1
|
King JS, Wan M, Wagley Y, Stestiv M, Kalajzic I, Hankenson KD, Sanjay A. Signaling pathways associated with Lgr6 to regulate osteogenesis. Bone 2024; 187:117207. [PMID: 39033993 DOI: 10.1016/j.bone.2024.117207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Fracture management largely relies on the bone's inherent healing capabilities and, when necessary, surgical intervention. Currently, there are limited osteoinductive therapies to promote healing, making targeting skeletal stem/progenitor cells (SSPCs) a promising avenue for therapeutic development. A limiting factor for this approach is our incomplete understanding of the molecular mechanisms governing SSPCs' behavior. We have recently identified that the Leucine-rich repeat-containing G-protein coupled receptor 6 (Lgr6) is expressed in sub-populations of SSPCs, and is required for maintaining bone volume during adulthood and for proper fracture healing. Lgr family members (Lgr4-6) are markers of stem cell niches and play a role in tissue regeneration primarily by binding R-Spondin (Rspo1-4). This interaction promotes canonical Wnt (cWnt) signaling by stabilizing Frizzled receptors. Interestingly, our findings here indicate that Lgr6 may also influence cWnt-independent pathways. Remarkably, Lgr6 expression was enhanced during Bmp-mediated osteogenesis of both human and murine cells. Using biochemical approaches, RNA sequencing, and bioinformatic analysis of published single-cell data, we found that elements of BMP signaling, including its target gene, pSMAD, and gene ontology pathways, are downregulated in the absence of Lgr6. Our findings uncover a molecular interdependency between the Bmp pathway and Lgr6, offering new insights into osteogenesis and potential targets for enhancing fracture healing.
Collapse
Affiliation(s)
- Justin S King
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Marta Stestiv
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Ivo Kalajzic
- Center for Regenerative Medicine and Skeletal Development, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA.
| |
Collapse
|
2
|
Menger MM, Emmerich M, Scheuer C, Hans S, Ehnert S, Nüssler AK, Herath SC, Steinestel K, Menger MD, Histing T, Laschke MW. Cilostazol Stimulates Angiogenesis and Accelerates Fracture Healing in Aged Male and Female Mice by Increasing the Expression of PI3K and RUNX2. Int J Mol Sci 2024; 25:755. [PMID: 38255829 PMCID: PMC10815626 DOI: 10.3390/ijms25020755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/15/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
Fracture healing in the aged is associated with a reduced healing capacity, which often results in delayed healing or non-union formation. Many factors may contribute to this deterioration of bone regeneration, including a reduced 'angiogenic trauma response'. The phosphodiesterase-3 (PDE-3) inhibitor cilostazol has been shown to exert pro-angiogenic and pro-osteogenic effects in preclinical studies. Therefore, we herein analyzed in a stable closed femoral fracture model whether this compound also promotes fracture healing in aged mice. Forty-two aged CD-1 mice (age: 16-18 months) were daily treated with 30 mg/kg body weight cilostazol (n = 21) or vehicle (control, n = 21) by oral gavage. At 2 and 5 weeks after fracture, the femora were analyzed by X-ray, biomechanics, micro-computed tomography (µCT), histology, immunohistochemistry, and Western blotting. These analyses revealed a significantly increased bending stiffness at 2 weeks (2.2 ± 0.4 vs. 4.3 ± 0.7 N/mm) and an enhanced bone formation at 5 weeks (4.4 ± 0.7 vs. 9.1 ± 0.7 mm3) in cilostazol-treated mice when compared to controls. This was associated with a higher number of newly formed CD31-positive microvessels (3.3 ± 0.9 vs. 5.5 ± 0.7 microvessels/HPF) as well as an elevated expression of phosphoinositide-3-kinase (PI3K) (3.6 ± 0.8 vs. 17.4 ± 5.5-pixel intensity × 104) and runt-related transcription factor (RUNX)2 (6.4 ± 1.2 vs. 18.2 ± 2.7-pixel intensity × 104) within the callus tissue. These findings indicate that cilostazol accelerates fracture healing in aged mice by stimulating angiogenesis and the expression of PI3K and RUNX2. Hence, cilostazol may represent a promising compound to promote bone regeneration in geriatric patients.
