151
|
Souza DCD, Abreu HDLD, Oliveira PVD, Capelo LP, Passos-Bueno MR, Catalani LH. A fast degrading PLLA composite with a high content of functionalized octacalcium phosphate mineral phase induces stem cells differentiation. J Mech Behav Biomed Mater 2019; 93:93-104. [DOI: 10.1016/j.jmbbm.2019.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/02/2019] [Accepted: 02/03/2019] [Indexed: 01/24/2023]
|
152
|
Azevedo PO, Paiva AE, Santos GSP, Lousado L, Andreotti JP, Sena IFG, Tagliati CA, Mintz A, Birbrair A. Cross-talk between lung cancer and bones results in neutrophils that promote tumor progression. Cancer Metastasis Rev 2019; 37:779-790. [PMID: 30203108 DOI: 10.1007/s10555-018-9759-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lung cancer is the leading cause of cancer mortality around the world. The lack of detailed understanding of the cellular and molecular mechanisms participating in the lung tumor progression restrains the development of efficient treatments. Recently, by using state-of-the-art technologies, including in vivo sophisticated Cre/loxP technologies in combination with lung tumor models, it was revealed that osteoblasts activate neutrophils that promote tumor growth in the lung. Strikingly, genetic ablation of osteoblasts abolished lung tumor progression via interruption of SiglecFhigh-expressing neutrophils supply to the tumor microenvironment. Interestingly, SiglecFhigh neutrophil signature was associated with worse lung adenocarcinoma patients outcome. This study identifies novel cellular targets for lung cancer treatment. Here, we summarize and evaluate recent advances in our understanding of lung tumor microenvironment.
Collapse
Affiliation(s)
- Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza Lousado
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos A Tagliati
- Department of Clinical and Toxicological Analysis, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
153
|
Fecher-Trost C, Lux F, Busch KM, Raza A, Winter M, Hielscher F, Belkacemi T, van der Eerden B, Boehm U, Freichel M, Weissgerber P. Maternal Transient Receptor Potential Vanilloid 6 (Trpv6) Is Involved In Offspring Bone Development. J Bone Miner Res 2019; 34:699-710. [PMID: 30786075 DOI: 10.1002/jbmr.3646] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 12/29/2022]
Abstract
Embryonic growth and bone development depend on placental Ca2+ transport across the feto-maternal barrier to supply minerals to the fetus. The individual factors and cellular mechanisms that regulate placental Ca2+ transfer, however, are only beginning to emerge. We find that the Ca2+ -selective transient receptor potential vanilloid 6 (TRPV6) channel is expressed in trophoblasts of the fetal labyrinth, in the yolk sac, and in the maternal part of the placenta. Lack of functional TRPV6 channels in the mother leads to a reduced Ca2+ content in both placenta and embryo. Ca2+ uptake in trophoblasts is impaired in the absence of Trpv6. Trpv6-deficient embryos are smaller, have a lower body weight, and shorter and less calcified femurs. The altered cortical bone microarchitecture persists in adulthood. We show that TRPV6's Ca2+ -conducting property causes this embryonic and bone phenotype. Our results show that TRPV6 is necessary for the Ca2+ uptake in trophoblasts and that TRPV6 deficiency in the placenta leads to reduced embryo growth, minor bone calcification, and impaired bone development. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Claudia Fecher-Trost
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Femke Lux
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Kai-Markus Busch
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Ahsan Raza
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Manuel Winter
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Franziska Hielscher
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Thabet Belkacemi
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Bram van der Eerden
- Department of Internal Medicine, Laboratory for Calcium and Bone Metabolism, Erasmus MC, Rotterdam, Netherlands
| | - Ulrich Boehm
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Petra Weissgerber
- Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany.,Transgenic Technologies, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| |
Collapse
|
154
|
Qi H, Kim JK, Ha P, Chen X, Chen E, Chen Y, Li J, Pan HC, Yu M, Mohazeb Y, Azer S, Baik L, Kwak JH, Ting K, Zhang X, Hu M, Soo C. Inactivation of Nell-1 in Chondrocytes Significantly Impedes Appendicular Skeletogenesis. J Bone Miner Res 2019; 34:533-546. [PMID: 30352124 PMCID: PMC6677149 DOI: 10.1002/jbmr.3615] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/18/2018] [Accepted: 10/06/2018] [Indexed: 12/29/2022]
Abstract
NELL-1, an osteoinductive protein, has been shown to regulate skeletal ossification. Interestingly, an interstitial 11p14.1-p15.3 deletion involving the Nell-1 gene was recently reported in a patient with short stature and delayed fontanelle closure. Here we sought to define the role of Nell-1 in endochondral ossification by investigating Nell-1-specific inactivation in Col2α1-expressing cell lineages. Nell-1flox/flox ; Col2α1-Cre+ (Nell-1Col2α1 KO) mice were generated for comprehensive analysis. Nell-1Col2α1 KO mice were born alive but displayed subtle femoral length shortening. At 1 and 3 months postpartum, Nell-1 inactivation resulted in dwarfism and premature osteoporotic phenotypes. Specifically, Nell-1Col2α1 KO femurs and tibias exhibited significantly reduced length, bone mineral density (BMD), bone volume per tissue volume (BV/TV), trabecular number/thickness, cortical volume/thickness/density, and increased trabecular separation. The decreased bone formation rate revealed by dynamic histomorphometry was associated with altered numbers and/or function of osteoblasts and osteoclasts. Furthermore, longitudinal observations by in vivo micro-CT showed delayed and reduced mineralization at secondary ossification centers in mutants. Histologically, reduced staining intensities of Safranin O, Col-2, Col-10, and fewer BrdU-positive chondrocytes were observed in thinner Nell-1Col2α1 KO epiphyseal plates along with altered distribution and weaker expression level of Ihh, Patched-1, PTHrP, and PTHrP receptor. Primary Nell-1Col2α1 KO chondrocytes also exhibited decreased proliferation and differentiation, and its downregulated expression of the Ihh-PTHrP signaling molecules can be partially rescued by exogenous Nell-1 protein. Moreover, intranuclear Gli-1 protein and gene expression of the Gli-1 downstream target genes, Hip-1 and N-Myc, were also significantly decreased with Nell-1 inactivation. Notably, the rescue effects were diminished/reduced with application of Ihh signaling inhibitors, cyclopamine or GANT61. Taken together, these findings suggest that Nell-1 is a pivotal modulator of epiphyseal homeostasis and endochondral ossification. The cumulative chondrocyte-specific Nell-1 inactivation significantly impedes appendicular skeletogenesis resulting in dwarfism and premature osteoporosis through inhibiting Ihh signaling and predominantly altering the Ihh-PTHrP feedback loop. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Huichuan Qi
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xiaoyan Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Yao Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jiayi Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Hsin Chuan Pan
