1
|
Dan-Jumbo SO, Riley SE, Cortes-Araya Y, Ho W, Lee S, Thrower T, Esteves CL, Donadeu FX. Derivation and long-term maintenance of porcine skeletal muscle progenitor cells. Sci Rep 2024; 14:9370. [PMID: 38653980 DOI: 10.1038/s41598-024-59767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Culture of muscle cells from livestock species has typically involved laborious enzyme-based approaches that yield heterogeneous populations with limited proliferative and myogenic differentiation capacity, thus limiting their use in physiologically-meaningful studies. This study reports the use of a simple explant culture technique to derive progenitor cell populations from porcine muscle that could be maintained and differentiated long-term in culture. Fragments of semitendinosus muscle from 4 to 8 week-old piglets (n = 4) were seeded on matrigel coated culture dishes to stimulate migration of muscle-derived progenitor cells (MDPCs). Cell outgrowths appeared within a few days and were serially passaged and characterised using RT-qPCR, immunostaining and flow cytometry. MDPCs had an initial mean doubling time of 1.4 days which increased to 2.5 days by passage 14. MDPC populations displayed steady levels of the lineage-specific markers, PAX7 and MYOD, up until at least passage 2 (positive immunostaining in about 40% cells for each gene), after which the expression of myogenic markers decreased gradually. Remarkably, MDPCs were able to readily generate myotubes in culture up until passage 8. Moreover, a decrease in myogenic capacity during serial passaging was concomitant with a gradual increase in the expression of the pre-adipocyte markers, CD105 and PDGFRA, and an increase in the ability of MDPCs to differentiate into adipocytes. In conclusion, explant culture provided a simple and efficient method to harvest enriched myogenic progenitors from pig skeletal muscle which could be maintained long-term and differentiated in vitro, thus providing a suitable system for studies on porcine muscle biology and applications in the expanding field of cultured meat.
Collapse
Affiliation(s)
- Susan O Dan-Jumbo
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Susanna E Riley
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Yennifer Cortes-Araya
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - William Ho
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Seungmee Lee
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Thomas Thrower
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Cristina L Esteves
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - F Xavier Donadeu
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK.
| |
Collapse
|
2
|
Tu B, Yu B, Wang W, Li J, Yuan F, Zhu J, Fan C. Inhibition of IL-17 prevents the progression of traumatic heterotopic ossification. J Cell Mol Med 2021; 25:7709-7719. [PMID: 34189826 PMCID: PMC8358870 DOI: 10.1111/jcmm.16617] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/05/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022] Open
Abstract
Traumatic heterotopic ossification (HO) is the abnormal formation of bone in soft tissues as a consequence of injury. However, the pathological mechanisms leading to traumatic HO remain unknown. Here, we report that aberrant expression of IL-17 promotes traumatic HO formation by activating β-catenin signalling in mouse model. We found that elevated IL-17 and β-catenin levels are correlated with a high degree of HO formation in specimens from patients and HO animals. We also show that IL-17 initiates and promotes HO progression in mice. Local injection of an IL-17 neutralizing antibody attenuates ectopic bone formation in a traumatic mouse model. IL-17 enhances the osteoblastic differentiation of mesenchymal stem cells (MSCs) by activating β-catenin signalling. Moreover, inhibition of IL-17R or β-catenin signalling by neutralizing antibodies or drugs prevents the osteogenic differentiation of isolated MSCs and decreases HO formation in mouse models. Together, our study identifies a novel role for active IL-17 as the inducer and promoter of ectopic bone formation and suggests that IL-17 inhibition might be a potential therapeutic target in traumatic HO.
Collapse
Affiliation(s)
- Bing Tu
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bo Yu
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Wang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Yuan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jing Zhu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
3
|
MyD88 Is Not Required for Muscle Injury-Induced Endochondral Heterotopic Ossification in a Mouse Model of Fibrodysplasia Ossificans Progressiva. Biomedicines 2021; 9:biomedicines9060630. [PMID: 34206078 PMCID: PMC8227787 DOI: 10.3390/biomedicines9060630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 01/09/2023] Open
Abstract
Excess inflammation and canonical BMP receptor (BMPR) signaling are coinciding hallmarks of the early stages of injury-induced endochondral heterotopic ossification (EHO), especially in the rare genetic disease fibrodysplasia ossificans progressiva (FOP). Multiple inflammatory signaling pathways can synergistically enhance BMP-induced Smad1/5/8 activity in multiple cell types, suggesting the importance of pathway crosstalk in EHO and FOP. Toll-like receptors (TLRs) and IL-1 receptors mediate many of the earliest injury-induced inflammatory signals largely via MyD88-dependent pathways. Thus, the hypothesis that MyD88-dependent signaling is required for EHO was tested in vitro and in vivo using global or Pdgfrα-conditional deletion of MyD88 in FOP mice. As expected, IL-1β or LPS synergistically increased Activin A (ActA)-induced phosphorylation of Smad 1/5 in fibroadipoprogenitors (FAPs) expressing Alk2R206H. However, conditional deletion of MyD88 in Pdgfrα-positive cells of FOP mice did not significantly alter the amount of muscle injury-induced EHO. Even more surprisingly, injury-induced EHO was not significantly affected by global deletion of MyD88. These studies demonstrate that MyD88-dependent signaling is dispensable for injury-induced EHO in FOP mice.
Collapse
|
4
|
Jann J, Drevelle O, Lauzon MA, Faucheux N. Adhesion, intracellular signalling and osteogenic differentiation of mesenchymal progenitor cells and preosteoblasts on poly(epsilon)caprolactone films functionalized by peptides derived from fibronectin and/or BMP-9. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:111088. [DOI: 10.1016/j.msec.2020.111088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/14/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022]
|
5
|
Goloviznina NA, Xie N, Dandapat A, Iaizzo PA, Kyba M. Prospective isolation of human fibroadipogenic progenitors with CD73. Heliyon 2020; 6:e04503. [PMID: 32728644 PMCID: PMC7381701 DOI: 10.1016/j.heliyon.2020.e04503] [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] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/17/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle relies on coordination between myogenic and non-myogenic interstitial cells for homeostasis and for regeneration and response to injury. Fibroadipogenic progenitors (FAPs) have recently been recognized as key modulators of signaling to promote myogenesis following injury. FAPs are also responsible for the fibrosis and fatty replacement of muscle tissue seen in many diseased states. While extensive use of surface markers to purify FAPs has been undertaken in the mouse system, in particular PDGFRA, markers for human FAPs are less well understood. Here, we show that CD73 can be used as a single positive marker to purify FAPs from the lineage-negative (CD45-neg, CD31-neg) fraction of skeletal muscle mononuclear cells. Although CD73 was previously found to be expressed in cultured myogenic cells, we find that this marker is only acquired upon culture and that the CD73+ fraction of human skeletal muscle has no myogenic activity. We show that Lin-neg CD73+ cells from human muscle undergo fat differentiation as well as fibrogenesis when exposed to appropriate activating signals in vitro. This simple single positive marker approach effectively enables isolation of human FAPs from fresh human skeletal muscle biopsies.