Collapse
Affiliation(s)
- Maximilian M. Menger
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Maximilian Emmerich
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Sandra Hans
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Andreas K. Nüssler
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Steven C. Herath
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Konrad Steinestel
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, 89081 Ulm, Germany
| | - Michael D. Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Matthias W. Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
3
|
Menger MM, Tobias AL, Bauer D, Bleimehl M, Scheuer C, Menger MD, Histing T, Laschke MW. Parathyroid hormone stimulates bone regeneration in an atrophic non-union model in aged mice. J Transl Med 2023; 21:844. [PMID: 37996876 PMCID: PMC10668449 DOI: 10.1186/s12967-023-04661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Non-union formation still represents a major burden in trauma and orthopedic surgery. Moreover, aged patients are at an increased risk for bone healing failure. Parathyroid hormone (PTH) has been shown to accelerate fracture healing in young adult animals. However, there is no information whether PTH also stimulates bone regeneration in atrophic non-unions in the aged. Therefore, the aim of the present study was to analyze the effect of PTH on bone regeneration in an atrophic non-union model in aged CD-1 mice. METHODS After creation of a 1.8 mm segmental defect, mice femora were stabilized by pin-clip fixation. The animals were treated daily with either 200 mg/kg body weight PTH 1-34 (n = 17) or saline (control; n = 17) subcutaneously. Bone regeneration was analyzed by means of X-ray, biomechanics, micro-computed tomography (µCT) imaging as well as histological, immunohistochemical and Western blot analyses. RESULTS In PTH-treated animals bone formation was markedly improved when compared to controls. This was associated with an increased bending stiffness as well as a higher number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts and CD31-positive microvessels within the callus tissue. Furthermore, PTH-treated aged animals showed a decreased inflammatory response, characterized by a lower number of MPO-positive granulocytes and CD68-positive macrophages within the bone defects when compared to controls. Additional Western blot analyses demonstrated a significantly higher expression of cyclooxygenase (COX)-2 and phosphoinositide 3-kinase (PI3K) in PTH-treated mice. CONCLUSION Taken together, these findings indicate that PTH is an effective pharmacological compound for the treatment of non-union formation in aged animals.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany.
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany.
| | - Anne L Tobias
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - David Bauer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Michelle Bleimehl
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| |
Collapse
|
4
|
Doherty L, Wan M, Peterson A, Youngstrom DW, King JS, Kalajzic I, Hankenson KD, Sanjay A. Wnt-associated adult stem cell marker Lgr6 is required for osteogenesis and fracture healing. Bone 2023; 169:116681. [PMID: 36708855 PMCID: PMC10015414 DOI: 10.1016/j.bone.2023.116681] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
Despite the remarkable regenerative capacity of skeletal tissues, nonunion of bone and failure of fractures to heal properly presents a significant clinical concern. Stem and progenitor cells are present in bone and become activated following injury; thus, elucidating mechanisms that promote adult stem cell-mediated healing is important. Wnt-associated adult stem marker Lgr6 is implicated in the regeneration of tissues with well-defined stem cell niches in stem cell-reliant organs. Here, we demonstrate that Lgr6 is dynamically expressed in osteoprogenitors in response to fracture injury. We used an Lgr6-null mouse model and found that Lgr6 expression is necessary for maintaining bone volume and efficient postnatal bone regeneration in adult mice. Skeletal progenitors isolated from Lgr6-null mice have reduced colony-forming potential and reduced osteogenic differentiation capacity due to attenuated cWnt signaling. Lgr6-null mice consist of a lower proportion of self-renewing stem cells. In response to fracture injury, Lgr6-null mice have a deficiency in the proliferation of periosteal progenitors and reduced ALP activity. Further, analysis of the bone regeneration phase and remodeling phase of fracture healing in Lgr6-null mice showed impaired endochondral ossification and decreased mineralization. We propose that in contrast to not being required for successful skeletal development, Lgr6-positive cells have a direct role in endochondral bone repair.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA; School of Dental Medicine, UConn Health, Farmington, CT 06030, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Anna Peterson
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Justin S King
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Ivo Kalajzic
- School of Dental Medicine, UConn Health, Farmington, CT 06030, USA; Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmington, CT 06030, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA.
| |
Collapse
|
5
|
Pan Y, Tang Y, Gu H, Ge W. Ubiquitin modification in osteogenic differentiation and bone formation: From mechanisms to clinical significance. Front Cell Dev Biol 2022; 10:1033223. [PMID: 36340031 PMCID: PMC9634082 DOI: 10.3389/fcell.2022.1033223] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/10/2022] [Indexed: 01/03/2024] Open
Abstract
The ubiquitin-proteasome system is an important pathway for mediating posttranslational modification and protein homeostasis and exerts a wide range of functions in diverse biological processes, including stem cell differentiation, DNA repair, and cell cycle regulation. Many studies have shown that ubiquitination modification plays a critical role in regulating the osteogenic differentiation of stem cells and bone formation through various mechanisms. This review summarizes current progress on the effects and mechanisms of ubiquitin modification on transcription factors and signaling pathways involved in osteogenic differentiation. Moreover, the review highlights the latest advances in the clinical application of drugs in bone tissue engineering. A thorough understanding of ubiquitin modifications may provide promising therapeutic targets for stem cell-based bone tissue engineering.
