1
|
Hamada H, Takashima K, Higuchi R, Ando W, Sugano N, Uemura K. Which Radiographic View Visualizes the Initial Collapse in Osteonecrosis of the Femoral Head? A Computed Tomography Scan Study. J Arthroplasty 2025:S0883-5403(25)00586-8. [PMID: 40414370 DOI: 10.1016/j.arth.2025.05.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/16/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND An accurate diagnosis for staging osteonecrosis of the femoral head (ONFH), particularly in early-stage collapse, is essential for determining therapeutic strategies. Various radiographic views in different femoral positions have been utilized to detect femoral head collapse. However, previous studies have not established the optimal femoral position that can sensitively detect initial collapse on plain radiography. This study aimed to identify the most sensitive radiographic view for visualizing collapse in early-stage ONFH by analyzing reconstructed frontal images of the femoral head at multiple femoral positions on computed tomography (CT). METHODS This study included 30 hips with early-stage ONFH (10 hips without collapse and 20 hips with collapse [< three mm] based on the antero-posterior and lateral radiographic images). The presence or absence of collapse in 10 reconstructed frontal images of the femoral head on CT scans, corresponding to 10 different femoral positions, was classified. Furthermore, the ability to detect collapse in each image was compared. RESULTS The reconstructed frontal image of the femoral head on CT scans at 45° flexion and 20° abduction had the highest sensitivity for detecting collapse among the analyzed positions. Hence, it had a significantly greater sensitivity than the neutral position (86 versus 53%, P < 0.01). Of 70% who did not present with collapse on plain radiography had collapse on the reconstructed frontal image at 45° flexion and 20° abduction. CONCLUSION The plain radiographic image taken at 45° flexion and 20° abduction, referred to as the 45° Dunn view, had a greater diagnostic potential for early collapse in ONFH compared with the antero-posterior radiographic image. Nevertheless, further research should be performed to comprehensively investigate the areas where collapse occurs in ONFH and to identify the most effective femoral position for detecting collapse on plain radiography.
Collapse
Affiliation(s)
- Hidetoshi Hamada
- Department of Orthopaedic Medical Engineering, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Kazuma Takashima
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Higuchi
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Wataru Ando
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Nobuhiko Sugano
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keisuke Uemura
- Department of Orthopaedic Medical Engineering, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
2
|
Shinohara I, Tsubosaka M, Toya M, Lee ML, Kushioka J, Murayama M, Gao Q, Li X, Zhang N, Chow SKH, Matsumoto T, Kuroda R, Goodman SB. C-C Motif Chemokine Ligand 2 Enhances Macrophage Chemotaxis, Osteogenesis, and Angiogenesis during the Inflammatory Phase of Bone Regeneration. Biomolecules 2023; 13:1665. [PMID: 38002347 PMCID: PMC10669364 DOI: 10.3390/biom13111665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Local cell therapy has recently gained attention for the treatment of joint diseases and fractures. Mesenchymal stem cells (MSCs) are not only involved in osteogenesis and angiogenesis, but they also have immunomodulatory functions, such as inducing macrophage migration during bone regeneration via macrophage crosstalk. C-C motif chemokine ligand 2 (CCL2), a known inflammatory mediator, is associated with the migration of macrophages during inflammation. This study examined the utility of CCL2 as a therapeutic target for local cell therapy. Using lentiviral vectors for rabbit MSCs, genetically modified CCL2 overexpressing MSCs were generated. Osteogenic differentiation assays were performed using MSCs with or without macrophages in co-culture, and cell migration assays were also performed. Additionally, co-cultures were performed with endothelial cells (ECs), and angiogenesis was evaluated using a tube formation assay. Overexpression of CCL2 did not affect bone formation under monoculture conditions but promoted chemotaxis and osteogenesis when co-cultured with macrophages. Furthermore, CCL2-overexpression promoted tube formation in co-culture with ECs. These results suggest that CCL2 induces macrophage chemotaxis and osteogenesis by promoting crosstalk between MSCs and macrophages; CCL2 also stimulates ECs to induce angiogenesis. These findings indicate that CCL2 may be a useful therapeutic target for local cell therapy in areas of bone loss.
Collapse
Affiliation(s)
- Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (T.M.); (R.K.)
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (T.M.); (R.K.)
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
| | - Max L. Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
| | - Ning Zhang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong;
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (T.M.); (R.K.)
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (T.M.); (R.K.)
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94063, USA; (I.S.); (M.T.); (M.T.); (M.L.L.); (J.K.); (M.M.); (Q.G.); (X.L.); (S.K.-H.C.)