Collapse
Affiliation(s)
- Natalya A Goloviznina
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ning Xie
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Abhijit Dandapat
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Paul A Iaizzo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
6
|
Zhao Y, Ouyang N, Chen L, Zhao H, Shen G, Dai J. Stimulating Factors and Origins of Precursor Cells in Traumatic Heterotopic Ossification Around the Temporomandibular Joint in Mice. Front Cell Dev Biol 2020; 8:445. [PMID: 32626707 PMCID: PMC7314999 DOI: 10.3389/fcell.2020.00445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
The contributing factors and the origins of precursor cells in traumatic heterotopic ossification around the temporomandibular joint (THO-TMJ), which causes obvious restriction of mouth opening and maxillofacial malformation, remain unclear. In this study, our findings demonstrated that injured chondrocytes in the condylar cartilage, but not osteoblasts in the injured subchondral bone, played definite roles in the development of THO-TMJ in mice. Injured condylar chondrocytes without articular disc reserves might secrete growth factors, such as IGF1 and TGFβ2, that stimulate precursor cells, such as endothelial cells and muscle-derived cells, to differentiate into chondrocytes or osteoblasts and induce THO-TMJ. Preserved articular discs can alleviate the pressure on the injured cartilage and inhibit the development of THO-TMJ by inhibiting the secretion of these growth factors from injured chondrocytes. However, the exact molecular relationships among trauma, the injured condylar cartilage, growth factors such as TGFβ2, and pressure need to be explored in detail in the future.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Oral & Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Disease, Shanghai, China
| | - Ningjuan Ouyang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Chen
- Department of Oral & Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Disease, Shanghai, China
| | - Hanjiang Zhao
- Department of Oral & Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Disease, Shanghai, China
| | - Guofang Shen
- Department of Oral & Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Disease, Shanghai, China
| | - Jiewen Dai
- Department of Oral & Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, National Clinical Research Center for Oral Disease, Shanghai, China
| |
Collapse
|
7
|
Zhang J, Song GY, Feng H. Development of heterotopic ossification after multiple-ligament reconstruction of the knee joint: Incidence and explanatory factor analysis. Knee 2020; 27:642-648. [PMID: 32563418 DOI: 10.1016/j.knee.2020.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/02/2020] [Accepted: 04/14/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND The development of heterotopic ossification (HO) might nullify any benefit of multiple-ligament reconstruction of the knee joint. The purpose of this study was to investigate the incidence and the specific explanatory factors for the development of HO after multiple-ligament reconstruction of the knee joint. METHODS From January 2011 to June 2016, 72 consecutive patients with knee dislocations received multiple-ligament reconstructions, of which 57 (79%) were available for a minimum follow up of 12 months and were included in this study. Anteroposterior (AP) and lateral radiographs were reviewed for all patients. This knee dislocation cohort was separated into two groups based on the presence or absence of HO for comparisons. In addition, the HO group was divided into three subgroups based on a modified quadrant grading system introduced by the senior author for further evaluation. Multivariate logistic regression analysis was then performed to identify specific explanatory factors predicting development of HO in patients after multiple-ligament reconstructions of the knee joint. RESULTS Among the 72 consecutive patients, 57 (79%) were available for the clinical evaluations with an average period of 28.4 months (range, 12-51 months). Twenty-one patients (37%) showed radiological evidence of HO. The HO group (n = 21) showed significantly inferior results of knee flexion angle compared with the non-HO group (n = 36) (HO group vs. non-HO group: 124 ± 13° vs. 132 ± 5°; P<0.01). According to the quadrant grading system, there were seven patients with grade I, nine with grade II, and five with grade III HO. Subgroup analysis further revealed that higher HO grade would lead to lower knee flexion angle. In addition, multivariate regression analysis showed that concomitant posterior cruciate ligament reconstruction was the only independent explanatory factor predicting the development of HO after multiple-ligament reconstruction of the knee joint (P=0.018; odds ratio, 8.75; 95% confidence interval, 1.69-39.7). CONCLUSION In this cohort of knee dislocations, the incidence of HO development following multiple-ligament reconstruction was 37%, with grade III HO showing the most inferior range of motion outcome. Moreover, concomitant posterior cruciate ligament reconstruction was the only independent predictor for the development of HO.
Collapse
Affiliation(s)
- Jin Zhang
- Sports Medicine Service, Beijing Jishuitan Hospital, Beijing, China
| | - Guan-Yang Song
- Sports Medicine Service, Beijing Jishuitan Hospital, Beijing, China
| | - Hua Feng
- Sports Medicine Service, Beijing Jishuitan Hospital, Beijing, China.
| |
Collapse
|
8
|
Both Human Hematoma Punctured from Pelvic Fractures and Serum Increase Muscle Resident Stem Cells Response to BMP9: A Multivariate Statistical Approach. J Clin Med 2020; 9:jcm9041175. [PMID: 32325892 PMCID: PMC7231246 DOI: 10.3390/jcm9041175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Hematoma and skeletal muscles play a crucial role in bone fracture healing. The muscle resident mesenchymal stromal cells (mrSCs) can promote bone formation by differentiating into osteoblasts upon treatment by bone morphogenetic proteins (BMP), such as BMP9. However, the influence of hematoma fracture extracts (Hema) on human mrSC (hmrSC) response to BMP9 is still unknown. We therefore determined the influence of Hema, human healthy serum (HH), and fetal bovine serum (FBS, control) on BMP9-induced osteoblast commitment of hmrSC by measuring alkaline phosphatase activity. Multiplex assays of 90 cytokines were performed to characterize HH and Hema composition and allow their classification by a multivariate statistical approach depending on their expression levels. We confirmed that BMP9 had a greater effect on osteoblastic differentiation of hmrSCs than BMP2 in presence of FBS. The hmrSCs response to BMP9 was enhanced by both Hema and HH, even though several cytokines were upregulated (IL-6, IL-8, MCP-1, VEGF-A and osteopontin), downregulated (BMP9, PDGF) or similar (TNF-alpha) in Hema compared with HH. Thus, hematoma may potentiate BMP9-induced osteogenic differentiation of hmrSCs during bone fracture healing. The multivariate statistical analyses will help to identify the cytokines involved in such phenomenon leading to normal or pathological bone healing.
Collapse
|
9
|
Characterization and therapeutic applications of mesenchymal stem cells for regenerative medicine. Tissue Cell 2020; 64:101330. [PMID: 32473704 DOI: 10.1016/j.tice.2020.101330] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/04/2020] [Accepted: 01/05/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent, genomic stable, self-renewable, and culturally expandable adult stem cells. MSCs facilitate tissue development, maintenance and repair, and produce secretory factors that support engraftment and trophic functions, marking them an attractive option in cell therapy, regenerative medicine and tissue engineering. METHOD In this review, we summarize the recent researches regarding the isolation and characterization of MSCs, therapeutic applications and advanced engineering techniques. We also discuss the advantages and limitations that remain to be overcome for MSCs based therapy. RESULTS It has been demonstrated that MSCs are able to modulate endogenous tissue and immune cells. Preclinical studies and early phase clinical trials have shown their great potential for tissue engineering of bone, cartilage, marrow stroma, muscle, fat, and other connective tissues. CONCLUSIONS MSC-based therapy show considerable promise to rebuild damaged or diseased tissues, which could be a promising therapeutic method for regeneration medicine.
Collapse
|
10
|
Zhao Y, Liu P, Chen Q, Ouyang N, Lin Y, Zhang W, Dai J, Shen G. Development process of traumatic heterotopic ossification of the temporomandibular joint in mice. J Craniomaxillofac Surg 2019; 47:1155-1161. [DOI: 10.1016/j.jcms.2018.11.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/22/2018] [Accepted: 11/28/2018] [Indexed: 10/27/2022] Open
|
11
|
Wasnik S, Lakhan R, Baylink DJ, Rundle CH, Xu Y, Zhang J, Qin X, Lau KHW, Carreon EE, Tang X. Cyclooxygenase 2 augments osteoblastic but suppresses chondrocytic differentiation of CD90 + skeletal stem cells in fracture sites. SCIENCE ADVANCES 2019; 5:eaaw2108. [PMID: 31392271 PMCID: PMC6669009 DOI: 10.1126/sciadv.aaw2108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/21/2019] [Indexed: 05/07/2023]
Abstract
Cyclooxygenase 2 (COX-2) is essential for normal tissue repair. Although COX-2 is known to enhance the differentiation of mesenchymal stem cells (MSCs), how COX-2 regulates MSC differentiation into different tissue-specific progenitors to promote tissue repair remains unknown. Because it has been shown that COX-2 is critical for normal bone repair and local COX-2 overexpression in fracture sites accelerates fracture repair, this study aimed to determine the MSC subsets that are targeted by COX-2. We showed that CD90+ mouse skeletal stem cells (mSSCs; i.e., CD45-Tie2-AlphaV+ MSCs) were selectively recruited by macrophage/monocyte chemoattractant protein 1 into fracture sites following local COX-2 overexpression. In addition, local COX-2 overexpression augmented osteoblast differentiation and suppressed chondrocyte differentiation in CD90+ mSSCs, which depended on canonical WNT signaling. CD90 depletion data demonstrated that local COX-2 overexpression targeted CD90+ mSSCs to accelerate fracture repair. In conclusion, CD90+ mSSCs are promising targets for the acceleration of bone repair.