Collapse
Affiliation(s)
- Yuan Pan
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yiman Tang
- Fourth Clinical Division, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Hang Gu
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Wenshu Ge
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
6
|
Bone morphogenetic protein 9 enhances osteogenic and angiogenic responses of human amniotic mesenchymal stem cells cocultured with umbilical vein endothelial cells through the PI3K/AKT/m-TOR signaling pathway. Aging (Albany NY) 2021; 13:24829-24849. [PMID: 34837694 PMCID: PMC8660623 DOI: 10.18632/aging.203718] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/25/2021] [Indexed: 01/06/2023]
Abstract
Background: Neovascularization plays an essential part in bone fracture and defect healing, constructing tissue engineered bone that targets bone regeneration. Bone morphogenetic protein 9 (BMP9) is a regular indicator that potentiates osteogenic and angiogenic differentiation of MSCs. Objectives: To investigate the effects of BMP9 on osteogenesis and angiogenesis of human amniotic mesenchymal stem cells (hAMSCs) cocultured with human umbilical vein endothelial cells (HUVECs) and determine the possible underlying molecular mechanism. Results: The isolated hAMSCs expressed surface markers of MSCs. hAMSCs cocultured with HUVECs enhance osteogenic differentiation and upregulate the expression of angiogenic factors. BMP9 not only potentiates angiogenic signaling of hAMSCs cocultured with HUVECs also increases ectopic bone formation and subcutaneous vessel invasion. Mechanically, the coupling effect between osteogenesis and angiogenesis induced by BMP9 was activated by the BMP/Smad and PI3K/AKT/m-TOR signaling pathways. Conclusions: BMP9-enhanced osteoblastic and angiogenic differentiation in cocultivation with hAMSCs and HUVECs in vitro and in vivo also provide a chance to harness the BMP9-regulated coordinated effect between osteogenic and angiogenic pathways through BMP/Smad and PI3K/AKT/m-TOR signalings. Materials and Methods: The ALP and Alizarin Red S staining assay to determine the effects of osteoblastic differentiation. RT-qPCR and western blot was measured the expression of angiogenesis-related factors. Ectopic bone formation was established and retrieved bony masses were subjected to histochemical staining. The angiogenesis ability and vessel invasion were subsequently determined by immunofluorescence staining. Molecular mechanisms such as the BMP/Smad and PI3K/AKT/m-TOR signaling pathways were detected by ELISA and western blot analysis.
Collapse
|
7
|
Fowlkes JL, Thrailkill KM, Bunn RC. RASopathies: The musculoskeletal consequences and their etiology and pathogenesis. Bone 2021; 152:116060. [PMID: 34144233 PMCID: PMC8316423 DOI: 10.1016/j.bone.2021.116060] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 01/07/2023]
Abstract
The RASopathies comprise an ever-growing number of clinical syndromes resulting from germline mutations in components of the RAS/MAPK signaling pathway. While multiple organs and tissues may be affected by these mutations, this review will focus on how these mutations specifically impact the musculoskeletal system. Herein, we review the genetics and musculoskeletal phenotypes of these syndromes in humans. We discuss how mutations in the RASopathy syndromes have been studied in translational mouse models. Finally, we discuss how signaling molecules within the RAS/MAPK pathway are involved in normal and abnormal bone biology in the context of osteoblasts, osteoclasts and chondrocytes.
Collapse
Affiliation(s)
- John L Fowlkes
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America.