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
3
|
Tsubosaka M, Maruyama M, Lui E, Moeinzadeh S, Huang EE, Kushioka J, Hirata H, Jain C, Storaci HW, Chan C, Toya M, Gao Q, Teissier V, Shen H, Li X, Zhang N, Matsumoto T, Kuroda R, Goodman SB, Yang YP. The efficiency of genetically modified mesenchymal stromal cells combined with a functionally graded scaffold for bone regeneration in corticosteroid-induced osteonecrosis of the femoral head in rabbits. J Biomed Mater Res A 2023; 111:1120-1134. [PMID: 36606330 PMCID: PMC10277231 DOI: 10.1002/jbm.a.37495] [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: 09/07/2022] [Revised: 12/18/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
Core decompression (CD) with mesenchymal stromal cells (MSCs) is an effective therapy for early-stage osteonecrosis of the femoral head (ONFH). Preconditioning of MSCs, using inflammatory mediators, is widely used in immunology and various cell therapies. We developed a three-dimensional printed functionally graded scaffold (FGS), made of β-TCP and PCL, for cell delivery at a specific location. The present study examined the efficacy of CD treatments with genetically modified (GM) MSCs over-expressing PDGF-BB (PDGF-MSCs) or GM MSCs co-over-expressing IL-4 and PDGF-BB and preconditioned for three days of exposure to lipopolysaccharide and tumor necrosis factor-alpha (IL-4-PDGF-pMSCs) using the FGS for treating steroid-induced ONFH in rabbits. We compared CD without cell-therapy, with IL-4-PDGF-pMSCs alone, and with FGS loaded with PDGF-MSCs or IL-4-PDGF-pMSCs. For the area inside the CD, the bone volume in the CD alone was higher than in both FGS groups. The IL-4-PDGF-pMSCs alone and FGS + PDGF-MSCs reduced the occurrence of empty lacunae and improved osteoclastogenesis. There was no significant difference in angiogenesis among the four groups. The combined effect of GM MSCs or pMSCs and the FGS was not superior to the effect of each alone. To establish an important adjunctive therapy for CD for early ONFH in the future, it is necessary and essential to develop an FGS that delivers biologics appropriately and provides structural and mechanical support.
Collapse
Affiliation(s)
- Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Charu Jain
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hunter W. Storaci
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin Chan
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Victoria Teissier
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Material Science and Engineering, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
4
|
Quan H, Ren C, He Y, Wang F, Dong S, Jiang H. Application of Biomaterials in Treating Early Osteonecrosis of the Femoral Head: Research Progress and Future Perspectives. Acta Biomater 2023; 164:15-73. [PMID: 37080444 DOI: 10.1016/j.actbio.2023.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/24/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023]
Abstract
Osteonecrosis of the femoral head (ONFH), a progressive pathological process of femoral head ischemia and osteocyte necrosis, is a refractory orthopedic disease caused by multiple etiologies and there is no complete cure at present. With the extension of ONFH duration, osteocyte apoptosis and trabecular bone loss can decrease the load-bearing capacity of the femoral head, which leads to the collapse of the articular cartilage and subchondral bone. Therefore, an urgent clinical need exists to develop effective treatment strategies of early-stage ONFH for maintaining the hip joint function and preventing femoral head collapse. In recent years, extensive attention has been paid to the application of diverse biomaterials in treating early ONFH for sustaining the normal morphology and function of the autologous femoral head, and slowing disease progression. Herein, we review the research progress of bone grafts, metallic materials, bioceramics, bioglasses and polymer materials for early ONFH treatment, and discuss the biological mechanisms of bone repair and regeneration in the femoral-head necrotic area. We propose suggestions for future research directions, from a special perspective of improving the local microenvironment in femoral head by facilitating vessel-associated osteoclasts (VAOs) generation and coupling of bone-specific angiogenesis and osteogenesis, as well as inhibiting bone-associated osteoclasts (BAOs) and BAO-mediated bone resorption. This review can provide ideas for the research, development, and clinical application of biomaterials for treating early ONFH. STATEMENT OF SIGNIFICANCE: We believe that at least three aspects of this manuscript make it interesting to readers of the Acta Biomaterialia. First, we briefly summarize the incidence, pathogenesis, risk factors, classification criteria and treatment of early osteonecrosis of the femoral head (ONFH). Second, we review the research progress in biomaterials for early ONFH treatment and the biological mechanisms of bone repair and regeneration in femoral-head necrotic area. Third, we propose future research progress on improving the local microenvironment in femoral head by facilitating vessel-associated osteoclasts generation and coupling of bone-specific angiogenesis and osteogenesis, as well as inhibiting bone-associated osteoclasts and bone resorption. We hope this review can provide ideas for the research, development, and clinical application of biomaterials for treating early ONFH.