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Mengliu Yu
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Center of Stomatology, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Chaoyang District, Beijing, P. R. China
| | - Yasamin Mohazeb
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Sophia Azer
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Lloyd Baik
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jin Hee Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Min Hu
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
155
|
Micrornas at the Interface between Osteogenesis and Angiogenesis as Targets for Bone Regeneration. Cells 2019; 8:cells8020121. [PMID: 30717449 PMCID: PMC6406308 DOI: 10.3390/cells8020121] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
Bone formation and regeneration is a multistep complex process crucially determined by the formation of blood vessels in the growth plate region. This is preceded by the expression of growth factors, notably the vascular endothelial growth factor (VEGF), secreted by osteogenic cells, as well as the corresponding response of endothelial cells, although the exact mechanisms remain to be clarified. Thereby, coordinated coupling between osteogenesis and angiogenesis is initiated and sustained. The precise interplay of these two fundamental processes is crucial during times of rapid bone growth or fracture repair in adults. Deviations in this balance might lead to pathologic conditions such as osteoarthritis and ectopic bone formation. Besides VEGF, the recently discovered important regulatory and modifying functions of microRNAs also support this key mechanism. These comprise two principal categories of microRNAs that were identified with specific functions in bone formation (osteomiRs) and/or angiogenesis (angiomiRs). However, as hypoxia is a major driving force behind bone angiogenesis, a third group involved in this process is represented by hypoxia-inducible microRNAs (hypoxamiRs). This review was focused on the identification of microRNAs that were found to have an active role in osteogenesis as well as angiogenesis to date that were termed "CouplingmiRs (CPLGmiRs)". Outlined representatives therefore represent microRNAs that already have been associated with an active role in osteogenic-angiogenic coupling or are presumed to have its potential. Elucidation of the molecular mechanisms governing bone angiogenesis are of great relevance for improving therapeutic options in bone regeneration, tissue-engineering, and the treatment of bone-related diseases.
Collapse
|
156
|
Tang S, Xie Z, Wang P, Li J, Wang S, Liu W, Li M, Wu X, Su H, Cen S, Ye G, Zheng G, Wu Y, Shen H. LncRNA-OG Promotes the Osteogenic Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells Under the Regulation of hnRNPK. Stem Cells 2019; 37:270-283. [PMID: 30372559 PMCID: PMC7379496 DOI: 10.1002/stem.2937] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 09/19/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are the main source of osteoblasts in vivo and are widely used in stem cell therapy. Previously, we analyzed long noncoding RNA (lncRNA) expression profiles during BM-MSC osteogenesis, and further investigation is needed to elucidate how lncRNAs regulate BM-MSC osteogenesis. Herein, we used customized microarrays to determine lncRNA expression profiles in BM-MSCs on days 0 and 10 of osteogenic differentiation. In addition, we identified a novel osteogenesis-associated lncRNA (lncRNA-OG) that is upregulated during this process. Functional assays showed that lncRNA-OG significantly promotes BM-MSC osteogenesis. Mechanistically, lncRNA-OG interacts with heterogeneous nuclear ribonucleoprotein K (hnRNPK) protein to regulate bone morphogenetic protein signaling pathway activation. Surprisingly, hnRNPK positively regulates lncRNA-OG transcriptional activity by promoting H3K27 acetylation of the lncRNA-OG promoter. Therefore, our study revealed a novel lncRNA with a positive function on BM-MSC osteogenic differentiation and proposed a new interaction between hnRNPK and lncRNA. Stem Cells 2018 Stem Cells 2019;37:270-283.
Collapse
Affiliation(s)
- Su'an Tang
- Department of OrthopedicsThe Eighth Affiliated Hospital, Sun Yat‐sen UniversityShenzhenPeople's Republic of China
- Department of OrthopedicsZhujiang Hospital, Southern Medical UniversityGuangzhouPeople's Republic of China
| | - Zhongyu Xie
- Department of OrthopedicsThe Eighth Affiliated Hospital, Sun Yat‐sen UniversityShenzhenPeople's Republic of China
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Peng Wang
- Department of OrthopedicsThe Eighth Affiliated Hospital, Sun Yat‐sen UniversityShenzhenPeople's Republic of China
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Jinteng Li
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Shan Wang
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Wenjie Liu
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Ming Li
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Xiaohua Wu
- Center for BiotherapySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Hongjun Su
- Center for BiotherapySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Shuizhong Cen
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Guiwen Ye
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Guan Zheng
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Yanfeng Wu
- Center for BiotherapySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Huiyong Shen
- Department of OrthopedicsThe Eighth Affiliated Hospital, Sun Yat‐sen UniversityShenzhenPeople's Republic of China
- Department of OrthopedicsSun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
157
|
New Insights into the Interplay between Non-Coding RNAs and RNA-Binding Protein HnRNPK in Regulating Cellular Functions. Cells 2019; 8:cells8010062. [PMID: 30658384 PMCID: PMC6357021 DOI: 10.3390/cells8010062] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/08/2019] [Accepted: 01/15/2019] [Indexed: 12/15/2022] Open
Abstract
The emerging data indicates that non-coding RNAs (ncRNAs) epresent more than the “junk sequences” of the genome. Both miRNAs and long non-coding RNAs (lncRNAs) are involved in fundamental biological processes, and their deregulation may lead to oncogenesis and other diseases. As an important RNA-binding protein (RBP), heterogeneous nuclear ribonucleoprotein K (hnRNPK) is known to regulate gene expression through the RNA-binding domain involved in various pathways, such as transcription, splicing, and translation. HnRNPK is a highly conserved gene that is abundantly expressed in mammalian cells. The interaction of hnRNPK and ncRNAs defines the novel way through which ncRNAs affect the expression of protein-coding genes and form autoregulatory feedback loops. This review summarizes the interactions of hnRNPK and ncRNAs in regulating gene expression at transcriptional and post-transcriptional levels or by changing the genomic structure, highlighting their involvement in carcinogenesis, glucose metabolism, stem cell differentiation, virus infection and other cellular functions. Drawing connections between such discoveries might provide novel targets to control the biological outputs of cells in response to different stimuli.