Collapse
Affiliation(s)
- Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Ram Lakhan
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Charles H. Rundle
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA, USA
| | - Yi Xu
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jintao Zhang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Henan, China
| | - Xuezhong Qin
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA, USA
| | - Kin-Hing William Lau
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA, USA
| | - Edmundo E. Carreon
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Xiaolei Tang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Corresponding author.
| |
Collapse
|
12
|
Drouin G, Couture V, Lauzon MA, Balg F, Faucheux N, Grenier G. Muscle injury-induced hypoxia alters the proliferation and differentiation potentials of muscle resident stromal cells. Skelet Muscle 2019; 9:18. [PMID: 31217019 PMCID: PMC6582603 DOI: 10.1186/s13395-019-0202-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Trauma-induced heterotopic ossification (HO) is a complication that develops under three conditions: the presence of an osteogenic progenitor cell, an inducing factor, and a permissive environment. We previously showed that a mouse multipotent Sca1+ CD31− Lin− muscle resident stromal cell (mrSC) population is involved in the development of HO in the presence of inducing factors, members of the bone morphogenetic protein family. Interestingly, BMP9 unlike BMP2 causes HO only if the muscle is damaged by injection of cardiotoxin. Because acute trauma often results in blood vessel breakdown, we hypothesized that a hypoxic state in damaged muscles may foster mrSCs activation and proliferation and trigger differentiation toward an osteogenic lineage, thus promoting the development of HO. Methods Three- to - six-month-old male C57Bl/6 mice were used to induce muscle damage by injection of cardiotoxin intramuscularly into the tibialis anterior and gastrocnemius muscles. mrSCs were isolated from damaged (hypoxic state) and contralateral healthy muscles and counted, and their osteoblastic differentiation with or without BMP2 and BMP9 was determined by alkaline phosphatase activity measurement. The proliferation and differentiation of mrSCs isolated from healthy muscles was also studied in normoxic incubator and hypoxic conditions. The effect of hypoxia on BMP synthesis and Smad pathway activation was determined by qPCR and/or Western blot analyses. Differences between normally distributed groups were compared using a Student’s paired t test or an unpaired t test. Results The hypoxic state of a severely damaged muscle increased the proliferation and osteogenic differentiation of mrSCs. mrSCs isolated from damaged muscles also displayed greater sensitivity to osteogenic signals, especially BMP9, than did mrSCs from a healthy muscle. In hypoxic conditions, mrSCs isolated from a control muscle were more proliferative and were more prone to osteogenic differentiation. Interestingly, Smad1/5/8 activation was detected in hypoxic conditions and was still present after 5 days, while Smad1/5/8 phosphorylation could not be detected after 3 h of normoxic incubator condition. BMP9 mRNA transcripts and protein levels were higher in mrSCs cultured in hypoxic conditions. Our results suggest that low-oxygen levels in damaged muscle influence mrSC behavior by facilitating their differentiation into osteoblasts. This effect may be mediated partly through the activation of the Smad pathway and the expression of osteoinductive growth factors such as BMP9 by mrSCs. Conclusion Hypoxia should be considered a key factor in the microenvironment of damaged muscle that triggers HO. Electronic supplementary material The online version of this article (10.1186/s13395-019-0202-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Geneviève Drouin
- Centre de Recherche du CHUS, 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada.,Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C8, Canada
| | - Vanessa Couture
- Centre de Recherche du CHUS, 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Marc-Antoine Lauzon
- Laboratory of 3D Cell Culture Systems, Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul Universite, Sherbrooke, QC, J1K 2R1, Canada
| | - Frédéric Balg
- Centre de Recherche du CHUS, 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada.,Department of Orthopedic Surgery, Faculty of Medicine, Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Nathalie Faucheux
- Centre de Recherche du CHUS, 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada. .,Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul Universite, Sherbrooke, QC, J1K 2R1, Canada.
| | - Guillaume Grenier
- Centre de Recherche du CHUS, 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada.,Department of Orthopedic Surgery, Faculty of Medicine, Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| |
Collapse
|
13
|
Zhang L, Xu J, Fu S, Qin B, Liu Y, Yang Y, Wang M, Li D, Zhong S, Huang W. Distribution and Morphological Measurement of Bony Spurs on the Coracoid Process in a Chinese Population. Med Sci Monit 2019; 25:2527-2534. [PMID: 30953435 PMCID: PMC6463619 DOI: 10.12659/msm.913658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background There are few studies on distributions or morphological measurements for bony spurs form at the attachment points of the ligaments and tendons on the coracoid process. The aim of this study was to investigate their most common sites and morphological characteristics, and to propose possible reasons. Material/Methods Scapulae with bony spurs on the coracoid process were selected from 377 intact and dry Chinese scapulae. The distribution, height, and transverse and longitudinal diameter of the bony spurs were measured in each coracoid process. Results We selected 71 scapulae, 36 left and 35 right, that had bony spurs, from 377 scapulae. The bony spurs were most commonly located at the attachment point of the superior transverse scapular ligament (STSL) (31, 23.66%), while the trapezoid ligament (TL) accounted for the smaller proportion (8, 6.11%). The TSL was the highest, with the minimum transverse and longitudinal diameter, while the TL had the greatest transverse and longitudinal diameters. Only the TSL and TL had a statistically significant difference between the left and the right bony spur regarding the longitudinal diameter (P<0.05). Conclusions Bony spurs are more likely to form at the attachment points of ligaments and tendons on the coracoid process, which has a greater risk of traction injuries or attachment points avulsion fractures.
Collapse
Affiliation(s)
- Lei Zhang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China (mainland).,Academician Workstation in Luzhou, Luzhou, Sichuan, China (mainland)
| | - Jie Xu
- School of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China (mainland).,Southern Medical University Technology, Ltd., Shunde Science Park, Guangzhou, Guangdong, China (mainland)
| | - Shijie Fu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China (mainland).,Academician Workstation in Luzhou, Luzhou, Sichuan, China (mainland)
| | - Bo Qin
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China (mainland).,Academician Workstation in Luzhou, Luzhou, Sichuan, China (mainland)
| | - Yang Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China (mainland).,Academician Workstation in Luzhou, Luzhou, Sichuan, China (mainland)
| | - Yang Yang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Southern Medical University Technology, Ltd., Shunde Science Park, Guangzhou, Guangdong, China (mainland).,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangzhou, Guangdong, China (mainland)
| | - Mian Wang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Southern Medical University Technology, Ltd., Shunde Science Park, Guangzhou, Guangdong, China (mainland).,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangzhou, Guangdong, China (mainland)
| | - Ding Li
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Southern Medical University Technology, Ltd., Shunde Science Park, Guangzhou, Guangdong, China (mainland).,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangzhou, Guangdong, China (mainland)
| | - Shizhen Zhong
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Academician Workstation in Luzhou, Luzhou, Sichuan, China (mainland)
| | - Wenhua Huang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Academician Workstation in Luzhou, Luzhou, Sichuan, China (mainland).,Southern Medical University Technology, Ltd., Shunde Science Park, Guangzhou, Guangdong, China (mainland).,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
14
|
Rosina M, Langone F, Giuliani G, Cerquone Perpetuini A, Reggio A, Calderone A, Fuoco C, Castagnoli L, Gargioli C, Cesareni G. Osteogenic differentiation of skeletal muscle progenitor cells is activated by the DNA damage response. Sci Rep 2019; 9:5447. [PMID: 30931986 PMCID: PMC6443689 DOI: 10.1038/s41598-019-41926-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/19/2019] [Indexed: 12/27/2022] Open
Abstract
Heterotopic ossification (HO) is a pathological condition characterized by the deposition of mineralized tissue in ectopic locations such as the skeletal muscle. The precise cellular origin and molecular mechanisms underlying HO are still debated. In our study we focus on the differentiation of mesoangioblasts (MABs), a population of multipotent skeletal muscle precursors. High-content screening for small molecules that perturb MAB differentiation decisions identified Idoxuridine (IdU), an antiviral and radiotherapy adjuvant, as a molecule that promotes MAB osteogenic differentiation while inhibiting myogenesis. IdU-dependent osteogenesis does not rely on the canonical BMP-2/SMADs osteogenic pathway. At pro-osteogenic conditions IdU induces a mild DNA Damage Response (DDR) that activates ATM and p38 eventually promoting the phosphorylation of the osteogenesis master regulator RUNX2. By interfering with this pathway IdU-induced osteogenesis is severely impaired. Overall, our study suggests that induction of the DDR promotes osteogenesis in muscle resident MABs thereby offering a new mechanism that may be involved in the ectopic deposition of mineralized tissue in the muscle.