Collapse
Affiliation(s)
- Hongyu Quan
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China; College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chencan Ren
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China; College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yuwei He
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China
| | - Fuyou Wang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China; State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China.
| | - Hong Jiang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
5
|
Hernigou J, Verdonk P, Homma Y, Verdonk R, Goodman SB, Hernigou P. Nonoperative and Operative Bone and Cartilage Regeneration and Orthopaedic Biologics of the Hip: An Orthoregeneration Network (ON) Foundation Hip Review. Arthroscopy 2022; 38:643-656. [PMID: 34506886 DOI: 10.1016/j.arthro.2021.08.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/12/2021] [Indexed: 02/02/2023]
Abstract
Orthoregeneration is defined as a solution for orthopaedic conditions that harnesses the benefits of biology to improve healing, reduce pain, improve function, and, optimally, provide an environment for tissue regeneration. Options include drugs, surgical intervention, scaffolds, biologics as a product of cells, and physical and electromagnetic stimuli. The goal of regenerative medicine is to enhance the healing of tissue after musculoskeletal injuries as both isolated treatment and adjunct to surgical management, using novel therapies to improve recovery and outcomes. Various orthopaedic biologics (orthobiologics) have been investigated for the treatment of pathology involving the hip, including osteonecrosis (aseptic necrosis) involving bone marrow, bone, and cartilage, and chondral injuries involving articular cartilage, synovium, and bone marrow. Promising and established treatment modalities for osteonecrosis include nonweightbearing; pharmacological treatments including low molecular-weight heparin, prostacyclin, statins, bisphosphonates, and denosumab, a receptor activator of nuclear factor-kB ligand inhibitor; extracorporeal shock wave therapy; pulsed electromagnetic fields; core decompression surgery; cellular therapies including bone marrow aspirate comprising mesenchymal stromal cells (MSCs aka mesenchymal stem cells) and bone marrow autologous concentrate, with or without expanded or cultured cells, and possible addition of bone morphogenetic protein-2, vascular endothelial growth factor, and basic fibroblast growth factor; and arterial perfusion of MSCs that may be combined with addition of carriers or scaffolds including autologous MSCs cultured with beta-tricalcium phosphate ceramics associated with a free vascularized fibula. Promising and established treatment modalities for chondral lesions include autologous platelet-rich plasma; hyaluronic acid; MSCs (in expanded or nonexpanded form) derived from bone marrow or other sources such as fat, placenta, umbilical cord blood, synovial membrane, and cartilage; microfracture or microfracture augmented with membrane containing MSCs, collagen, HA, or synthetic polymer; mosaicplasty; 1-stage autologous cartilage translation (ACT) or 2-stage ACT using 3-dimensional spheroids; and autologous cartilage grafting; chondral flap repair, or flap fixation with fibrin glue. Hip pain is catastrophic in young patients, and promising therapies offer an alternative to premature arthroplasty. This may address both physical and psychological components of pain; the goal is to avoid or postpone an artificial joint. LEVEL OF EVIDENCE: Level V, expert opinion.