Collapse
|
158
|
Soper JR, Bonar SF, O’Sullivan DJ, McCredie J, Willert HG. Thalidomide and neurotrophism. Skeletal Radiol 2019; 48:517-525. [PMID: 30341712 PMCID: PMC6394469 DOI: 10.1007/s00256-018-3086-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND Following the thalidomide disaster (1958-62), Henkel and Willert analysed the pattern of dysmelia in the long bones (J Bone Joint Surg Br. 51:399-414, 1969) and the extremities, Willert and Henkel (Z Orthop Ihre Grenzgeb. 107:663-75, 1970). Willert's material from deformed extremities is re-examined here asking "How does thalidomide reduce the skeleton?" MATERIALS AND METHODS We reviewed the original data collection of Willert and Henkel (Z Orthop Ihre Grenzgeb. 107:663-75, 1970), comprising musculoskeletal histology slides from 30 children affected by thalidomide with radiographs of hands (19 cases) and feet (4 cases). RESULTS All original observations by Willert and Henkel (Z Orthop Ihre Grenzgeb. 107:663-75, 1970), were verified. Radial rays of the hand disappeared early, but the foot was spared until late. Radiology confirms that bone reduction in the hand (aplasia or hypoplasia in the thumb and index finger) coincides with sensory segmental nerve C6. In the foot, reduction of the toes is rare, but mesenchymal excess (polydactyly) occurs in the hallux (L5 sclerotome), usually associated with absent tibia (L4 sclerotome). Histology confirms skeletal mesenchymal components to be unremarkable, contrasting with grossly abnormal bony architecture, a striking discordance between microscopic and macroscopic findings. No necrosis or vascular pathology was seen. CONCLUSION The basic lesion was an abnormal quantity rather than quality of mesenchyme. Cell populations result from cellular proliferation, controlled in early limb bud formation by neurotrophism. Thalidomide is a known sensory neurotoxin in adults. In the embryo, sensorineural injury alters neurotrophism, causing increased or diminished cell proliferation in undifferentiated mesenchyme. Differentiation into normal cartilage occurs later, but within an altered mesenchymal mass. Reduction or excess deformity results, with normal histology, a significant finding. The primary pathological condition is not in the skeleton, but in the nerves.
Collapse
Affiliation(s)
- Judith R. Soper
- Diagnostic Radiology, Royal Prince Alfred Hospital, Missenden Rd, Camperdown, Sydney, NSW Australia ,Specialist Magnetic Resonance Imaging Carillon Ave Newtown, Sydney, NSW Australia
| | - S. Fiona Bonar
- Douglass Hanly Moir, Giffnock Ave Macquarie Park, Sydney, NSW Australia ,Anatomical Pathology Royal Prince Alfred Hospital, Missenden Rd Camperdown Sydney, Sydney, NSW Australia
| | | | - Janet McCredie
- Diagnostic Radiology, Faculty of Medicine University of Sydney, Sydney, NSW Australia
| | | |
Collapse
|
159
|
Cui J, Zhang W, Huang E, Wang J, Liao J, Li R, Yu X, Zhao C, Zeng Z, Shu Y, Zhang R, Yan S, Lei J, Yang C, Wu K, Wu Y, Huang S, Ji X, Li A, Gong C, Yuan C, Zhang L, Liu W, Huang B, Feng Y, An L, Zhang B, Dai Z, Shen Y, Luo W, Wang X, Huang A, Luu HH, Reid RR, Wolf JM, Thinakaran G, Lee MJ, He TC. BMP9-induced osteoblastic differentiation requires functional Notch signaling in mesenchymal stem cells. J Transl Med 2019; 99:58-71. [PMID: 30353129 PMCID: PMC6300564 DOI: 10.1038/s41374-018-0087-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/28/2018] [Accepted: 05/14/2018] [Indexed: 01/12/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitors that can differentiate into multiple lineages including osteoblastic lineage. Osteogenic differentiation of MSCs is a cascade that recapitulates most, if not all, of the molecular events occurring during embryonic skeletal development, which is regulated by numerous signaling pathways including bone morphogenetic proteins (BMPs). Through a comprehensive analysis of the osteogenic activity, we previously demonstrated that BMP9 is the most potent BMP for inducing bone formation from MSCs both in vitro and in vivo. However, as one of the least studied BMPs, the essential mediators of BMP9-induced osteogenic signaling remain elusive. Here we show that BMP9-induced osteogenic signaling in MSCs requires intact Notch signaling. While the expression of Notch receptors and ligands are readily detectable in MSCs, Notch inhibitor and dominant-negative Notch1 effectively inhibit BMP9-induced osteogenic differentiation in vitro and ectopic bone formation in vivo. Genetic disruption of Notch pathway severely impairs BMP9-induced osteogenic differentiation and ectopic bone formation from MSCs. Furthermore, while BMP9-induced expression of early-responsive genes is not affected by defective Notch signaling, BMP9 upregulates the expression of Notch receptors and ligands at the intermediate stage of osteogenic differentiation. Taken together, these results demonstrate that Notch signaling may play an essential role in coordinating BMP9-induced osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Jing Cui
- grid.412461.4Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China ,0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Wenwen Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA. .,Stem Cell Research Laboratory, Department of Obstetrics and Gynecology, the Affiliated University-Town Hospital, Chongqing Medical University, 401331, Chongqing, China.