| | - Kathryn M Thrailkill
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - R Clay Bunn
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| |
Collapse
|
8
|
Sun T, Yang D, Wu Y, Sheng Q. The function of microRNA-211 expression in post-fracture bone cell apoptosis involving the transforming growth factor-β/ phosphoinositide 3-kinase signaling pathway. J Int Med Res 2021; 48:300060520926353. [PMID: 32720595 PMCID: PMC7388126 DOI: 10.1177/0300060520926353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background The underlying mechanism of micro (mi)RNA-211 in bone cell apoptosis after fracture
remains unclear. This study aimed to determine the effect and function of miRNA-211 in
bone cell apoptosis in fracture patients. Methods Serum samples were collected from patients with fractures and healthy controls. Serum
miR-211 expression was detected by quantitative PCR. MC3T3-E1 cells were transfected
with a transforming growth factor (TGF)-β inhibitor and phosphoinositide 3-kinase (PI3K)
inhibitor. The viability of MC3T3-E1 cells was detected by the MTT assay, and apoptosis
was detected by flow cytometry. Caspase-3/9 activity and the protein expression of
TGF-β, PI3K, and p-Akt were detected by western blot and immunoprecipitation. Results In the fracture group, miRNA-211 expression was significantly up-regulated compared
with controls. We used miRNA-211 mimics to up-regulate miRNA-211 expression, and
observed inhibited cell viability and induced apoptosis and lactate dehydrogenase (LDH)
activity. miRNA-211 up-regulation also suppressed the expression of TGF-β, PI3K, and
p-Akt proteins. Conversely, miRNA-211 down-regulation increased cell viability and
reduced apoptosis and LDH activity, as well as inducing the expression of TGF-β, PI3K,
and p-Akt. Inhibiting TGF-β decreased the effect of anti-miRNA-211 on osteocyte
apoptosis. Conclusion Our data indicate that miRNA-211 functions via the TGF-β/PI3K/Akt signaling pathway in
patients with fractures.
Collapse
Affiliation(s)
- Tongxin Sun
- Department of Orthopedics, Dongying People's Hospital, Dongying, Shandong province, P. R. China
| | - Dai Yang
- Department of Orthopedics, Dongying People's Hospital, Dongying, Shandong province, P. R. China
| | - Yuanpeng Wu
- Department of Orthopedics, Dongying People's Hospital, Dongying, Shandong province, P. R. China
| | - Qingang Sheng
- Department of Orthopedics, Dongying District People's Hospital, Dongying, Shandong province, P. R. China
| |
Collapse
|
9
|
Doherty L, Wan M, Kalajzic I, Sanjay A. Diabetes impairs periosteal progenitor regenerative potential. Bone 2021; 143:115764. [PMID: 33221502 PMCID: PMC7770068 DOI: 10.1016/j.bone.2020.115764] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
Diabetics are at increased risk for fracture, and experience severely impaired skeletal healing characterized by delayed union or nonunion of the bone. The periosteum harbors osteochondral progenitors that can differentiate into chondrocytes and osteoblasts, and this connective tissue layer is required for efficient fracture healing. While bone marrow-derived stromal cells have been studied extensively in the context of diabetic skeletal repair and osteogenesis, the effect of diabetes on the periosteum and its ability to contribute to bone regeneration has not yet been explicitly evaluated. Within this study, we utilized an established murine model of type I diabetes to evaluate periosteal cell differentiation capacity, proliferation, and availability under the effect of a diabetic environment. Periosteal cells from diabetic mice were deficient in osteogenic differentiation ability in vitro, and diabetic mice had reduced periosteal populations of mesenchymal progenitors with a corresponding reduction in proliferation capacity following injury. Additionally, fracture callus mineralization and mature osteoblast activity during periosteum-mediated healing was impaired in diabetic mice compared to controls. We propose that the effect of diabetes on periosteal progenitors and their ability to aid in skeletal repair directly impairs fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn School of Dental Medicine, Farmington, CT, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| |
Collapse
|
10
|
Hyperhomocysteinemia inhibits tibial fracture healing in rats through PI3K/AKT signaling pathway. Exp Ther Med 2020; 19:2083-2088. [PMID: 32104269 PMCID: PMC7027308 DOI: 10.3892/etm.2020.8412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
To explore the influence of hyperhomocysteinemia (hHcys) on the tibial fracture healing in rats and its effect on the phosphatidylinositol 3-hydroxy kinase (PI3K)/protein kinase B (AKT) signaling pathway. A total of 36 Sprague-Dawley rats were randomly divided into sham group (n=12), tibial fracture group (n=12) and hHcys + fracture group (n=12). The rats in tibial fracture group underwent the tibial fracture surgery, while the model of tibial fracture and hHcys was established in hHcys + fracture group. The level of plasma homocysteine (Hcy) in each group was analyzed using the full-automatic biochemical analyzer, the fracture stress biomechanical measurement was performed, and the ultimate bending strength and torque were calculated. Moreover, the protein expressions of PI3K and phosphorylated (p)-AKT in tibial tissues were detected using western blotting, the messenger ribonucleic acid (mRNA) levels of Bcl-2 associated X protein (Bax) and caspase-3 were detected using quantitative polymerase chain reaction (qPCR), the apoptosis was detected via terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining, and the expressions of inflammatory factors were detected via immunohistochemistry. Compared with sham group, tibial fracture group and hHcys + fracture group had a significantly increased level of plasma Hcy, significantly decreased ultimate bending strength and torque, obviously decreased relative protein expressions of PI3K and p-AKT, increased mRNA levels of Bax and caspase-3 and an increased expression of pro-inflammatory factor tumor necrosis factor-α (TNF-α). Compared with tibial fracture group, hHcys + fracture group had a higher level of plasma Hcy, lower ultimate bending strength and torque, lower relative protein expressions of PI3K and p-AKT, higher mRNA levels of Bax and caspase-3, a higher apoptosis rate and a higher expression of TNF-α. hHcys blocks the downstream apoptotic signal transduction, promotes apoptosis and inflammatory response, and affects fracture healing through affecting the PI3K/AKT signaling pathway.