Collapse
Affiliation(s)
| | | | - Yasuhiro Homma
- Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - René Verdonk
- Department of Orthopaedics & Trauma, ULB University Clinic Erasme, Brussels, Belgium
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University Medical Center Outpatient Center, Stanford, California, U.S.A
| | | |
Collapse
|
6
|
Tsubosaka M, Maruyama M, Huang EE, Zhang N, Utsunomiya T, Gao Q, Shen H, Li X, Kushioka J, Hirata H, Yao Z, Yang YP, Goodman SB. Effect on Osteogenic Differentiation of Genetically Modified IL4 or PDGF-BB Over-Expressing and IL4-PDGF-BB Co-Over-Expressing Bone Marrow-Derived Mesenchymal Stromal Cells In Vitro. Bioengineering (Basel) 2021; 8:bioengineering8110165. [PMID: 34821731 PMCID: PMC8614682 DOI: 10.3390/bioengineering8110165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022] Open
Abstract
The use of genetically modified (GM) mesenchymal stromal cells (MSCs) and preconditioned MSCs (pMSCs) may provide further opportunities to improve the outcome of core decompression (CD) for the treatment of early-stage osteonecrosis of the femoral head (ONFH). GM interleukin-4 (IL4) over-expressing MSCs (IL4-MSCs), platelet-derived growth factor (PDGF)-BB over-expressing MSCs (PDGF-BB-MSCs), and IL4-PDGF-BB co-over-expressing MSCs (IL4-PDGF-BB-MSCs) and their respective pMSCs were used in this in vitro study and compared with respect to cell proliferation and osteogenic differentiation. IL4-MSCs, PDGF-BB-MSCs, IL4-PDGF-BB-MSCs, and each pMSC treatment significantly increased cell proliferation compared to the MSC group alone. The percentage of Alizarin red-stained area in the IL4-MSC and IL4-pMSC groups was significantly lower than in the MSC group. However, the percentage of Alizarin red-stained area in the PDGF-BB-MSC group was significantly higher than in the MSC and PDGF-BB-pMSC groups. The percentage of Alizarin red-stained area in the IL4-PDGF-BB-pMSC was significantly higher than in the IL4-PDGF-BB-MSC group. There were no significant differences in the percentage of Alizarin red-stained area between the MSC and IL4-PDGF-BB-pMSC groups. The use of PDGF-BB-MSCs or IL4-PDGF-BB-pMSCs increased cell proliferation. Furthermore, PDGF-BB-MSCs promoted osteogenic differentiation. The addition of GM MSCs may provide a useful supplementary cell-based therapy to CD for treatment of ONFH.
Collapse
Affiliation(s)
- Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
- Department of Material Science and Engineering, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063, USA; (M.T.); (M.M.); (E.E.H.); (N.Z.); (T.U.); (Q.G.); (H.S.); (X.L.); (J.K.); (H.H.); (Z.Y.); (Y.P.Y.)
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
- Correspondence: ; Tel.: +1-650-498-4343
| |
Collapse
|
7
|
Guzman RA, Maruyama M, Moeinzadeh S, Lui E, Zhang N, Storaci HW, Tam K, Huang EE, Utsunomiya T, Rhee C, Gao Q, Yao Z, Yang YP, Goodman SB. The effect of genetically modified platelet-derived growth factor-BB over-expressing mesenchymal stromal cells during core decompression for steroid-associated osteonecrosis of the femoral head in rabbits. Stem Cell Res Ther 2021; 12:503. [PMID: 34526115 PMCID: PMC8444495 DOI: 10.1186/s13287-021-02572-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/27/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Approximately one third of patients undergoing core decompression (CD) for early-stage osteonecrosis of the femoral head (ONFH) experience progression of the disease, and subsequently require total hip arthroplasty (THA). Thus, identifying adjunctive treatments to optimize bone regeneration during CD is an unmet clinical need. Platelet-derived growth factor (PDGF)-BB plays a central role in cell growth and differentiation. The aim of this study was to characterize mesenchymal stromal cells (MSCs) that were genetically modified to overexpress PDGF-BB (PDGF-BB-MSCs) in vitro and evaluate their therapeutic effect when injected into the bone tunnel at the time of CD in an in vivo rabbit model of steroid-associated ONFH. METHODS In vitro studies: Rabbit MSCs were transduced with a lentivirus vector carrying the human PDGF-BB gene under the control of either the cytomegalovirus (CMV) or phosphoglycerate (PGK) promoter. The proliferative rate, PDGF-BB expression level, and osteogenic differentiation capacity of unmodified MSCs, CMV-PDGF-BB-MSCs, and PGK-PDGF-BB-MSCs were assessed. In vivo studies: Twenty-four male New Zealand white rabbits received an intramuscular (IM) injection of methylprednisolone 20 mg/kg. Four weeks later, the rabbits were divided into four groups: the CD group, the hydrogel [HG, (a collagen-alginate mixture)] group, the MSC group, and the PGK-PDGF-BB-MSC group. Eight weeks later, the rabbits were sacrificed, their femurs were harvested, and microCT, mechanical testing, and histological analyses were performed. RESULTS In vitro studies: PGK-PDGF-BB-MSCs proliferated more rapidly than unmodified MSCs (P < 0.001) and CMV-PDGF-BB-MSCs (P < 0.05) at days 3 and 7. CMV-PDGF-BB-MSCs demonstrated greater PDGF-BB expression than PGK-PDGF-BB-MSCs (P < 0.01). However, PGK-PDGF-BB-MSCs exhibited greater alkaline phosphatase staining at 14 days (P < 0.01), and osteogenic differentiation at 28 days (P = 0.07) than CMV-PDGF-BB-MSCs. In vivo: The PGK-PDGF-BB-MSC group had a trend towards greater bone mineral density (BMD) than the CD group (P = 0.074). The PGK-PDGF-BB-MSC group demonstrated significantly lower numbers of empty lacunae (P < 0.001), greater osteoclast density (P < 0.01), and greater angiogenesis (P < 0.01) than the other treatment groups. CONCLUSION The use of PGK-PDGF-BB-MSCs as an adjunctive treatment with CD may reduce progression of osteonecrosis and enhance bone regeneration and angiogenesis in the treatment of early-stage ONFH.