Collapse
Affiliation(s)
- M Rosina
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - F Langone
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - G Giuliani
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | | | - A Reggio
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - A Calderone
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - C Fuoco
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - L Castagnoli
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - C Gargioli
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
| | - G Cesareni
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.
| |
Collapse
|
15
|
Łęgosz P, Drela K, Pulik Ł, Sarzyńska S, Małdyk P. Challenges of heterotopic ossification-Molecular background and current treatment strategies. Clin Exp Pharmacol Physiol 2018; 45:1229-1235. [DOI: 10.1111/1440-1681.13025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 01/27/2023]
Affiliation(s)
- Paweł Łęgosz
- Department of Orthopaedics and Traumatology; 1st Faculty of Medicine; Medical University of Warsaw; Warsaw Poland
| | - Katarzyna Drela
- NeuroRepair Department; Mossakowski Medical Research Centre; Polish Academy of Sciences; Warsaw Poland
| | - Łukasz Pulik
- Department of Orthopaedics and Traumatology; 1st Faculty of Medicine; Medical University of Warsaw; Warsaw Poland
| | - Sylwia Sarzyńska
- Department of Orthopaedics and Traumatology; 1st Faculty of Medicine; Medical University of Warsaw; Warsaw Poland
| | - Paweł Małdyk
- Department of Orthopaedics and Traumatology; 1st Faculty of Medicine; Medical University of Warsaw; Warsaw Poland
| |
Collapse
|
16
|
Immunological and morphological analysis of heterotopic ossification differs to healthy controls. BMC Musculoskelet Disord 2018; 19:327. [PMID: 30205831 PMCID: PMC6134767 DOI: 10.1186/s12891-018-2246-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Background Formation of lamellar bone in non-osseus tissue is a pathological process called heterotopic ossification. It is the aim of this study to analyse the morphology and immunological status of patients with heterotopic ossification compared to individual healthy persons. Methods Human bone marrow and blood samples were obtained from 6 systemically healthy individuals and 4 patients during resection of heterotopic ossification from bone at hip arthroplasty. Bone was fragmented and treated with purified collagenase. Immunofluorescence surface staining was performed and analyzed with flow cytometry. Microcomputed tomography scanning was done performed at a resolution of 11 and 35 μm isometric voxel size respectively using a two different cone beam X-computer tomography systems and a microfocus X-ray tube. Subsequently the volume data was morphometrically analysed. Results The monocytes, stem cells, stroma cells and granulocytes progenitor cells were strongly reduced in the heterotopic ossification patient. Additionally a significant reduction of stromal stem cells cells and CD34 positive stem cells was observed. The frequency of NK-cells, B cells and T cells were not altered in the patients with heterotopic ossification compared to a healthy person. Micromorphometric parameters showed a lower content of mineralized bone tissue compared to normal bone. Mean trabecular thickness showed a high standard deviation, indicating a high variation in trabecular thickness, anisotropy and reducing bone strength. Conclusions This work shows altered immunological distribution that is accompanied by a low decrease in bone volume fraction and tissue mineral density in the heterotopic ossification sample compared to normal bone. Compared to healthy subjects, this might reflect an immunological participation in the development of this entity.
Collapse
|
17
|
Perez JR, Kouroupis D, Li DJ, Best TM, Kaplan L, Correa D. Tissue Engineering and Cell-Based Therapies for Fractures and Bone Defects. Front Bioeng Biotechnol 2018; 6:105. [PMID: 30109228 PMCID: PMC6079270 DOI: 10.3389/fbioe.2018.00105] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/09/2018] [Indexed: 12/25/2022] Open
Abstract
Bone fractures and segmental bone defects are a significant source of patient morbidity and place a staggering economic burden on the healthcare system. The annual cost of treating bone defects in the US has been estimated to be $5 billion, while enormous costs are spent on bone grafts for bone injuries, tumors, and other pathologies associated with defective fracture healing. Autologous bone grafts represent the gold standard for the treatment of bone defects. However, they are associated with variable clinical outcomes, postsurgical morbidity, especially at the donor site, and increased surgical costs. In an effort to circumvent these limitations, tissue engineering and cell-based therapies have been proposed as alternatives to induce and promote bone repair. This review focuses on the recent advances in bone tissue engineering (BTE), specifically looking at its role in treating delayed fracture healing (non-unions) and the resulting segmental bone defects. Herein we discuss: (1) the processes of endochondral and intramembranous bone formation; (2) the role of stem cells, looking specifically at mesenchymal (MSC), embryonic (ESC), and induced pluripotent (iPSC) stem cells as viable building blocks to engineer bone implants; (3) the biomaterials used to direct tissue growth, with a focus on ceramic, biodegradable polymers, and composite materials; (4) the growth factors and molecular signals used to induce differentiation of stem cells into the osteoblastic lineage, which ultimately leads to active bone formation; and (5) the mechanical stimulation protocols used to maintain the integrity of the bone repair and their role in successful cell engraftment. Finally, a couple clinical scenarios are presented (non-unions and avascular necrosis—AVN), to illustrate how novel cell-based therapy approaches can be used. A thorough understanding of tissue engineering and cell-based therapies may allow for better incorporation of these potential therapeutic approaches in bone defects allowing for proper bone repair and regeneration.
Collapse
Affiliation(s)
- Jose R Perez
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Deborah J Li
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Lee Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
18
|
Abstract
Heterotopic ossification (HO) is abnormal formation of mature lamellar bone in soft tissues. HO is most commonly diagnosed in the setting of localized trauma, which results in improper differentiation of progenitor cells, leading to aberrant tissue formation. In the pediatric population, nongenetic causes of HO have rarely been reported, especially HO involving the tendons of the ankle. We present a case of HO of the peroneus brevis tendon without systemic disease in a pediatric patient. The patient was a 7-year-old female with a normal birth and developmental history who first presented 6 weeks after a right ankle sprain with pain localized to the lateral calcaneus. Prominent swelling and tenderness to palpation were noted over the peroneal tubercle. Radiographic imaging showed dystrophic calcification within the peroneus brevis tendon. After failed conservative management, the heterotopic ossified mass (1.5 × 0.3 cm) was excised from the peroneus brevis tendon. The tendon was primarily repaired. The patient was followed up for 12 weeks postoperatively and achieved full resolution of her pain with a return to normal activity. HO has been theorized to be the result of an imbalance between bone mineralization and demineralization. In the setting of localized trauma, inductive agents have been implicated in pathologic bone formation. In the pediatric population, HO has rarely been diagnosed in the absence of genetic causes. In patients presenting with lateral foot and ankle pain, HO of the peroneal tendons should be considered in the differential diagnosis. In a patient with pain secondary to HO, surgical excision of the heterotopic mass can achieve symptom resolution.