Collapse
|
11
|
Chen L, Song Z, Wu J, Huang Q, Shen Z, Wei X, Lin Z. LncRNA DANCR sponges miR-216a to inhibit odontoblast differentiation through upregulating c-Cbl. Exp Cell Res 2019; 387:111751. [PMID: 31805275 DOI: 10.1016/j.yexcr.2019.111751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 11/20/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022]
Abstract
Enhanced odontoblast differentiation of human dental pulp cells (hDPCs) is considered a keystone in dentin-pulp complex formation. We have revealed lncRNA DANCR was implicated in this differentiation program, however, its mechanism in odontoblast differentiation of hDPCs remains further explored. In this study, by employing loss-of-function approach, we identified downregulation of DANCR drived odontoblast differentiaion of hDPCs. Bioinformatics analysis was utilized to show that DANCR contained binding site for miR-216a and an inverse correlation between DANCR and miR-216a was obtained. Dual luciferase reporter assay and RNA-binding protein immunoprecipitation (RIP) were applied to further confirm that DANCR conferred its functions by directly binding to miR-216a. Notably, miR-216a was able to bind to the 3'-UTR of c-Cbl and repressed its expression. In addition, the protein level of c-CBL was significantly downregulated during hDPCs differentiation, while c-Cbl overexpression inhibited odontoblast differentiation of hDPCs. Moreover, downregulation of miR-216a efficiently reversed the suppression of c-Cbl level and odontoblast differentiation induced by knockdown of DANCR. Taken together, these analyses indicated that DANCR positively regulated the expression of c-Cbl, through sponging miR-216a, and inhibited odontoblast differentiation of hDPCs. Our results will extend the field of clinical application for cell-based therapy in regenerative medicine.
Collapse
Affiliation(s)
- Lingling Chen
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| | - Zhi Song
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| | - Jinyan Wu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| | - Qiting Huang
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| | - Zongshan Shen
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| | - Xi Wei
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| | - Zhengmei Lin
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong, China.
| |
Collapse
|
12
|
Doherty L, Yu J, Wang X, Hankenson KD, Kalajzic I, Sanjay A. A PDGFRβ-PI3K signaling axis mediates periosteal cell activation during fracture healing. PLoS One 2019; 14:e0223846. [PMID: 31665177 PMCID: PMC6821073 DOI: 10.1371/journal.pone.0223846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022] Open
Abstract
Insufficient and delayed fracture healing remain significant public health problems with limited therapeutic options. Phosphoinositide 3-kinase (PI3K) signaling, a major pathway involved in regulation of fracture healing, promotes proliferation, migration, and differentiation of osteoprogenitors. We have recently reported that knock-in mice with a global increase in PI3K signaling (gCblYF) show enhanced femoral fracture healing characterized by an extraordinary periosteal response to injury. Interestingly, of all growth factor receptors involved in fracture healing, PI3K directly binds only to PDGFR. Given these findings, we hypothesized a PDGFR-PI3K interaction is necessary for mediating robust periosteal cell activation following fracture. In this study, we isolated primary periosteal cells from gCblYF mice to analyze cross-talk between the PDGFRβ and PI3K signaling pathways. We found PDGFRβ signaling contributes to robust Akt phosphorylation in periosteal cells in comparison with other growth factor signaling pathways. Additionally, we performed femoral fractures on gCblYF mice with a conditional removal of PDGFRβ in mesenchymal progenitors using inducible alpha smooth muscle actin (αSMA) CreERT2 mice. Our studies showed that depletion of PDGFRβ signaling within these progenitors in the early phase of fracture healing significantly abrogates PI3K-mediated periosteal activation and proliferation three days after fracture. Combined, these results suggest that PDGFRβ signaling through PI3K is necessary for robust periosteal activation in the earliest phases of fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
| | - Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
| | - Xi Wang
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, United States of America
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, United States of America
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
13
|
Coates BA, McKenzie JA, Buettmann EG, Liu X, Gontarz PM, Zhang B, Silva MJ. Transcriptional profiling of intramembranous and endochondral ossification after fracture in mice. Bone 2019; 127:577-591. [PMID: 31369916 PMCID: PMC6708791 DOI: 10.1016/j.bone.2019.07.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/27/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022]
Abstract