Collapse
Affiliation(s)
- Roberto Alfonso Guzman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA.,Department of Mechanical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Hunter W Storaci
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Kaysie Tam
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA. .,Department of Material Science and Engineering, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA. .,Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA.
| |
Collapse
|
8
|
Li T, Liu Y, Zhang Q, Sun W, Dong Y. A steroid-induced osteonecrosis model established using an organ-on-a-chip platform. Exp Ther Med 2021; 22:1070. [PMID: 34447463 PMCID: PMC8355687 DOI: 10.3892/etm.2021.10504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
Bone microvascular endothelial cells (BMECs) constitute the central part of the femoral head's intramural microenvironment network and have an essential role in the development of steroid-induced osteonecrosis of the femoral head. Recently, the rapid development of microfluidic technology has led to innovations in the fields of chemistry, medicine and life sciences. It is now possible to use microfluidics organ-on-a-chip techniques to assess osteonecrosis. In the present study, BMECs were cultured on a microfluidic organ-on-a-chip platform to explore the pathogenesis of femoral-head necrosis. The aim of the present study was to explore the effects of different interventions on BMECs and study the pathogenesis of steroid-induced osteonecrosis through a microfluidic organ-on-a-chip platform. Methods including SU-8 lithography were used to produce a microfluidic organ-on-a-chip and human umbilical vein endothelial cells (HUVECs) were used to test whether it was possible to culture cells on the chip. Subsequently, a set of methods were applied for the isolation, purification, culture and identification of BMECs. Hydroxyapatite (HA) was used for co-culture, dexamethasone was used at different concentrations as an intervention in the cells and icariin was used for protection. BMECs were isolated and cultured from the femoral head obtained following total hip arthroplasty and were then inoculated into the microfluidic organ-on-a-chip for further treatment. In part I of the experiment, HUVECs and BMECs both successfully survived on the chip and a comparison of the growth and morphology was performed. HA and BMECs were then co-cultured for comparison with the control group. The cell growth was observed by confocal microscopy after 24 h. In part II, the effects of different concentrations of glucocorticoid (0.4 or 0.6 mg/ml dexamethasone) and the protection of icariin were evaluated. The morphology of BMECs and the cleaved caspase-3/7 content were observed by immunofluorescence staining and confocal microscopy after 24 h. In the microfluidic organ-on-a-chip, the response of the cells was able to be accurately observed. In part I, at the same concentration of injected cells, BMECs exhibited improved viability compared with HUVECs (P<0.05). In addition, it was indicated that HA was not only able to promote the germination and growth of BMECs but also improve the survival of the cells (P<0.05). In part II, it was identified that dexamethasone was able to induce BMECs to produce cleaved caspase 3/7; the caspase 3/7 content was significantly higher than that in the blank control group (P<0.05) and a dose correlation was observed. Icariin was able to inhibit this process and protect the microvascular structure of BMECs. The content of cleaved caspase 3/7 in the icariin-protected group was significantly lower than that in the group without icariin (P<0.05). It was concluded that BMECs are more likely to survive than HUVECs and HA promoted the growth of BMECs on the microfluidic organ-on-a-chip platform. Glucocorticoid caused damage to BMECs through the production of cleaved caspase 3/7, which was observed through the microfluidic organ-on-a-chip platform, and icariin protected BMECs from damage.