Collapse
Affiliation(s)
- Karan Dua
- Surgeon, Department of Orthopaedic Surgery and Rehabilitation Medicine, SUNY Downstate Medical Center, Brooklyn, NY
| | - James M Barsi
- Assistant Professor, Department of Orthopaedic Surgery, Stony Brook University School of Medicine, Stony Brook, NY.
| |
Collapse
|
19
|
Eisenstein N, Stapley S, Grover L. Post-Traumatic Heterotopic Ossification: An Old Problem in Need of New Solutions. J Orthop Res 2018; 36:1061-1068. [PMID: 29193256 DOI: 10.1002/jor.23808] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/12/2017] [Indexed: 02/04/2023]
Abstract
Heterotopic ossification (HO) is the formation of pathological bone in ectopic sites and it can have serious consequences for functional outcomes. For many years, its main clinical relevance was as a rare complication of elective joint arthroplasty or CNS injury and a number of prophylaxes were developed to mitigate against it in these settings. As a consequence of changes in patterns of wounding and survival in conflicts since the turn of the century, post-traumatic HO has become much more common and case severity has increased. It represents one of the main barriers to rehabilitation in a large cohort of combat-injured patients. However, extant prophylaxes have not been shown to be effective or appropriate in this patient cohort. In addition, the lack of reliable early detection or means of predicting which patients will develop HO is another barrier to effective prevention. This review examines the current state of understanding of post-traumatic HO including the historical context, epidemiology, pathophysiology, clinical issues, currently prophylaxis and detection, management, and potential future approaches. Our aims are to highlight the current lack of effective means of early detection and prevention of HO after major trauma and to stimulate research into novel solutions to this challenging problem. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1061-1068, 2018.
Collapse
Affiliation(s)
- Neil Eisenstein
- Royal Centre for Defence Medicine, Birmingham Research Park, ICT Centre, Vincent Drive, Birmingham, B15 2SQ, United Kingdom.,School of Chemical Engineering, University of Birmingham, Edgbaston, B15 2TT, United Kingdom
| | - Sarah Stapley
- Royal Centre for Defence Medicine, Birmingham Research Park, ICT Centre, Vincent Drive, Birmingham, B15 2SQ, United Kingdom.,School of Chemical Engineering, University of Birmingham, Edgbaston, B15 2TT, United Kingdom
| | - Liam Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, B15 2TT, United Kingdom
| |
Collapse
|
20
|
Brady RD, Shultz SR, McDonald SJ, O'Brien TJ. Neurological heterotopic ossification: Current understanding and future directions. Bone 2018; 109:35-42. [PMID: 28526267 DOI: 10.1016/j.bone.2017.05.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 12/30/2022]
Abstract
Neurological heterotopic ossification (NHO) involves the formation of bone in soft tissue following a neurological condition, of which the most common are brain and spinal cord injuries. NHO often forms around the hip, knee and shoulder joints, causing severe pain and joint deformation which is associated with significant morbidity and reduced quality of life. The cellular and molecular events that initiate NHO have been the focus of an increasing number of human and animal studies over the past decade, with this work largely driven by the need to unearth potential therapeutic interventions to prevent the formation of NHO. This review provides an overview of the present understanding of NHO pathogenesis and pathobiology, current treatments, novel therapeutic targets, potential biomarkers and future directions.
Collapse
Affiliation(s)
- Rhys D Brady
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC, 3010, Australia.
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC, 3010, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC, 3086, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC, 3010, Australia
| |
Collapse
|
21
|
Cholok D, Chung MT, Ranganathan K, Ucer S, Day D, Davis TA, Mishina Y, Levi B. Heterotopic ossification and the elucidation of pathologic differentiation. Bone 2018; 109:12-21. [PMID: 28987285 PMCID: PMC6585944 DOI: 10.1016/j.bone.2017.09.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 01/23/2023]
Abstract
Tissue regeneration following acute or persistent inflammation can manifest a spectrum of phenotypes ranging from the adaptive to the pathologic. Heterotopic Ossification (HO), the endochondral formation of bone within soft-tissue structures following severe injury serves as a prominent example of pathologic differentiation; and remains a persistent clinical issue incurring significant patient morbidity and expense to adequately diagnose and treat. The pathogenesis of HO provides an intriguing opportunity to better characterize the cellular and cell-signaling contributors to aberrant differentiation. Indeed, recent work has continued to resolve the unique cellular lineages, and causative pathways responsible for ectopic bone development yielding promising avenues for the development of novel therapeutic strategies shown to be successful in analogous animal models of HO development. This review details advances in the understanding of HO in the context of inciting inflammation, and explains how these advances inform the current standards of diagnosis and treatment.
Collapse
Affiliation(s)
- David Cholok
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Michael T Chung
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Kavitha Ranganathan
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Serra Ucer
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Devaveena Day
- Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - Thomas A Davis
- Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, MD, USA; Department of Surgery, Uniformed Services University of the Health Sciences & the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Yuji Mishina
- School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Cell-Surface Protein Profiling Identifies Distinctive Markers of Progenitor Cells in Human Skeletal Muscle. Stem Cell Reports 2017; 7:263-78. [PMID: 27509136 PMCID: PMC4983081 DOI: 10.1016/j.stemcr.2016.07.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/08/2016] [Accepted: 07/08/2016] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle contains two distinct stem/progenitor populations. One is the satellite cell, which acts as a muscle stem cell, and the other is the mesenchymal progenitor, which contributes to muscle pathogeneses such as fat infiltration and fibrosis. Detailed and accurate characterization of these progenitors in humans remains elusive. Here, we performed comprehensive cell-surface protein profiling of the two progenitor populations residing in human skeletal muscle and identified three previously unrecognized markers: CD82 and CD318 for satellite cells and CD201 for mesenchymal progenitors. These markers distinguish myogenic and mesenchymal progenitors, and enable efficient isolation of the two types of progenitors. Functional study revealed that CD82 ensures expansion and preservation of myogenic progenitors by suppressing excessive differentiation, and CD201 signaling favors adipogenesis of mesenchymal progenitors. Thus, cell-surface proteins identified here are not only useful markers but also functionally important molecules, and provide valuable insight into human muscle biology and diseases.
Collapse
|
23
|
Torossian F, Guerton B, Anginot A, Alexander KA, Desterke C, Soave S, Tseng HW, Arouche N, Boutin L, Kulina I, Salga M, Jose B, Pettit AR, Clay D, Rochet N, Vlachos E, Genet G, Debaud C, Denormandie P, Genet F, Sims NA, Banzet S, Levesque JP, Lataillade JJ, Le Bousse-Kerdilès MC. Macrophage-derived oncostatin M contributes to human and mouse neurogenic heterotopic ossifications. JCI Insight 2017; 2:96034. [PMID: 29093266 DOI: 10.1172/jci.insight.96034] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/26/2017] [Indexed: 02/04/2023] Open
Abstract
Neurogenic heterotopic ossification (NHO) is the formation of ectopic bone generally in muscles surrounding joints following spinal cord or brain injury. We investigated the mechanisms of NHO formation in 64 patients and a mouse model of spinal cord injury-induced NHO. We show that marrow from human NHOs contains hematopoietic stem cell (HSC) niches, in which mesenchymal stromal cells (MSCs) and endothelial cells provide an environment supporting HSC maintenance, proliferation, and differentiation. The transcriptomic signature of MSCs from NHOs shows a neuronal imprinting associated with a molecular network required for HSC support. We demonstrate that oncostatin M (OSM) produced by activated macrophages promotes osteoblastic differentiation and mineralization of human muscle-derived stromal cells surrounding NHOs. The key role of OSM was confirmed using an experimental model of NHO in mice defective for the OSM receptor (OSMR). Our results provide strong evidence that macrophages contribute to NHO formation through the osteogenic action of OSM on muscle cells within an inflammatory context and suggest that OSM/OSMR could be a suitable therapeutic target. Altogether, the evidence of HSCs in ectopic bones growing at the expense of soft tissue in spinal cord/brain-injured patients indicates that inflammation and muscle contribute to HSC regulation by the brain-bone-blood triad.