Bone fracture repair represents an important clinical challenge with nearly 1 million non-union fractures occurring annually in the U.S. Gene expression differs between non-union and healthy repair, suggesting there is a pattern of gene expression that is indicative of optimal repair. Despite this, the gene expression profile of fracture repair remains incompletely understood. In this work, we used RNA-seq of two well-established murine fracture models to describe gene expression of intramembranous and endochondral bone formation. We used top differentially expressed genes, enriched gene ontology terms and pathways, callus cellular phenotyping, and histology to describe and contrast these bone formation processes across time. Intramembranous repair, as modeled by ulnar stress fracture, and endochondral repair, as modeled by femur full fracture, exhibited vastly different transcriptional profiles throughout repair. Stress fracture healing had enriched differentially expressed genes associated with bone repair and osteoblasts, highlighting the strong osteogenic repair process of this model. Interestingly, the PI3K-Akt signaling pathway was one of only a few pathways uniquely enriched in stress fracture repair. Full fracture repair involved a higher level of inflammatory and immune cell related genes than did stress fracture repair. Full fracture repair also differed from stress fracture in a robust downregulation of ion channel genes following injury, the role of which in fracture repair is unclear. This study offers a broad description of gene expression in intramembranous and endochondral ossification across several time points throughout repair and suggests several potentially intriguing genes, pathways, and cells whose role in fracture repair requires further study.
Collapse
Affiliation(s)
- Brandon A Coates
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America.
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America
| | - Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America
| | - Xiaochen Liu
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America
| | - Paul M Gontarz
- Department of Developmental Biology, Washington University in St. Louis, MO, United States of America
| | - Bo Zhang
- Department of Developmental Biology, Washington University in St. Louis, MO, United States of America
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America
| |
Collapse
|
14
|
Walia B, Huang AH. Tendon stem progenitor cells: Understanding the biology to inform therapeutic strategies for tendon repair. J Orthop Res 2019; 37:1270-1280. [PMID: 30270569 PMCID: PMC6823601 DOI: 10.1002/jor.24156] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
Tendon and ligament injuries are a leading cause of healthcare visits with significant impact in terms of economic cost and reduced quality of life. To date, reparative strategies remain largely restricted to conservative treatment or surgical repair. However, these therapies fail to restore native tendon structure and function; thus, the tissue may re-rupture or degenerate with time. To improve tendon healing, one promising strategy may be harnessing the innate potential of resident tendon stem/progenitor cells (TSPCs) to guide tenogenic regeneration. In this review, we outline recent advances in the identification and characterization of putative TSPC populations, and discuss biochemical, biomechanical, and biomaterial methods employed for their culture and differentiation. Finally, we identify limitations in our current understanding of TSPC biology, key challenges for their use, and potential therapeutic strategies to inform cell-based tendon repair. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1270-1280, 2019.
Collapse
Affiliation(s)
- Bhavita Walia
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice H. Huang
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
15
|
Adapala NS, Root S, Lorenzo J, Aguila H, Sanjay A. PI3K activation increases SDF-1 production and number of osteoclast precursors, and enhances SDF-1-mediated osteoclast precursor migration. Bone Rep 2019; 10:100203. [PMID: 30989092 PMCID: PMC6449702 DOI: 10.1016/j.bonr.2019.100203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/26/2019] [Accepted: 03/19/2019] [Indexed: 01/07/2023] Open
Abstract
Our previous studies showed that in a mouse model in which PI3K-AKT activation was increased (YF mice), osteoclast numbers and levels of SDF-1, a chemokine, were augmented. The purpose of this study was to delineate the role of PI3K activation in regulating SDF-1 production and examine whether SDF-1 can stimulate differentiation and/or migration of osteoclast precursors. Using flow cytometric analysis, we demonstrated that compared to wild type mice, bone marrow of YF mice had increased numbers of CXCL12 abundant reticular (CAR) cells, that are a major cell type responsible for producing SDF-1. At the molecular level, transcription factor specificity protein 1 (Sp1) induced an increased transcription of SDF-1 that was dependent on PI3K/AKT activation. YF mice also contained an increased number of osteoclast precursors, in which expression of CXCR4, a major receptor for SDF-1, was increased. SDF-1 did not induce differentiation of osteoclast precursors into mature osteoclasts; compared to cells derived from WT mice, cells obtained from YF mice were more responsive to SDF-1. In conclusion, we demonstrate that PI3K activation resulted in increased SDF-1, increased the number of osteoclast precursors, and enhanced osteoclast precursor migration in response to SDF-1. PI3K activation regulates the number of CAR cells in mouse bone marrow. PI3K activation regulates SDF-1/CXCL12 production by CAR cells in bone marrow. PI3K/AKT activation mediates transcription of SDF-1 by regulating transcription factor Sp1. SDF-1 enhances migration of osteoclast precursors via CXCR4.