| | - Enyi Huang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Jia Wang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Stem Cell Research Laboratory, Department of Obstetrics and Gynecology, the Affiliated University-Town Hospital, Chongqing Medical University, 401331 Chongqing, China
| | - Junyi Liao
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Ruidong Li
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Xinyi Yu
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Chen Zhao
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Zongyue Zeng
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Yi Shu
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Ruyi Zhang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Shujuan Yan
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Jiayan Lei
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Chao Yang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Ke Wu
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Ying Wu
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0001 1431 9176grid.24695.3cDepartment of Immunology and Microbiology, Beijing University of Chinese Medicine, 100029 Beijing, China
| | - Shifeng Huang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Xiaojuan Ji
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Alexander Li
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Cheng Gong
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,grid.413247.7Department of Surgery, the Affiliated Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Chengfu Yuan
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0001 0033 6389grid.254148.eDepartment of Biochemistry and Molecular Biology, China Three Gorges University School of Medicine, 443002 Yichang, China
| | - Linghuan Zhang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Wei Liu
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Bo Huang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China ,grid.412455.3Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, China
| | - Yixiao Feng
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Liping An
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0004 1798 9345grid.411294.bKey Laboratory of Orthopaedic Surgery of Gansu Province and the Department of Orthopaedic Surgery, the Second Hospital of Lanzhou University, 730030 Lanzhou, China
| | - Bo Zhang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0004 1798 9345grid.411294.bKey Laboratory of Orthopaedic Surgery of Gansu Province and the Department of Orthopaedic Surgery, the Second Hospital of Lanzhou University, 730030 Lanzhou, China
| | - Zhengyu Dai
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,Department of Orthopaedic Surgery, Chongqing Hospital of Traditional Chinese Medicine, 400021 Chongqing, China
| | - Yi Shen
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0004 1803 0208grid.452708.cDepartment of Orthopaedic Surgery, Xiangya Second Hospital of Central South University, 410011 Changsha, China
| | - Wenping Luo
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Xi Wang
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8653 0555grid.203458.8Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016 Chongqing, China
| | - Ailong Huang
- grid.412461.4Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hue H. Luu
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Russell R. Reid
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA ,0000 0000 8736 9513grid.412578.dDepartment of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Jennifer Moriatis Wolf
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Gopal Thinakaran
- 0000 0000 8736 9513grid.412578.dDepartment of Neurobiology, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Michael J. Lee
- 0000 0000 8736 9513grid.412578.dMolecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637 USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA. .,Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
160
|
Wan Y, Lantz B, Cusack BJ, Szabo-Rogers HL. Prickle1 regulates differentiation of frontal bone osteoblasts. Sci Rep 2018; 8:18021. [PMID: 30575813 PMCID: PMC6303328 DOI: 10.1038/s41598-018-36742-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/27/2018] [Indexed: 11/08/2022] Open
Abstract
Enlarged fontanelles and smaller frontal bones result in a mechanically compromised skull. Both phenotypes could develop from defective migration and differentiation of osteoblasts in the skull bone primordia. The Wnt/Planar cell polarity (Wnt/PCP) signaling pathway regulates cell migration and movement in other tissues and led us to test the role of Prickle1, a core component of the Wnt/PCP pathway, in the skull. For these studies, we used the missense allele of Prickle1 named Prickle1Beetlejuice (Prickle1Bj). The Prickle1Bj/Bj mutants are microcephalic and develop enlarged fontanelles between insufficient frontal bones, while the parietal bones are normal. Prickle1Bj/Bj mutants have several other craniofacial defects including a midline cleft lip, incompletely penetrant cleft palate, and decreased proximal-distal growth of the head. We observed decreased Wnt/β-catenin and Hedgehog signaling in the frontal bone condensations of the Prickle1Bj/Bj mutants. Surprisingly, the smaller frontal bones do not result from defects in cell proliferation or death, but rather significantly delayed differentiation and decreased expression of migratory markers in the frontal bone osteoblast precursors. Our data suggests that Prickle1 protein function contributes to both the migration and differentiation of osteoblast precursors in the frontal bone.
Collapse
Affiliation(s)
- Yong Wan
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brandi Lantz
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J Cusack
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heather L Szabo-Rogers
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
161
|
Abstract
During cartilage development chondrocytes undergo a multi-step process characterized by consecutive changes in cell morphology and gene expression. Cell proliferation, polarity, differentiation, and migration are influenced by chemical and mechanical signaling between the extracellular matrix (ECM) and the cell. Several structurally diverse transmembrane receptors such as integrins, discoidin domain receptor 2 (DDR 2), and CD44 mediate the crosstalk between cells and their ECM. However, the contribution of cell-matrix interactions during early chondrogenesis and further cartilage development through cell receptors and their signal transduction pathways is still not fully understood. Determination of receptor signaling pathways and the function of downstream targets will aid in a better understanding of musculoskeletal pathologies such as chondrodysplasia, and the development of new approaches for the treatment of cartilage disorders. We will summarize recent findings, linking cell receptors and their potential signaling pathways to the control of chondrocyte behavior during early chondrogenesis and endochondral ossification.
Collapse
Affiliation(s)
- Carina Prein
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, and Western University Bone and Joint Institute, University of Western Ontario, London, ON, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, and Western University Bone and Joint Institute, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
162
|
Abstract
It is from the discovery of leptin and the central nervous system as a regulator of bone remodeling that the presence of autonomic nerves within the skeleton transitioned from a mere histological observation to the mechanism whereby neurons of the central nervous system communicate with cells of the bone microenvironment and regulate bone homeostasis. This shift in paradigm sparked new preclinical and clinical investigations aimed at defining the contribution of sympathetic, parasympathetic, and sensory nerves to the process of bone development, bone mass accrual, bone remodeling, and cancer metastasis. The aim of this article is to review the data that led to the current understanding of the interactions between the autonomic and skeletal systems and to present a critical appraisal of the literature, bringing forth a schema that can put into physiological and clinical context the main genetic and pharmacological observations pointing to the existence of an autonomic control of skeletal homeostasis. The different types of nerves found in the skeleton, their functional interactions with bone cells, their impact on bone development, bone mass accrual and remodeling, and the possible clinical or pathophysiological relevance of these findings are discussed.
Collapse
Affiliation(s)
- Florent Elefteriou
- Department of Molecular and Human Genetics and Orthopedic Surgery, Center for Skeletal Medicine and Biology, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
163
|
Beck L. Expression and function of Slc34 sodium-phosphate co-transporters in skeleton and teeth. Pflugers Arch 2018; 471:175-184. [PMID: 30511265 DOI: 10.1007/s00424-018-2240-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022]
Abstract
Under normal physiological condition, the biomineralization process is limited to skeletal tissues and teeth and occurs throughout the individual's life. Biomineralization is an actively regulated process involving the progressive mineralization of the extracellular matrix secreted by osteoblasts in bone or odontoblasts and ameloblasts in tooth. Although the detailed molecular mechanisms underlying the formation of calcium-phosphate apatite crystals are still debated, it is suggested that calcium and phosphate may need to be transported across the membrane of the mineralizing cell, suggesting a pivotal role of phosphate transporters in bone and tooth mineralization. In this context, this short review describes the current knowledge on the role of Slc34 Na+-phosphate transporters in skeletal and tooth mineralization.