Collapse
Affiliation(s)
- Frédéric Torossian
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Bernadette Guerton
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Adrienne Anginot
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Kylie A Alexander
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | | | - Sabrina Soave
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Hsu-Wen Tseng
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nassim Arouche
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Laetitia Boutin
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Irina Kulina
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Marjorie Salga
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.,Université de Versailles Saint-Quentin-en-Yvelines, Evolution of neuromuscular diseases: innovative concepts and practices, Inserm U1179, Montigny le Bretonneux, France
| | - Beulah Jose
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Allison R Pettit
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Denis Clay
- UMS33, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Nathalie Rochet
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Erica Vlachos
- Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Guillaume Genet
- Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Charlotte Debaud
- Université de Versailles Saint-Quentin-en-Yvelines, Evolution of neuromuscular diseases: innovative concepts and practices, Inserm U1179, Montigny le Bretonneux, France.,Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Philippe Denormandie
- Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - François Genet
- Université de Versailles Saint-Quentin-en-Yvelines, Evolution of neuromuscular diseases: innovative concepts and practices, Inserm U1179, Montigny le Bretonneux, France.,Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research and Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Sébastien Banzet
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France.,Centre de Transfusion Sanguine des Armées, L'Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Jean-Pierre Levesque
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Jean-Jacques Lataillade
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France.,Centre de Transfusion Sanguine des Armées, L'Institut de Recherche Biomédicale des Armées, Clamart, France
| | | |
Collapse
|
24
|
Porras DP, Abbaszadeh M, Bhattacharya D, D'Souza NC, Edjiu NR, Perry CGR, Scimè A. p107 Determines a Metabolic Checkpoint Required for Adipocyte Lineage Fates. Stem Cells 2017; 35:1378-1391. [PMID: 28233396 DOI: 10.1002/stem.2576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/07/2017] [Indexed: 12/14/2022]
Abstract
We show that the transcriptional corepressor p107 orchestrates a metabolic checkpoint that determines adipocyte lineage fates for non-committed progenitors. p107 accomplishes this when stem cell commitment would normally occur in growth arrested cells. p107-deficient embryonic progenitors are characterized by a metabolic state resembling aerobic glycolysis that is necessary for their pro-thermogenic fate. Indeed, during growth arrest they have a reduced capacity for NADH partitioning between the cytoplasm and mitochondria. Intriguingly, this occurred despite an increase in the capacity for mitochondrial oxidation of non-glucose substrates. The significance of metabolic reprogramming is underscored by the disruption of glycolytic capacities in p107-depleted progenitors that reverted their fates from pro-thermogenic to white adipocytes. Moreover, the manipulation of glycolytic capacity on nonspecified embryonic and adult progenitors forced their beige fat commitment. These innovative findings introduce a new approach to increase pro-thermogenic adipocytes based on simply promoting aerobic glycolysis to manipulate nonspecified progenitor fate decisions. Stem Cells 2017;35:1378-1391.
Collapse
Affiliation(s)
- Deanna P Porras
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Maryam Abbaszadeh
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Debasmita Bhattacharya
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Ninoschka C D'Souza
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Nareh R Edjiu
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Christopher G R Perry
- Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Anthony Scimè
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Supanc HRH, Gorman S, Tuan RS. Traumatized muscle-derived multipotent progenitor cells recruit endothelial cells through vascular endothelial growth factor-A action. J Tissue Eng Regen Med 2017; 11:3038-3047. [PMID: 28078807 DOI: 10.1002/term.2205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 02/24/2016] [Accepted: 03/27/2016] [Indexed: 12/21/2022]
Abstract
Traumatized muscle, such as that debrided from blast injury sites, is considered a promising and convenient tissue source for multipotent progenitor cells (MPCs), a population of adult mesenchymal stem cell (MSC)-like cells. The present study aimed to assess the regenerative therapeutic potential of human traumatized muscle-derived MPCs, e.g., for injury repair in the blast-traumatized extremity, by comparing their pro-angiogenic potential in vitro and capillary recruitment activity in vivo to those of MSCs isolated from human bone marrow, a widely-used tissue source. MPCs were tested for their direct and indirect effects on human microvascular endothelial cells (ECs) in vitro. The findings reported here showed that MPC-conditioned culture medium (MPC-CM), like MSC-CM, promoted EC-cord network branching. Silent (si)RNA-mediated silencing of vascular endothelial growth factor-A (VEGF-A) expression in MPCs attenuated this effect. In a chick embryonic chorioallantoic membrane in vivo angiogenesis assay, MPCs encapsulated in photocrosslinked gelatin scaffold recruited blood vessels more efficiently than either MSCs or human foreskin fibroblasts. Together, these findings support the potential application of traumatized muscle-derived MPCs in cell-based regenerative medicine therapies as a result of their influence on EC organization. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Heidi R H Supanc
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, School of Medicine.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA
| | - Shannon Gorman
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, School of Medicine.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
26
|
Inhibition of connexin 43 prevents trauma-induced heterotopic ossification. Sci Rep 2016; 6:37184. [PMID: 27849058 PMCID: PMC5111117 DOI: 10.1038/srep37184] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/26/2016] [Indexed: 12/14/2022] Open
Abstract
Heterotopic ossification (HO) can result from traumatic injury, surgery or genetic diseases. Here, we demonstrate that overexpression of connexin 43 (Cx43) is critical for the development and recurrence of traumatic HO in patients. Inhibition of Cx43 by shRNA substantially suppressed the osteogenic differentiation of MC-3T3 cells and the expression of osteogenic genes. We employed a tenotomy mouse model to explore the hypothesis that Cx43 is vital to the development of HO. Inhibition of Cx43 by a specific shRNA decreased extraskeletal bone formation in vivo. In addition, we demonstrated that ERK signaling activated by Cx43 plays an important role in promoting HO. ERK signaling was highly activated in HO tissue collected from patient and mouse models. Importantly, de novo soft tissue HO was significantly attenuated in mice treated with U0126. Inhibition of Cx43 and ERK led to decreased expressions of Runx2, BSP and Col-1 in vivo and in vitro. Moreover, HO patients with low Cx43 expression or ERK activation had a lower risk of recurrence after the lesions were surgically removed. Our findings indicate that Cx43 promotes trauma-induced HO formation by activating the ERK pathway and enhances the expression of osteogenic markers.
Collapse
|
27
|
miR-203 inhibits the traumatic heterotopic ossification by targeting Runx2. Cell Death Dis 2016; 7:e2436. [PMID: 27787524 PMCID: PMC5133990 DOI: 10.1038/cddis.2016.325] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 12/25/2022]
Abstract
Emerging evidence has indicated that dysregulated microRNAs (miRNAs) have an important role in bone formation. However, the pathophysiological role of miRNAs in traumatic heterotopic ossification (HO) remains to be elucidated. Using gene expression profile analyses and subsequent confirmation with real-time PCR assays, we identified the decreased expression of miRNA-203 (miR-203) and increased expression of Runx2 as responses to the development of traumatic HO. We found that miR-203 expression was markedly higher in primary and recurrent HO tissues than in normal bones. The upregulation of miR-203 significantly decreased the level of Runx2 expression, whereas miR-203 downregulation increased Runx2 expression. Mutation of the putative miR-203-binding sites in Runx2 mRNA abolished miR-203-mediated repression of Runx2 3'-untranslated region luciferase reporter activity, indicating that Runx2 is an important target of miR-203 in osteoblasts. We also found that miR-203 is negatively correlated with osteoblast differentiation. Furthermore, in vitro osteoblast activity and matrix mineralization were promoted by antagomir-203 and decreased by agomir-203. We showed that miR-203 suppresses osteoblast activity by inhibiting the β-catenin and extracellular signal-regulated kinase pathways. Moreover, using a tenotomy mouse HO model, we found an inhibitory role of miR-203 in regulating HO in vivo; pretreatment with antagomiR-203 increased the development of HO. These data suggest that miR-203 has a crucial role in suppressing HO by directly targeting Runx2 and that the therapeutic overexpression of miR-203 may be a potential strategy for treating traumatic HO.