Collapse
Affiliation(s)
- Tengqi Li
- Department of Orthopedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, P.R. China
| | - Yadi Liu
- Department of Bioanalysis, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Qingyu Zhang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Wei Sun
- Department of Orthopedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, P.R. China.,Department of Orthopedic Surgery, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Yiyang Dong
- Department of Bioanalysis, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| |
Collapse
|
9
|
Maruyama M, Lin T, Kaminow NI, Thio T, Storaci HW, Pan CC, Yao Z, Takagi M, Goodman SB, Yang YP. The efficacy of core decompression for steroid-associated osteonecrosis of the femoral head in rabbits. J Orthop Res 2021; 39:1441-1451. [PMID: 33095462 PMCID: PMC8204476 DOI: 10.1002/jor.24888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/10/2020] [Accepted: 10/21/2020] [Indexed: 02/04/2023]
Abstract
Although core decompression (CD) is often performed in the early stage of osteonecrosis of the femoral head (ONFH), the procedure does not always prevent subsequent deterioration and the effects of CD are not fully clarified. The aim of this study is to evaluate the efficacy of CD for steroid-associated ONFH in rabbits. Twelve male and 12 female New Zealand rabbits were injected intramuscularly 20 mg/kg of methylprednisolone once and were divided into the disease control and CD groups. In the disease control group, rabbits had no treatment and were euthanized at 12 weeks postinjection. In the CD group, rabbits underwent left femoral CD at 4 weeks postinjection and were euthanized 8 weeks postoperatively. The left femurs were collected to perform morphological, biomechanical, and histological analysis. Bone mineral density and bone volume fraction in the femoral head in the CD group were significantly higher than in the disease control group. However, no difference in the mechanical strength was observed between the two groups. Histological analysis showed that alkaline phosphatase and CD31 positive cells significantly increased in the males after CD treatment. The number of empty lacunae in the surrounding trabecular bone was significantly higher in the CD group. The current study indicated that CD improved the morphological properties, but did not improve the mechanical strength in the femoral head at early-stage ONFH. These data suggest the need for additional biological, mechanical strategies, and therapeutic windows to improve the outcome of early-stage steroid-associated ONFH.
Collapse
Affiliation(s)
- Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Nicolas I. Kaminow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Thio
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Hunter W. Storaci
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Chi-Chun Pan
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA,Department of Mechanical Engineering, Stanford University School of Medicine, Stanford, California, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA,Department of Bioengineering, Stanford University School of Engineering, Stanford, California, USA
| | - Yunzhi P. Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA,Department of Bioengineering, Stanford University School of Engineering, Stanford, California, USA,Department of Material Science and Engineering, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
10
|
Maruyama M, Moeinzadeh S, Guzman RA, Zhang N, Storaci HW, Utsunomiya T, Lui E, Huang EE, Rhee C, Gao Q, Yao Z, Takagi M, Yang YP, Goodman SB. The efficacy of lapine preconditioned or genetically modified IL4 over-expressing bone marrow-derived mesenchymal stromal cells in corticosteroid-associated osteonecrosis of the femoral head in rabbits. Biomaterials 2021; 275:120972. [PMID: 34186237 DOI: 10.1016/j.biomaterials.2021.120972] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/29/2021] [Accepted: 06/10/2021] [Indexed: 02/09/2023]
Abstract
Cell-based therapy for augmentation of core decompression (CD) using mesenchymal stromal cells (MSCs) is a promising treatment for early stage osteonecrosis of the femoral head (ONFH). Recently, the therapeutic potential for immunomodulation of osteogenesis using preconditioned (with pro-inflammatory cytokines) MSCs (pMSCs), or by the timely resolution of inflammation using MSCs that over-express anti-inflammatory cytokines has been described. Here, pMSCs exposed to tumor necrosis factor-alpha and lipopolysaccharide for 3 days accelerated osteogenic differentiation in vitro. Furthermore, injection of pMSCs encapsulated with injectable hydrogels into the bone tunnel facilitated angiogenesis and osteogenesis in the femoral head in vivo, using rabbit bone marrow-derived MSCs and a model of corticosteroid-associated ONFH in rabbits. In contrast, in vitro and in vivo studies demonstrated that genetically-modified MSCs that over-express IL4 (IL4-MSCs), established by using a lentiviral vector carrying the rabbit IL4 gene under the cytomegalovirus promoter, accelerated proliferation of MSCs and decreased the percentage of empty lacunae in the femoral head. Therefore, adjunctive cell-based therapy of CD using pMSCs and IL4-MSCs may hold promise to heal osteonecrotic lesions in the early stage ONFH. These interventions must be applied in a temporally sensitive fashion, without interfering with the mandatory acute inflammatory phase of bone healing.