Collapse
Affiliation(s)
- Naga Suresh Adapala
- Department of Orthopaedic Surgery, Farmington, CT, USA.,U Conn Health, Farmington, CT, USA
| | - Sierra Root
- Department of Immunology, Farmington, CT, USA.,U Conn Health, Farmington, CT, USA
| | - Joseph Lorenzo
- Department of Endocrinology and Metabolism, Farmington, CT, USA.,U Conn Health, Farmington, CT, USA
| | - Hector Aguila
- Department of Immunology, Farmington, CT, USA.,U Conn Health, Farmington, CT, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, Farmington, CT, USA.,U Conn Health, Farmington, CT, USA
| |
Collapse
|
16
|
Wu R, Ruan J, Sun Y, Liu M, Sha Z, Fan C, Wu Q. Long non-coding RNA HIF1A-AS2 facilitates adipose-derived stem cells (ASCs) osteogenic differentiation through miR-665/IL6 axis via PI3K/Akt signaling pathway. Stem Cell Res Ther 2018; 9:348. [PMID: 30545407 PMCID: PMC6293597 DOI: 10.1186/s13287-018-1082-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/10/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Background This study was aimed to investigate the role and specific molecular mechanism of HIF1A-AS2/miR-665/IL6 axis in regulating osteogenic differentiation of adipose-derived stem cells (ASCs) via the PI3K/Akt signaling pathway. Methods RNAs’ expression profile in normal/osteogenic differentiation-induced ASCs (osteogenic group) was from the Gene Expression Omnibus database. The analysis was carried out using Bioconductor of R. Gene Set Enrichment Analysis and Kyoto Encyclopedia of Genes and Genomes dataset were applied to identify up- and downregulated signaling pathways. Co-expression network of specific lncRNAs and mRNAs was structured by Cytoscape, while binding sites amongst lncRNA, mRNA, and miRNA were predicted by TargetScan and miRanda. ASCs were derived from human adipose tissue and were authenticated by flow cytometry. ASC cell function was surveyed by alizarin red and alkaline phosphatase (ALP) staining. Molecular mechanism of HIF1A-AS2/miR-665/IL6 axis was investigated by RNAi, cell transfection, western blot, and qRT-PCR. RNA target relationships were validated by dual-luciferase assay. Results HIF1A-AS2 and IL6 were highly expressed while miR-665 was lowly expressed in induced ASCs. HIF1A-AS2 and IL6 improved the expression level of osteoblast markers Runx2, Osterix, and Osteocalcin and also accelerated the formation of calcium nodule and ALP activity, yet miR-665 had opposite effects. HIF1A-AS2 directly targeted miR-665, whereas miR-665 repressed IL6 expression. Moreover, the HIF1A-AS2/miR-665/IL6 regulating axis activated the PI3K/Akt signaling pathway. Conclusions LncRNA HIF1A-AS2 could sponge miR-665 and hence upregulate IL6, activate the PI3K/Akt signaling pathway, and ultimately promote ASC osteogenic differentiation. Electronic supplementary material The online version of this article (10.1186/s13287-018-1082-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ruoyu Wu
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jihao Ruan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Yongjin Sun
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Mengyu Liu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Zhuang Sha
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China.
| | - Qingkai Wu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Xuhui District, Shanghai, 200233, China. .,Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
17
|
Deubiquitinating Enzymes and Bone Remodeling. Stem Cells Int 2018; 2018:3712083. [PMID: 30123285 PMCID: PMC6079350 DOI: 10.1155/2018/3712083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/29/2018] [Indexed: 02/05/2023] Open
Abstract
Bone remodeling, which is essential for bone homeostasis, is controlled by multiple factors and mechanisms. In the past few years, studies have emphasized the role of the ubiquitin-dependent proteolysis system in regulating bone remodeling. Deubiquitinases, which are grouped into five families, remove ubiquitin from target proteins and are involved in several cell functions. Importantly, a number of deubiquitinases mediate bone remodeling through regulating differentiation and/or function of osteoblast and osteoclasts. In this review, we review the functions and mechanisms of deubiquitinases in mediating bone remodeling.