Collapse
|
28
|
Agarwal S, Loder SJ, Sorkin M, Li S, Shrestha S, Zhao B, Mishina Y, James AW, Levi B. Analysis of Bone-Cartilage-Stromal Progenitor Populations in Trauma Induced and Genetic Models of Heterotopic Ossification. Stem Cells 2016; 34:1692-701. [PMID: 27068890 DOI: 10.1002/stem.2376] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 03/14/2016] [Indexed: 11/09/2022]
Abstract
Heterotopic ossification (HO), the formation of extra-skeletal bone in soft tissues, is a pathologic process occurring after substantial burns or trauma, or in patients with type I bone morphogenetic protein (BMP) receptor hyperactivating mutations. Identifying the cells responsible for de novo bone formation during adulthood is of critical importance for therapeutic and regenerative purposes. Using a model of trauma-induced HO with hind limb Achilles' tenotomy and dorsal burn injury and a genetic nontrauma HO model (Nfatc1-Cre/caAcvr1(fl/wt) ), we demonstrate enrichment of previously defined bone-cartilage-stromal progenitor cells (BCSP: AlphaV+/CD105+/Tie2-/CD45-/Thy1-/6C3-) at the site of HO formation when compared with marrow isolated from the ipsilateral hind limb, or from tissue of the contralateral, uninjured hind limb. Upon transplantation into tenotomy sites soon after injury, BCSPs isolated from neonatal mice or developing HO incorporate into the developing lesion in cartilage and bone and express chondrogenic and osteogenic transcription factors. Additionally, BCSPs isolated from developing HO similarly incorporate into new HO lesions upon transplantation. Finally, adventitial cells, but not pericytes, appear to play a supportive role in HO formation. Our findings indicate that BCSPs contribute to de novo bone formation during adulthood and may hold substantial regenerative potential. Stem Cells 2016;34:1692-1701.
Collapse
Affiliation(s)
- Shailesh Agarwal
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Shawn J Loder
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Sorkin
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Shuli Li
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Swati Shrestha
- Department of Pathology & Laboratory Medicine and Orthopaedic Hospital Research Center, University of California, Los Angeles
| | - Bin Zhao
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yuji Mishina
- School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Aaron W James
- Department of Pathology & Laboratory Medicine and Orthopaedic Hospital Research Center, University of California, Los Angeles
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Agarwal S, Loder S, Brownley C, Cholok D, Mangiavini L, Li J, Breuler C, Sung HH, Li S, Ranganathan K, Peterson J, Tompkins R, Herndon D, Xiao W, Jumlongras D, Olsen BR, Davis TA, Mishina Y, Schipani E, Levi B. Inhibition of Hif1α prevents both trauma-induced and genetic heterotopic ossification. Proc Natl Acad Sci U S A 2016; 113:E338-47. [PMID: 26721400 PMCID: PMC4725488 DOI: 10.1073/pnas.1515397113] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pathologic extraskeletal bone formation, or heterotopic ossification (HO), occurs following mechanical trauma, burns, orthopedic operations, and in patients with hyperactivating mutations of the type I bone morphogenetic protein receptor ACVR1 (Activin type 1 receptor). Extraskeletal bone forms through an endochondral process with a cartilage intermediary prompting the hypothesis that hypoxic signaling present during cartilage formation drives HO development and that HO precursor cells derive from a mesenchymal lineage as defined by Paired related homeobox 1 (Prx). Here we demonstrate that Hypoxia inducible factor-1α (Hif1α), a key mediator of cellular adaptation to hypoxia, is highly expressed and active in three separate mouse models: trauma-induced, genetic, and a hybrid model of genetic and trauma-induced HO. In each of these models, Hif1α expression coincides with the expression of master transcription factor of cartilage, Sox9 [(sex determining region Y)-box 9]. Pharmacologic inhibition of Hif1α using PX-478 or rapamycin significantly decreased or inhibited extraskeletal bone formation. Importantly, de novo soft-tissue HO was eliminated or significantly diminished in treated mice. Lineage-tracing mice demonstrate that cells forming HO belong to the Prx lineage. Burn/tenotomy performed in lineage-specific Hif1α knockout mice (Prx-Cre/Hif1α(fl:fl)) resulted in substantially decreased HO, and again lack of de novo soft-tissue HO. Genetic loss of Hif1α in mesenchymal cells marked by Prx-cre prevents the formation of the mesenchymal condensations as shown by routine histology and immunostaining for Sox9 and PDGFRα. Pharmacologic inhibition of Hif1α had a similar effect on mesenchymal condensation development. Our findings indicate that Hif1α represents a promising target to prevent and treat pathologic extraskeletal bone.
Collapse
MESH Headings
- Activin Receptors, Type I/metabolism
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Animals
- Burns/complications
- Burns/genetics
- Chondrogenesis/drug effects
- Chondrogenesis/genetics
- Disease Models, Animal
- Gene Regulatory Networks/drug effects
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Integrases/metabolism
- Luminescent Measurements
- Mesenchymal Stem Cells/drug effects
- Mice, Knockout
- Models, Biological
- Mustard Compounds/pharmacology
- Ossification, Heterotopic/diagnostic imaging
- Ossification, Heterotopic/drug therapy
- Ossification, Heterotopic/genetics
- Ossification, Heterotopic/prevention & control
- Phenylpropionates/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- SOX9 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Sirolimus/pharmacology
- Tendons/drug effects
- Tendons/pathology
- Tendons/surgery
- Tenotomy
- Up-Regulation/drug effects
- Wound Healing/drug effects
- Wounds and Injuries/complications
- Wounds and Injuries/pathology
- X-Ray Microtomography
Collapse
Affiliation(s)
- Shailesh Agarwal
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Shawn Loder
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Cameron Brownley
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - David Cholok
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Laura Mangiavini
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI 48109
| | - John Li
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | | | - Hsiao H Sung
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Shuli Li
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | | | - Joshua Peterson
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Ronald Tompkins
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114
| | - David Herndon
- Department of Surgery, Shriners Hospital for Children and University of Texas Medical Branch, Galveston, TX 77555
| | - Wenzhong Xiao
- Department of Surgery, Genome Technology Center, Stanford University, Palo Alto, CA 94305
| | - Dolrudee Jumlongras
- Department of Developmental Biology, Harvard Dental School, Boston, MA 02115
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard Dental School, Boston, MA 02115
| | - Thomas A Davis
- Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, MD 20910
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109
| | - Ernestina Schipani
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI 48109;
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
30
|
Davies OG, Grover LM, Eisenstein N, Lewis MP, Liu Y. Identifying the Cellular Mechanisms Leading to Heterotopic Ossification. Calcif Tissue Int 2015; 97:432-44. [PMID: 26163233 DOI: 10.1007/s00223-015-0034-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/02/2015] [Indexed: 12/19/2022]
Abstract
Heterotopic ossification (HO) is a debilitating condition defined by the de novo development of bone within non-osseous soft tissues, and can be either hereditary or acquired. The hereditary condition, fibrodysplasia ossificans progressiva is rare but life threatening. Acquired HO is more common and results from a severe trauma that produces an environment conducive for the formation of ectopic endochondral bone. Despite continued efforts to identify the cellular and molecular events that lead to HO, the mechanisms of pathogenesis remain elusive. It has been proposed that the formation of ectopic bone requires an osteochondrogenic cell type, the presence of inductive agent(s) and a permissive local environment. To date several lineage-tracing studies have identified potential contributory populations. However, difficulties identifying cells in vivo based on the limitations of phenotypic markers, along with the absence of established in vitro HO models have made the results difficult to interpret. The purpose of this review is to critically evaluate current literature within the field in an attempt identify the cellular mechanisms required for ectopic bone formation. The major aim is to collate all current data on cell populations that have been shown to possess an osteochondrogenic potential and identify environmental conditions that may contribute to a permissive local environment. This review outlines the pathology of endochondral ossification, which is important for the development of potential HO therapies and to further our understanding of the mechanisms governing bone formation.