Collapse
Affiliation(s)
- Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberto Alfonso Guzman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hunter W Storaci
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Mechanical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Material Science and Engineering, Stanford University School of Medicine, Stanford, CA, USA; Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Kuroda Y, Tanaka T, Miyagawa T, Hamada H, Abe H, Ito-Ihara T, Asada R, Fujimoto Y, Takahashi D, Tetsunaga T, Kaneuji A, Takagi M, Inaba Y, Morita S, Sugano N, Tanaka S, Matsuda S, Akiyama H. Recombinant human FGF-2 for the treatment of early-stage osteonecrosis of the femoral head: TRION, a single-arm, multicenter, Phase II trial. Regen Med 2021; 16:535-548. [PMID: 34075804 DOI: 10.2217/rme-2021-0049] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: This study aimed to evaluate the 2-year outcomes from a clinical trial of recombinant human FGF-2 (rhFGF-2) for osteonecrosis of the femoral head (ONFH). Patients & methods: Sixty-four patients with nontraumatic, precollapse and large ONFHs were percutaneously administered with 800 μg rhFGF-2 contained in gelatin hydrogel. Setting the end point of radiological collapse, we analyzed the joint preservation period of the historical control. Changes in two validated clinical scores, bone regeneration and safety were evaluated. Results: Radiological joint preservation time was significantly higher in the rhFGF-2 group than in the control group. The ONFHs tended to improve to smaller ONFHs. The postoperative clinical scores significantly improved. Thirteen serious adverse events showed recovery. Conclusion: rhFGF-2 treatment increases joint preservation time with clinical efficacy, radiological bone regeneration and safety.
Collapse
Affiliation(s)
- Yutaka Kuroda
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, Shogoin, Kawahara-cho 54, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takeyuki Tanaka
- Department of Orthopedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Takaki Miyagawa
- Department of Orthopedic Surgery, Gifu University, Gifu, 501-1194, Japan
| | - Hidetoshi Hamada
- Department of Orthopedic Surgery, Osaka University, Osaka, 565-0871, Japan
| | - Hiroyasu Abe
- Department of Biomedical Statistics & Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Toshiko Ito-Ihara
- The Clinical & Translational Research Center, Kyoto Prefectural University of Medicine, Kyoto, 602-0841, Japan
| | - Ryuta Asada
- Innovative & Clinical Research Promotion Center, Graduate School of Medicine, Gifu University, Gifu, 501-1194, Japan
| | - Yusuke Fujimoto
- Department of Medical Joint Materials, Graduate School of Medical & Dental Sciences, Kagoshima University, Kagoshima, 890-8520, Japan
| | - Daisuke Takahashi
- Department of Orthopedic Surgery, Faculty of Medicine & Graduate School of Medicine, Hokkaido University, Hokkaido, 060-8648, Japan
| | - Tomonori Tetsunaga
- Department of Orthopedic Surgery, Okayama University, Okayama, 700-0914, Japan
| | - Ayumi Kaneuji
- Department of Orthopedic Surgery, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Michiaki Takagi
- Department of Orthopedic Surgery, Yamagata University Faculty of Medicine, Yamagata, 990-2331, Japan
| | - Yutaka Inaba
- Department of Orthopedic Surgery, Yokohama City University, Kanagawa, 236-0004, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics & Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Nobuhiko Sugano
- Department of Orthopedic Medical Engineering, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Sakae Tanaka
- Department of Orthopedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Shuichi Matsuda
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, Shogoin, Kawahara-cho 54, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Haruhiko Akiyama
- Department of Orthopedic Surgery, Gifu University, Gifu, 501-1194, Japan
| | | |
Collapse
|
12
|
Kuroda Y, Okuzu Y, Kawai T, Goto K, Matsuda S. Difference in Therapeutic Strategies for Joint-Preserving Surgery for Non-Traumatic Osteonecrosis of the Femoral Head between the United States and Japan: A Review of the Literature. Orthop Surg 2021; 13:742-748. [PMID: 33821555 PMCID: PMC8126904 DOI: 10.1111/os.12979] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/05/2021] [Accepted: 02/02/2021] [Indexed: 01/18/2023] Open
Abstract
For patients with non-traumatic osteonecrosis of the femoral head (ONFH), core decompression (CD) and bone grafts (BG) are mainly performed in the West, while osteotomy is found to be predominant in Japan. It is not well recognized how the surgical procedures for joint preservation in patients with ONFH are completely different between the United States and Japan. This paper identifies the contexts and the differences in treatment strategies for ONFH between the two countries. We compared the surgical trends of the two countries over three periods, 1997-2001, 2002-2006, and 2007-2011 (the US data for the third period was 2007-2008), based on a 2014 US paper and a 2013 national publication in Japan. We compared the details of surgery for non-traumatic ONFH under the same conditions in the two reports. For the period 1997-2001, the rates of surgeries for ONFH in the US were as follows: total hip arthroplasty (THA), 86%; CD, 10%; and osteotomy, 0.4%. In Japan, THA was 61%, osteotomy 38%, and CD 0%. For the recent period, 2007-2011 (US 2007-2008), the rate of THA was 91%, CD 6%, and osteotomy 0.1%, in the US, compared to a THA rate of 73%, CD 0%, and osteotomy 26% in Japan. The results for the interim period (2002-2006) were between the old and new data. The use of joint-preserving surgery for ONFH differs greatly between the US and Japan. The first-line joint-preserving surgery was CD in the US and osteotomy in Japan. Each procedure was rarely done in the other country. From about 2000 to 2010, the percentage of THA increased in both countries. The proportion of joint-preserving surgery (CD in the US and osteotomy in Japan) declined. The decrease in joint-preserving procedures may be largely attributed to improved long-term outcomes of THA due to technological advances. There is also a reluctance for young ONFH patients to undergo joint-preserving procedures, such as osteotomy, that require long-term hospitalization.
