Mmp9(-/-) periosteal grafts healed via intramembranous ossification in wild type hosts, but healed via endochondral ossification in Mmp9(-/-) hosts. We observed that macrophages accumulated at the periosteal surface in Mmp9(-/-) mice, suggesting that cell differentiation in the periosteum is influenced by factors such as BMP2 that are produced locally by inflammatory cells. Taken together, these results show that MMP9 mediates indirect effects on skeletal cell differentiation by regulating the inflammatory response and the distribution of inflammatory cells, leading to the local regulation of periosteal cell differentiation.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Yan Yiu Yu
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Shirley Lieu
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Frank Yang
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Jeffrey Lang
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Chuanyong Lu
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Zena Werb
- Department of Anatomy, University of California at San Francisco, San Francisco, USA
| | - Diane Hu
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
| | - Céline Colnot
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, USA
- INSERM U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
39
|
Jash A, Yun K, Sahoo A, So JS, Im SH. Looping mediated interaction between the promoter and 3' UTR regulates type II collagen expression in chondrocytes. PLoS One 2012; 7:e40828. [PMID: 22815835 PMCID: PMC3397959 DOI: 10.1371/journal.pone.0040828] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 06/13/2012] [Indexed: 11/18/2022] Open
Abstract
Type II collagen is the major component of articular cartilage and is mainly synthesized by chondrocytes. Repeated sub-culturing of primary chondrocytes leads to reduction of type II collagen gene (Col2a1) expression, which mimics the process of chondrocyte dedifferentiation. Although the functional importance of Col2a1 expression has been extensively investigated, mechanism of transcriptional regulation during chondrocyte dedifferentiation is still unclear. In this study, we have investigated the crosstalk between cis-acting DNA element and transcription factor on Col2a1 expression in primary chondrocytes. Bioinformatic analysis revealed the potential regulatory regions in the Col2a1 genomic locus. Among them, promoter and 3′ untranslated region (UTR) showed highly accessible chromatin architecture with enriched recruitment of active chromatin markers in primary chondrocytes. 3′ UTR has a potent enhancer function which recruits Lef1 (Lymphoid enhancer binding factor 1) transcription factor, leading to juxtaposition of the 3′ UTR with the promoter through gene looping resulting in up-regulation of Col2a1 gene transcription. Knock-down of endogenous Lef1 level significantly reduced the gene looping and subsequently down-regulated Col2a1 expression. However, these regulatory loci become inaccessible due to condensed chromatin architecture as chondrocytes dedifferentiate which was accompanied by a reduction of gene looping and down-regulation of Col2a1 expression. Our results indicate that Lef1 mediated looping between promoter and 3′ UTR under the permissive chromatin architecture upregulates Col2a1 expression in primary chondrocytes.
Collapse
Affiliation(s)
- Arijita Jash
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Kangsun Yun
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Anupama Sahoo
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Jae-Seon So
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Sin-Hyeog Im
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
- * E-mail:
| |
Collapse
|
40
|
Lauing KL, Roper PM, Nauer RK, Callaci JJ. Acute alcohol exposure impairs fracture healing and deregulates β-catenin signaling in the fracture callus. Alcohol Clin Exp Res 2012; 36:2095-103. [PMID: 22691115 DOI: 10.1111/j.1530-0277.2012.01830.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 03/11/2012] [Indexed: 01/02/2023]
Abstract
BACKGROUND Alcohol abuse is a risk factor for bone damage and fracture-related complications. Through precise β-catenin signaling, canonical Wnt signaling plays a key role in fracture repair by promoting the differentiation of new bone and cartilage cells. In this study, we examined the effects of alcohol on the Wnt pathway in injured bone using a murine model of alcohol-induced impaired fracture healing. METHODS Male C57Bl/6 or T cell factor (TCF)-transgenic mice were administered 3 daily intraperitoneal doses of alcohol or saline. One hour following the final injection, mice were subjected to a stabilized, mid-shaft tibial fracture. Injured and contralateral tibias were harvested at 6, 9, or 14 days post-fracture for the analysis of biomechanical strength, callus tissue composition, and Wnt/β-catenin signaling. RESULTS Acute alcohol treatment was associated with a significant decrease in fracture callus volume, diameter, and biomechanical strength at day 14 post-fracture. Histology revealed an alcohol-related reduction in cartilage and bone formation at the fracture site, and that alcohol inhibited normal cartilage maturation. Acute alcohol exposure caused a significant 2.3-fold increase in total β-catenin protein at day 6 and a significant decrease of 53 and 56% at days 9 and 14, respectively. lacZ staining in β-galactosidase-expressing TCF-transgenic mice revealed spatial and quantitative differences in Wnt-specific transcriptional activation at day 6 in the alcohol group. Days 9 and 14 post-fracture showed that acute alcohol exposure decreased Wnt transcriptional activation, which correlates with the modulation of total β-catenin protein levels observed at these time points. CONCLUSIONS Acute alcohol exposure resulted in significant impairment of fracture callus tissue formation, perturbation of the key Wnt pathway protein β-catenin, and disruption of normal Wnt-mediated transcription. These data suggest that the canonical Wnt pathway is a target for alcohol in bone and may partially explain why impaired fracture healing is observed in alcohol-abusing individuals.
Collapse
Affiliation(s)
- Kristen L Lauing
- Burn and Shock Trauma Institute, Loyola University Stritch School of Medicine, Maywood, Illinois 60153, USA.
| | | | | | | |
Collapse
|