Collapse
Affiliation(s)
- O G Davies
- School of Mechanical and Manufacturing Engineering, Loughborough University, Ashby Road, Loughborough, LE11 3TU, UK.
- Centre for Biological Engineering, Loughborough University, Loughborough, LE11 3TU, UK.
| | - L M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - N Eisenstein
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - M P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Epinal Way, Loughborough, LE11 3TU, UK
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Loughborough, UK
- National Centre for Sport and Exercise Medicine, Loughborough University, Epinal Way, Loughborough, LE11 3TU, UK
| | - Y Liu
- School of Mechanical and Manufacturing Engineering, Loughborough University, Ashby Road, Loughborough, LE11 3TU, UK
| |
Collapse
|
31
|
Abstract
An 18-year-old man presented with mid left thigh pain after sequential lacrosse injuries 1 month and 2 weeks prior. Physical examination was significant for a tender mass in the mid left thigh.
Collapse
|
32
|
Agarwal S, Loder SJ, Brownley C, Eboda O, Peterson JR, Hayano S, Wu B, Zhao B, Kaartinen V, Wong VC, Mishina Y, Levi B. BMP signaling mediated by constitutively active Activin type 1 receptor (ACVR1) results in ectopic bone formation localized to distal extremity joints. Dev Biol 2015; 400:202-9. [PMID: 25722188 DOI: 10.1016/j.ydbio.2015.02.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/11/2015] [Accepted: 02/15/2015] [Indexed: 10/24/2022]
Abstract
BMP signaling mediated by ACVR1 plays a critical role for development of multiple structures including the cardiovascular and skeletal systems. While deficient ACVR1 signaling impairs normal embryonic development, hyperactive ACVR1 function (R206H in humans and Q207D mutation in mice, ca-ACVR1) results in formation of heterotopic ossification (HO). We developed a mouse line, which conditionally expresses ca-ACVR1 with Nfatc1-Cre(+) transgene. Mutant mice developed ectopic cartilage and bone at the distal joints of the extremities including the interphalangeal joints and hind limb ankles as early as P4 in the absence of trauma or exogenous bone morphogenetic protein (BMP) administration. Micro-CT showed that even at later time points (up to P40), cartilage and bone development persisted at the affected joints most prominently in the ankle. Interestingly, this phenotype was not present in areas of bone outside of the joints - tibia are normal in mutants and littermate controls away from the ankle. These findings demonstrate that this model may allow for further studies of heterotopic ossification, which does not require the use of stem cells, direct trauma or activation with exogenous Cre gene administration.
Collapse
Affiliation(s)
- Shailesh Agarwal
- University of Michigan Medical School, Department of Surgery, Ann Arbor, MI, USA
| | - Shawn J Loder
- University of Michigan Medical School, Department of Surgery, Ann Arbor, MI, USA
| | - Cameron Brownley
- University of Michigan Medical School, Department of Surgery, Ann Arbor, MI, USA
| | - Oluwatobi Eboda
- University of Michigan Medical School, Department of Surgery, Ann Arbor, MI, USA
| | - Jonathan R Peterson
- University of Michigan Medical School, Department of Surgery, Ann Arbor, MI, USA
| | - Satoru Hayano
- University of Michigan, School of Dentistry, Department of Biologic and Materials Sciences, Ann Arbor, MI, USA
| | - Bingrou Wu
- Albert Einstein College of Medicine, Department of Genetics, Bronx, New York, USA
| | - Bin Zhao
- Albert Einstein College of Medicine, Department of Genetics, Bronx, New York, USA
| | - Vesa Kaartinen
- University of Michigan, School of Dentistry, Department of Biologic and Materials Sciences, Ann Arbor, MI, USA
| | - Victor C Wong
- Johns Hopkins University, Department of Plastic Surgery, Baltimore, MD, USA
| | - Yuji Mishina
- University of Michigan, School of Dentistry, Department of Biologic and Materials Sciences, Ann Arbor, MI, USA.
| | - Benjamin Levi
- University of Michigan Medical School, Department of Surgery, Ann Arbor, MI, USA.
| |
Collapse
|
33
|
Trensz F, Lucien F, Couture V, Söllrald T, Drouin G, Rouleau AJ, Grandbois M, Lacraz G, Grenier G. Increased microenvironment stiffness in damaged myofibers promotes myogenic progenitor cell proliferation. Skelet Muscle 2015; 5:5. [PMID: 25729564 PMCID: PMC4343274 DOI: 10.1186/s13395-015-0030-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/19/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The stiffness of the myogenic stem cell microenvironment markedly influences the ability to regenerate tissue. We studied the effect of damaged myofibers on myogenic progenitor cell (MPC) proliferation and determined whether the structural integrity of the microenvironment contributes to phenotypic changes. METHODS Individual myofibers were isolated and cultured for 6 days. During this period, the cytoskeleton of myofibers and transcription factors regulating MPC differentiation were characterized by immunostaining. Atomic Force Microscopy (AFM) was performed to measure stiffness of cultured myofibers. Healthy and damaged myofibers, and their associated MPCs, were studied in skeletal muscle from dystrophic and tenotomy mouse models. MPCs were cultured on stiffness-tunable substrates, and their phenotypes were assessed by immunostaining of myogenic transcription factors. RESULTS We showed that individual myofibers tend to shrink or collapse when cultured ex vivo starting from day 1 and that this is associated with a marked increase in the number of proliferative MPCs (Pax7(+)MyoD(+)). The myofibers collapsed due to a loss of viability as shown by Evans blue dye uptake and the disorganization of their cytoskeletons. Interestingly, collapsed myofibers in mdx skeletal muscles were similar to damaged myofibers in that they lose their viability, have a disorganized cytoskeleton (actin and α-actinin), and display local MPC (MyoD(+)) proliferation at their periphery. In a tenotomy model that causes loss of muscle tension, the cytoskeletal disorganization of myofibers also correlated with the activation/proliferation of MPCs. A deeper analysis of collapsed myofibers revealed that they produce trophic factors that influence MPC proliferation. In addition, collapsed myofibers expressed several genes related to the basal lamina. Immunostaining revealed the presence of fibronectin in the basal lamina and the cytoplasm of damaged myofibers. Lastly, using atomic force microscopy (AFM), we showed that collapsed myofibers exhibit greater stiffness than intact myofibers. Growing MPCs on a 2-kPa polyacrylamide-based substrate, exempt of additional microenvironmental cues, recapitulated proliferation and reduced spontaneous differentiation compared to growth on a 0.5-kPa substrate. CONCLUSIONS Our results support the notion that collapsed or damaged myofibers increase the structural stiffness of the satellite cell microenvironment, which in addition to other cues such as trophic factors and changes in extracellular matrix composition, promotes the proliferation and maintenance of MPCs, required for myofiber repair.
Collapse
Affiliation(s)
- Frédéric Trensz
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada
| | - Fabrice Lucien
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada
| | - Vanessa Couture
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada
| | - Thomas Söllrald
- Department of Electrical and Computer Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Geneviève Drouin
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada
| | - André-Jean Rouleau
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada
| | - Michel Grandbois
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada ; Department of Pharmacology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Gregory Lacraz
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada ; New address: Hubrecht Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Guillaume Grenier
- Research Centre of the Centre Hospitalier de l'Université de Sherbrooke (CRCHUS), Université de Sherbrooke, Sherbrooke, QC Canada ; Department of Orthopedic Surgery, Faculty of Medicine, Université de Sherbrooke, 3001-12th Avenue North, Sherbrooke, J1H 5N4, QC Canada
| |
Collapse
|