Collapse
Affiliation(s)
- Laurent Beck
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Faculté de Chirurgie Dentaire, Université de Nantes, ONIRIS, 1 place Alexis Ricordeau, 44042, Nantes, France. .,Université de Nantes, UFR Odontologie, 44042, Nantes, France.
| |
Collapse
|
164
|
Kimura T, Ozaki T, Fujita K, Yamashita A, Morioka M, Ozono K, Tsumaki N. Proposal of patient-specific growth plate cartilage xenograft model for FGFR3 chondrodysplasia. Osteoarthritis Cartilage 2018; 26:1551-1561. [PMID: 30086379 DOI: 10.1016/j.joca.2018.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/12/2018] [Accepted: 07/21/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE FGFR3 chondrodysplasia is caused by a gain-of-function mutation of the FGFR3 gene. The disease causes abnormal growth plate cartilage and lacks effective drug treatment. We sought to establish an in vivo model for the study of FGFR3 chondrodysplasia pathology and drug testing. DESIGN We created cartilage from human induced pluripotent stem cells (hiPSCs) and transplanted the cartilage into the subcutaneous spaces of immunodeficient mice. We then created cartilage from the hiPSCs of patients with FGFR3 chondrodysplasia and transplanted them into immunodeficient mice. We treated some mice with a FGFR inhibitor after the transplantation. RESULTS Xenografting the hiPSC-derived cartilage reproduced human growth plate cartilage consisting of zones of resting, proliferating, prehypertrophic and hypertrophic chondrocytes and bone in immunodeficient mice. Immunohistochemistry of xenografts using anti-human nuclear antigen antibody indicated that all chondrocytes in growth plate cartilage were human, whereas bone was composed of human and mouse cells. The pathology of small hypertrophic chondrocytes due to up-regulated FGFR3 signaling in FGFR3 skeletal dysplasia was recapitulated in growth plate cartilage formed in the xenografts of patient-specific hiPSC-derived cartilage. The mean diameters of hypertrophic chondrocytes between wild type and thanatophoric dysplasia were significantly different (95% CI: 13.2-26.9; n = 4 mice, one-way analysis of variance (ANOVA)). The pathology was corrected by systemic administration of a FGFR inhibitor to the mice. CONCLUSION The patient-specific growth plate cartilage xenograft model for FGFR3 skeletal dysplasia indicated recapitulation of pathology and effectiveness of a FGFR inhibitor for treatment and warrants more study for its usefulness to study disease pathology and drug testing.
Collapse
Affiliation(s)
- T Kimura
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan; Department of Pediatrics, Osaka University Graduate School of Medicine, Japan
| | - T Ozaki
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - K Fujita
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - A Yamashita
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - M Morioka
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - K Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Japan
| | - N Tsumaki
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan.
| |
Collapse
|
165
|
Schliermann A, Nickel J. Unraveling the Connection between Fibroblast Growth Factor and Bone Morphogenetic Protein Signaling. Int J Mol Sci 2018; 19:ijms19103220. [PMID: 30340367 PMCID: PMC6214098 DOI: 10.3390/ijms19103220] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Ontogeny of higher organisms as well the regulation of tissue homeostasis in adult individuals requires a fine-balanced interplay of regulating factors that individually trigger the fate of particular cells to either stay undifferentiated or to differentiate towards distinct tissue specific lineages. In some cases, these factors act synergistically to promote certain cellular responses, whereas in other tissues the same factors antagonize each other. However, the molecular basis of this obvious dual signaling activity is still only poorly understood. Bone morphogenetic proteins (BMPs) and fibroblast growth factors (FGFs) are two major signal protein families that have a lot in common: They are both highly preserved between different species, involved in essential cellular functions, and their ligands vastly outnumber their receptors, making extensive signal regulation necessary. In this review we discuss where and how BMP and FGF signaling cross paths. The compiled data reflect that both factors synchronously act in many tissues, and that antagonism and synergism both exist in a context-dependent manner. Therefore, by challenging a generalization of the connection between these two pathways a new chapter in BMP FGF signaling research will be introduced.
Collapse
Affiliation(s)
- Anna Schliermann
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
| | - Joachim Nickel
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
- Fraunhofer Institut für Silicatforschung, Translationszentrum TLZ-RT, Röntgenring 11, 97222 Würzburg, Germany.
| |
Collapse
|
166
|
Wang L, Jia H, Tower RJ, Levine MA, Qin L. Analysis of short-term treatment with the phosphodiesterase type 5 inhibitor tadalafil on long bone development in young rats. Am J Physiol Endocrinol Metab 2018; 315:E446-E453. [PMID: 29920215 PMCID: PMC6230700 DOI: 10.1152/ajpendo.00130.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclic GMP (cGMP) is an important intracellular regulator of endochondral bone growth and skeletal remodeling. Tadalafil, an inhibitor of the phosphodiesterase (PDE) type 5 (PDE5) that specifically hydrolyzes cGMP, is increasingly used to treat children with pulmonary arterial hypertension (PAH), but the effect of tadalafil on bone growth and strength has not been previously investigated. In this study, we first analyzed the expression of transcripts encoding PDEs in primary cultures of chondrocytes from newborn rat epiphyses. We detected robust expression of PDE5 as the major phosphodiesterase hydrolyzing cGMP. Time-course experiments showed that C-type natriuretic peptide increased intracellular levels of cGMP in primary chondrocytes with a peak at 2 min, and in the presence of tadalafil the peak level of intracellular cGMP was 37% greater ( P < 0.01) and the decline was significantly attenuated. Next, we treated 1-mo-old Sprague Dawley rats with vehicle or tadalafil for 3 wk. Although 10 mg·kg-1·day-1 tadalafil led to a significant 52% ( P < 0.01) increase in tissue levels of cGMP and a 9% reduction ( P < 0.01) in bodyweight gain, it did not alter long bone length, cortical or trabecular bone properties, and histological features. In conclusion, our results indicate that PDE5 is highly expressed in growth plate chondrocytes, and short-term tadalafil treatment of growing rats at doses comparable to those used in children with PAH has neither obvious beneficial effect on long bone growth nor any observable adverse effect on growth plate structure and trabecular and cortical bone structure.
Collapse
Affiliation(s)
- Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- Department of Orthopaedics, Shandong University Qilu Hospital, Shandong University , Jinan , China
| | - Haoruo Jia
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- Department of Orthopaedics, The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, China
| | - Robert J Tower
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Michael A Levine
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- Division of Endocrinology and Diabetes and the Center for Bone Health, The Children's Hospital of Philadelphia , Philadelphia, Pennsylvania
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
167
|
Onofrillo C, Duchi S, O'Connell CD, Blanchard R, O'Connor AJ, Scott M, Wallace GG, Choong PFM, Di Bella C. Biofabrication of human articular cartilage: a path towards the development of a clinical treatment. Biofabrication 2018; 10:045006. [PMID: 30088479 DOI: 10.1088/1758-5090/aad8d9] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cartilage injuries cause pain and loss of function, and if severe may result in osteoarthritis (OA). 3D bioprinting is now a tangible option for the delivery of bioscaffolds capable of regenerating the deficient cartilage tissue. Our team has developed a handheld device, the Biopen, to allow in situ additive manufacturing during surgery. Given its ability to extrude in a core/shell manner, the Biopen can preserve cell viability during the biofabrication process, and it is currently the only biofabrication tool tested as a surgical instrument in a sheep model using homologous stem cells. As a necessary step toward the development of a clinically relevant protocol, we aimed to demonstrate that our handheld extrusion device can successfully be used for the biofabrication of human cartilage. Therefore, this study is a required step for the development of a surgical treatment in human patients. In this work we specifically used human adipose derived mesenchymal stem cells (hADSCs), harvested from the infra-patellar fat pad of donor patients affected by OA, to also prove that they can be utilized as the source of cells for the future clinical application. With the Biopen, we generated bioscaffolds made of hADSCs laden in gelatin methacrylate, hyaluronic acid methacrylate and cultured in the presence of chondrogenic stimuli for eight weeks in vitro. A comprehensive characterisation including gene and protein expression analyses, immunohistology, confocal microscopy, second harmonic generation, light sheet imaging, atomic force mycroscopy and mechanical unconfined compression demonstrated that our strategy resulted in human hyaline-like cartilage formation. Our in situ biofabrication approach represents an innovation with important implications for customizing cartilage repair in patients with cartilage injuries and OA.