Collapse
Affiliation(s)
- Yutaka Kuroda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yaichiro Okuzu
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshiyuki Kawai
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Goto
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
13
|
Maruyama M, Pan CC, Moeinzadeh S, Storaci HW, Guzman RA, Lui E, Ueno M, Utsunomiya T, Zhang N, Rhee C, Yao Z, Takagi M, Goodman SB, Yang YP. Effect of porosity of a functionally-graded scaffold for the treatment of corticosteroid-associated osteonecrosis of the femoral head in rabbits. J Orthop Translat 2021; 28:90-99. [PMID: 33816112 PMCID: PMC7995660 DOI: 10.1016/j.jot.2021.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/24/2020] [Accepted: 01/07/2021] [Indexed: 11/24/2022] Open
Abstract
Background/Objective: Core decompression (CD) with scaffold and cell-based therapies is a promising strategy for providing both mechanical support and regeneration of the osteonecrotic area for early stage osteonecrosis of the femoral head (ONFH). We designed a new 3D printed porous functionally-graded scaffold (FGS) with a central channel to facilitate delivery of transplanted cells in a hydrogel to the osteonecrotic area. However, the optimal porous structural design for the FGS for the engineering of bone in ONFH has not been elucidated. The aim of this study was to fabricate and evaluate two different porous structures (30% or 60% porosity) of the FGSs in corticosteroid-associated ONFH in rabbits. Methods Two different FGSs with 30% or 60% porosity containing a 1-mm central channel were 3D printed using polycaprolactone and β-tricalcium phosphate. The FGS was 3-mm diameter and 32-mm length and was composed of three segments: 1-mm in length for the non-porous proximal segment, 22-mm in length for the porous (30% versus 60%) middle segment, and 9-mm in length for the 15% porous distal segment. Eighteen male New Zealand White rabbits were given a single dose of 20 mg/kg methylprednisolone acetate intramuscularly. Four weeks later, rabbits were divided into three groups: the CD group, the 30% porosity FGS group, and the 60% porosity FGS group. In the CD group, a 3-mm diameter drill hole was created into the left femoral head. In the FGS groups, a 30% or 60% porosity implant was inserted into the bone tunnel. Eight weeks postoperatively, femurs were harvested and microCT, mechanical, and histological analyses were performed. Results The actual porosity and pore size of the middle segments were 26.4% ± 2.3% and 699 ± 56 μm in the 30% porosity FGS, and 56.0% ± 4.5% and 999 ± 71 μm in the 60% porosity FGS, respectively using microCT analysis. Bone ingrowth ratio in the 30% porosity FGS group was 73.9% ± 15.8%, which was significantly higher than 39.5% ± 13.0% in the CD group on microCT (p < 0.05). Bone ingrowth ratio in the 60% porosity FGS group (61.3% ± 30.1%) showed no significant differences compared to the other two groups. The stiffness at the bone tunnel site in the 30% porosity FGS group was 582.4 ± 192.3 N/mm3, which was significantly higher than 338.7 ± 164.6 N/mm3 in the 60% porosity FGS group during push-out testing (p < 0.05). Hematoxylin and eosin staining exhibited thick and mature trabecular bone around the porous FGS in the 30% porosity FGS group, whereas thinner, more immature trabecular bone was seen around the porous FGS in the 60% porosity FGS group. Conclusion These findings indicate that the 30% porosity FGS may enhance bone regeneration and have superior biomechanical properties in the bone tunnel after CD in ONFH, compared to the 60% porosity FGS. Translation potential statement The translational potential of this article: This FGS implant holds promise for improving outcomes of CD for early stage ONFH.
Collapse
Affiliation(s)
- Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Chi-Chun Pan
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, CA, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hunter W Storaci
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberto Alfonso Guzman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, CA, USA
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Material Science and Engineering, Stanford University School of Medicine, Stanford, CA, USA.,Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|