Collapse
|
18
|
Walia B, Lingenheld E, Duong L, Sanjay A, Drissi H. A novel role for cathepsin K in periosteal osteoclast precursors during fracture repair. Ann N Y Acad Sci 2018; 1415:57-68. [PMID: 29479711 DOI: 10.1111/nyas.13629] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 11/29/2022]
Abstract
Osteoporosis management is currently centered around bisphosphonates, which inhibit osteoclast (OC) bone resorption but do not affect bone formation. This reduces fracture risk, but fails to restore healthy bone remodeling. Studies in animal models showed that cathepsin K (CatK) inhibition by genetic deletion or chemical inhibitors maintained bone formation while abrogating resorption during bone remodeling and stimulated periosteal bone modeling. Recently, periosteal mononuclear tartrate-resistant acid phosphatase-positive (TRAP+ ) osteoclast precursors (OCPs) were shown to augment angiogenesis-coupled osteogenesis. CatK gene deletion increased osteoblast differentiation via enhanced OCP and OC secretion of platelet-derived growth factor (PDGF)-BB and sphingosine 1 phosphate. The effects of periosteum-derived OCPs on bone remodeling are unknown, particularly with regard to fracture repair. We hypothesized that periosteal OCPs derived from CatK-null (Ctsk-/- ) mice may enhance periosteal bone formation during fracture repair. We found fewer periosteal OCPs in Ctsk-/- mice under homeostatic conditions; however, after fracture, this population increased in number relative to that seen in wild-type (WT) mice. Enhanced TRAP staining and greater expression of PDGF-BB were observed in fractured Ctsk-/- femurs relative to WT femurs. This early pattern of augmented PDGF-BB expression in Ctsk-/- mice may contribute to improved fracture healing by enhancing callus mineralization in Ctsk-/- mice.
Collapse
Affiliation(s)
- Bhavita Walia
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut
| | | | - Le Duong
- Bone Biology Group, Merck Research Laboratories, West Point, Pennsylvania
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
19
|
陈 燕, 陈 良, 李 正, 兰 泽. [Osteogenic effect of collagen/bioglass composites carrying noggin siRNA]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:106-111. [PMID: 33177016 PMCID: PMC6765610 DOI: 10.3969/j.issn.1673-4254.2018.01.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate osteogenic effect of collagen/bioglass composites loaded with a small interfering RNA (siRNA) targeting noggin. METHODS The collagen/bioglass composites loaded with the negative control siRNA or noggin siRNA were prepared by freeze-drying method. CCK8 test was used to evaluate the proliferation of MC3T3 cells exposed to the aqueous extracts of collagen/bioglass composites and the siRNA-loaded collagen/bioglass composites. ALP activity assay, quantitative real-time PCR and Alizarin Red staining were used to assess the effect of the 3 composites on mineralization in MC3T3 cells. RESULTS MC3T3 cells cultured for 3 and 5 days in the presence of the extracts of the 3 composites all showed significantly more active proliferation than the blank control cells (P < 0.05). Compared with the cells seeded on the scaffold without siRNA, MC3T3 cells seeded on collagen/bioglass scaffold loaded with noggin siRNA showed a significantly higher ALP activity at 14 days after seeding (P < 0.05) with significantly increased expression of ALP, Runx2 and BSP mRNAs (P < 0.05). Alizarin Red staining showed that the cells seeded on the noggin siRNA-loading collagen/bioglass scaffold contained significantly more mineralized nodules than the other cells (P < 0.05). CONCLUSIONS The collagen/bioglass composites loaded with noggin siRNA have a good biocompatibility, and the collagen/bioglass composites and noggin siRNA show a synergistic effect in promoting osteogenesis.
Collapse
Affiliation(s)
- 燕玲 陈
- 福建医科大学附属口腔医院正畸科,福建 福州 350004Department of Orthodontics, Stomatological Hospital Affiliated to Fujian Medical University, Fuzhou 350004, China
| | - 良娇 陈
- 广州医科大学附属口腔医院//广州口腔病研究所//口腔医学重点实验室,广东 广州 510140Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 510140, China
| | - 正茂 李
- 广州医科大学附属口腔医院//广州口腔病研究所//口腔医学重点实验室,广东 广州 510140Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 510140, China
| | - 泽栋 兰
- 南方医科大学深圳口腔医院,广东 深圳 518000Shenzhen Stomatological Hospital Affiliated to Southern Medical University, Shenzhen 518000, China
| |
Collapse
|