Collapse
Affiliation(s)
- Carmine Onofrillo
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, VIC, Australia. ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, Innovation Campus, University of Wollongong, NSW, Australia. BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital, Melbourne, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Bo X, Wu M, Xiao H, Wang H. Transcriptome analyses reveal molecular mechanisms that regulate endochondral ossification in amphibian Bufo gargarizans during metamorphosis. Biochim Biophys Acta Gen Subj 2018; 1862:2632-2644. [PMID: 30076880 DOI: 10.1016/j.bbagen.2018.07.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND A developmental transition from aquatic to terrestrial existence is one of the most important events in the evolution of terrestrial vertebrates. Amphibian metamorphosis is a classic model to study this transition. The development of the vertebrate skeleton can reflect its evolutionary history. Endochondral ossification serves a vital role in skeletal development. Thus, we sought to unravel molecular mechanisms that regulate endochondral ossification during Bufo gargarizans metamorphosis. METHODS The alizarin red-alcian blue double staining method was used to visualize the skeletal development of B. gargarizans during metamorphosis. RNA sequencing (RNA-seq) was used to explore the transcriptome of B. gargarizans in four key developmental stages during metamorphosis. Real-time quantitative PCR (RT-qPCR) was used to validate the expression patterns of endochondral ossification related genes. RESULTS Endochondral ossification increased gradually in skeletal system of B. gargarizans during metamorphosis. A total of 137,264 unigenes were assembled and 44,035 unigenes were annotated. 10,352 differentially expressed genes (DEGs) were further extracted among four key developmental stages. In addition, 28 endochondral ossification related genes were found by searching for DEG libraries in B. gargarizans. Of the 28 genes, 10 genes were validated using RT-qPCR. CONCLUSIONS The exquisite coordination of the 28 genes is essential for regulation of endochondral ossification during B. gargarizans metamorphosis. GENERAL SIGNIFICANCE The present study will not only provide an invaluable genomic resource and background for further research of endochondral ossification in amphibians but will also aid in enhancing our understanding of the evolution of terrestrial vertebrates.
Collapse
Affiliation(s)
- Xiaoxue Bo
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Minyao Wu
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Hui Xiao
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China.
| |
Collapse
|
169
|
Anterior Skull Base and Pericranial Flap Ossification after Frontofacial Monobloc Advancement. Plast Reconstr Surg 2018; 141:437-445. [PMID: 29036029 DOI: 10.1097/prs.0000000000004040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Frontofacial monobloc advancement creates a communication between the anterior cranial fossa and nasal cavities. To tackle this issue, transorbital pericranial pedicled flaps are routinely performed in the authors' center. This study aimed to assess the postoperative ossification of the anterior skull base and pedicled flaps following frontofacial monobloc advancement, and to identify factors influencing this ossification. METHODS Measurements of the skull base only and of the ossified pedicled flaps together with the skull base were performed on computed tomographic scans at the nasofrontal and the nasoethmoid frontal junctions. The total thickness of the skull vault was measured and a qualitative defect score for the anterior skull base was computed. RESULTS Twenty-two patients who underwent frontofacial monobloc advancement at a median age of 3.1 years (range, 1.9 to 3.6 years) were included: 14 with Crouzon, five with Pfeiffer, and three with Apert syndrome. One year and 5 years after surgery, the distraction gap was completely ossified in the anterior skull base midline in all patients. Ossified pedicled flaps together with the skull base were thicker in patients than in controls at these two time points (p < 0.005 and p < 0.02). Patients with Pfeiffer syndrome had a significantly thicker skull base only and ossified pedicled flaps together with the skull base thicknesses (p = 0.01 and p = 0.03) and lower defect scores than patients with Crouzon or Apert syndrome (p = 0.03) 1 year postoperatively. CONCLUSION As ossification of the pedicled flaps and total reossification of the anterior skull base midline were observed in all patients, the authors indicate that performing pedicled flaps in frontofacial monobloc advancement surgery could promote the reossification of the anterior skull base. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, IV.
Collapse
|
170
|
Hu H, Zhao X, Ma J, Shangguan Y, Pan Z, Chen L, Zhang X, Wang H. Prenatal nicotine exposure retards osteoclastogenesis and endochondral ossification in fetal long bones in rats. Toxicol Lett 2018; 295:249-255. [PMID: 29981921 DOI: 10.1016/j.toxlet.2018.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/06/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
This study investigated the mechanisms underlying the retarded development of long bone in fetus by prenatal nicotine exposure (PNE) which had been demonstrated by our previous work. Nicotine (2.0 mg/kg.d) or saline was injected subcutaneously into pregnant rats every morning from gestational day (GD) 9 to 20. Fetal femurs or tibias were harvested for analysis on GD 20. We found massive accumulation of hypertrophic chondrocytes and a delayed formation of primary ossification center (POC) in the fetal femur or tibia of rat fetus after PNE, which was accompanied by a decreased amount of osteoclasts in the POC and up-regulated expression of osteoprotegerin (OPG) but by no obvious change in the expression of receptor activator of NF-κB ligand (RANKL). In primary osteoblastic cells, both nicotine (0, 162, 1620, 16,200 ng/ml) and corticosterone (0, 50, 250, 1250 nM) promoted the mRNA expression of OPG but concentration-dependently suppressed that of RANKL. Furthermore, blocking α4β2-nicotinic acetylcholine receptor (α4β2-nAChR) or glucocorticoid receptor rescued the above effects of nicotine and corticosterone, respectively. In conclusion, retarded osteoclastogenesis may contribute to delayed endochondral ossification in long bone in fetal rats with PNE. The adverse effects of PNE may be mediated via the direct effect of nicotine and indirect effect of maternal corticosterone on osteoblastic cells.
Collapse
Affiliation(s)
- Hang Hu
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Physiology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Xin Zhao
- Department of Physiology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Jing Ma
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Physiology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Yangfan Shangguan
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhengqi Pan
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liaobin Chen
- Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xianrong Zhang
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
171
|
Eskinazi-Budge A, Manickavasagam D, Czech T, Novak K, Kunzler J, Oyewumi MO. Preparation of emulsifying wax/glyceryl monooleate nanoparticles and evaluation as a delivery system for repurposing simvastatin in bone regeneration. Drug Dev Ind Pharm 2018; 44:1583-1590. [DOI: 10.1080/03639045.2018.1483381] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Aaron Eskinazi-Budge
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Dharani Manickavasagam
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
- Department of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Tori Czech
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Kimberly Novak
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - James Kunzler
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Moses O. Oyewumi
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
- Department of Biomedical Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
172
|
Ikehata M, Yamada A, Fujita K, Yoshida Y, Kato T, Sakashita A, Ogata H, Iijima T, Kuroda M, Chikazu D, Kamijo R. Cooperation of Rho family proteins Rac1 and Cdc42 in cartilage development and calcified tissue formation. Biochem Biophys Res Commun 2018; 500:525-529. [DOI: 10.1016/j.bbrc.2018.04.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/04/2018] [Indexed: 01/14/2023]
|
173
|
Vas WJ, Shah M, Blacker TS, Duchen MR, Sibbons P, Roberts SJ. Decellularized Cartilage Directs Chondrogenic Differentiation: Creation of a Fracture Callus Mimetic. Tissue Eng Part A 2018; 24:1364-1376. [PMID: 29580181 DOI: 10.1089/ten.tea.2017.0450] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Complications that arise from impaired fracture healing have considerable socioeconomic implications. Current research in the field of bone tissue engineering predominantly aims to mimic the mature bone tissue microenvironment. This approach, however, may produce implants that are intrinsically unresponsive to the cues present during the initiation of fracture repair. As such, this study describes the development of decellularized xenogeneic hyaline cartilage matrix in an attempt to mimic the initial reparative phase of fracture repair. Three approaches based on vacuum-assisted osmotic shock (Vac-OS), Triton X-100 (Vac-STx), and sodium dodecyl sulfate (Vac-SDS) were investigated. The Vac-OS methodology reduced DNA content below 50 ng/mg of tissue, while retaining 85% of the sulfate glycosaminoglycan content, and as such was selected as the optimal methodology for decellularization. The resultant Vac-OS scaffolds (decellularized extracellular matrix [dcECM]) were also devoid of the immunogenic alpha-Gal epitope. Furthermore, minimal disruption to the structural integrity of the dcECM was demonstrated using differential scanning calorimetry and fluorescence lifetime imaging microscopy. The biological integrity of the dcECM was confirmed by its ability to drive the chondrogenic commitment and differentiation of human chondrocytes and periosteum-derived cells, respectively. Furthermore, histological examination of dcECM constructs implanted in immunocompetent mice revealed a predominantly M2 macrophage-driven regenerative response both at 2 and 8 weeks postimplantation. These findings contrasted with the implanted native costal cartilage that elicited a predominantly M1 macrophage-mediated inflammatory response. This study highlights the capacity of dcECM from the Vac-OS methodology to direct the key biological processes of endochondral ossification, thus potentially recapitulating the callus phase of fracture repair.
Collapse
Affiliation(s)
- Wollis J Vas
- 1 Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London , Stanmore, United Kingdom
| | - Mittal Shah
- 1 Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London , Stanmore, United Kingdom
| | - Thomas S Blacker
- 2 Department of Cell and Developmental Biology, University College London , London, United Kingdom .,3 Department of Physics and Astronomy, University College London , London, United Kingdom
| | - Michael R Duchen
- 2 Department of Cell and Developmental Biology, University College London , London, United Kingdom
| | - Paul Sibbons
- 4 Northwick Park Institute for Medical Research , Northwick Park Hospital, London, United Kingdom
| | - Scott J Roberts
- 1 Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London , Stanmore, United Kingdom
| |
Collapse
|
174
|
Facial bone fragmentation in blind cavefish arises through two unusual ossification processes. Sci Rep 2018; 8:7015. [PMID: 29725043 PMCID: PMC5934472 DOI: 10.1038/s41598-018-25107-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/13/2018] [Indexed: 01/04/2023] Open
Abstract
The precise mechanisms underlying cranial bone development, evolution and patterning remain incompletely characterised. This poses a challenge to understanding the etiologies of craniofacial malformations evolving in nature. Capitalising on natural variation, “evolutionary model systems” provide unique opportunities to identify underlying causes of aberrant phenotypes as a complement to studies in traditional systems. Mexican blind cavefish are a prime evolutionary model for cranial disorders since they frequently exhibit extreme alterations to the skull and lateral asymmetries. These aberrations occur in stark contrast to the normal cranial architectures of closely related surface-dwelling fish, providing a powerful comparative paradigm for understanding cranial bone formation. Using a longitudinal and in vivo analytical approach, we discovered two unusual ossification processes in cavefish that underlie the development of ‘fragmented’ and asymmetric cranial bones. The first mechanism involves the sporadic appearance of independent bony elements that fail to fuse together later in development. The second mechanism involves the “carving” of channels in the mature bone, a novel form of post-ossification remodeling. In the extreme cave environment, these novel mechanisms may have evolved to augment sensory input, and may indirectly result in a trade-off between sensory expansion and cranial bone development.
Collapse
|
175
|
Yan S, Zhang R, Wu K, Cui J, Huang S, Ji X, An L, Yuan C, Gong C, Zhang L, Liu W, Feng Y, Zhang B, Dai Z, Shen Y, Wang X, Luo W, Liu B, Haydon RC, Lee MJ, Reid RR, Wolf JM, Shi Q, Luu HH, He TC, Weng Y. Characterization of the essential role of bone morphogenetic protein 9 (BMP9) in osteogenic differentiation of mesenchymal stem cells (MSCs) through RNA interference. Genes Dis 2018; 5:172-184. [PMID: 30258947 PMCID: PMC6149187 DOI: 10.1016/j.gendis.2018.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells and capable of differentiating into multiple cell types including osteoblastic, chondrogenic and adipogenic lineages. We previously identified BMP9 as one of the most potent BMPs that induce osteoblastic differentiation of MSCs although exact molecular mechanism through which BMP9 regulates osteogenic differentiation remains to be fully understood. Here, we seek to develop a recombinant adenovirus system to optimally silence mouse BMP9 and then characterize the important role of BMP9 in osteogenic differentiation of MSCs. Using two different siRNA bioinformatic prediction programs, we design five siRNAs targeting mouse BMP9 (or simB9), which are expressed under the control of the converging H1 and U6 promoters in recombinant adenovirus vectors. We demonstrate that two of the five siRNAs, simB9-4 and simB9-7, exhibit the highest efficiency on silencing exogenous mouse BMP9 in MSCs. Furthermore, simB9-4 and simB9-7 act synergistically in inhibiting BMP9-induced expression of osteogenic markers, matrix mineralization and ectopic bone formation from MSCs. Thus, our findings demonstrate the important role of BMP9 in osteogenic differentiation of MSCs. The characterized simB9 siRNAs may be used as an important tool to investigate the molecular mechanism behind BMP9 osteogenic signaling. Our results also indicate that recombinant adenovirus-mediated expression of siRNAs is efficient and sustained, and thus may be used as an effective delivery vehicle of siRNA therapeutics.
Collapse
Affiliation(s)
- Shujuan Yan
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ruyi Zhang
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ke Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jing Cui
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Shifeng Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xiaojuan Ji
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Liping An
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Key Laboratory of Orthopaedic Surgery of Gansu Province and the Department of Orthopaedic Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Chengfu Yuan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Biochemistry and Molecular Biology, China Three Gorges University School of Medicine, Yichang 443002, China
| | - Cheng Gong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Surgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Linghuan Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Wei Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yixiao Feng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Bo Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Key Laboratory of Orthopaedic Surgery of Gansu Province and the Department of Orthopaedic Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Zhengyu Dai
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Orthopaedic Surgery, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Yi Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Orthopaedic Surgery, Xiangya Second Hospital of Central South University, Changsha 410011, China
| | - Xi Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Wenping Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Bo Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,The School of Pharmacy and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Rex C Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Surgery, Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Qiong Shi
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yaguang Weng
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
176
|
Miao J, Wang X, Bao J, Jin S, Chang T, Xia J, Yang L, Zhu B, Xu L, Zhang L, Gao X, Chen Y, Li J, Gao H. Multimarker and rare variants genomewide association studies for bone weight in Simmental cattle. J Anim Breed Genet 2018; 135:159-169. [DOI: 10.1111/jbg.12326] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/27/2018] [Indexed: 12/30/2022]
Affiliation(s)
- J. Miao
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
- College of Animal Sciences; Fujian Agriculture and Forestry University; Fujian China
| | - X. Wang
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - J. Bao
- Veterinary Bureau of Wulagai Precinct in Xilin Gol League; Wulagai China
| | - S. Jin
- Veterinary Bureau of Wulagai Precinct in Xilin Gol League; Wulagai China
| | - T. Chang
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - J. Xia
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - L. Yang
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province; Sichuan Agricultural University; Sichuan China
| | - B. Zhu
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - L. Xu
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - L. Zhang
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - X. Gao
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - Y. Chen
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - J. Li
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| | - H. Gao
- Laboratory of Molecular Biology and Bovine Breeding; Institute of Animal Sciences; Chinese Academy of Agricultural Sciences; Beijing China
| |
Collapse
|
177
|
Pacifici M. Retinoid roles and action in skeletal development and growth provide the rationale for an ongoing heterotopic ossification prevention trial. Bone 2018; 109:267-275. [PMID: 28826842 PMCID: PMC8011837 DOI: 10.1016/j.bone.2017.08.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/15/2017] [Indexed: 12/30/2022]
Abstract
The majority of skeletal elements develop via endochondral ossification. This process starts with formation of mesenchymal cell condensations at prescribed sites and times in the early embryo and is followed by chondrogenesis, growth plate cartilage maturation and hypertrophy, and replacement of cartilage with bone and marrow. This complex stepwise process is reactivated and recapitulated in physiologic conditions such as fracture repair, but can occur extraskeletally in pathologies including heterotopic ossification (HO), Ossification of the Posterior Longitudinal Ligament (OPLL) and Hereditary Multiple Exostoses (HME). One form of HO is common and is triggered by trauma, invasive surgeries or burns and is thus particularly common amongst severely wounded soldiers. There is also a congenital and very severe form of HO that occurs in children with Fibrodysplasia Ossificans Progressiva (FOP) and is driven by activating mutations in ACVR1 encoding the type I bone morphogenetic protein (BMP) receptor ALK2. Current treatments for acquired HO, including NSAIDs and local irradiation, are not always effective and can have side effects, and there is no effective treatment for HO in FOP. This review article describes the research path we took several years ago to develop a new and effective treatment for both congenital and acquired forms of HO and specifically, the testing of synthetic retinoid agonists to block the initial and critical chondrogenic step leading to HO onset and progression. We summarize studies with mouse models of injury-induced and congenital HO demonstrating the effectiveness and mode of action of the retinoid agonists, including Palovarotene. Our studies have provided the rationale for, directly led to, an ongoing phase 2 FDA clinical trial to test efficacy and safety of Palovarotene in FOP. Top-line results released a few months ago by the pharmaceutical sponsor Clementia are very encouraging. Given shared developmental pathways amongst pathologies of extraskeletal tissue formation, Palovarotene may also be effective in HME as preliminary in vitro data suggest.
Collapse
Affiliation(s)
- Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States.
| |
Collapse
|
178
|
Wang Y, Liu JA, Leung KKH, Sham MH, Chan D, Cheah KSE, Cheung M. Reprogramming of Mouse Calvarial Osteoblasts into Induced Pluripotent Stem Cells. Stem Cells Int 2018; 2018:5280793. [PMID: 29721022 PMCID: PMC5867603 DOI: 10.1155/2018/5280793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/09/2018] [Indexed: 11/22/2022] Open
Abstract
Previous studies have demonstrated the ability of reprogramming endochondral bone into induced pluripotent stem (iPS) cells, but whether similar phenomenon occurs in intramembranous bone remains to be determined. Here we adopted fluorescence-activated cell sorting-based strategy to isolate homogenous population of intramembranous calvarial osteoblasts from newborn transgenic mice carrying both Osx1-GFP::Cre and Oct4-EGFP transgenes. Following retroviral transduction of Yamanaka factors (Oct4, Sox2, Klf4, and c-Myc), enriched population of osteoblasts underwent silencing of Osx1-GFP::Cre expression at early stage of reprogramming followed by late activation of Oct4-EGFP expression in the resulting iPS cells. These osteoblast-derived iPS cells exhibited gene expression profiles akin to embryonic stem cells and were pluripotent as demonstrated by their ability to form teratomas comprising tissues from all germ layers and also contribute to tail tissue in chimera embryos. These data demonstrate that iPS cells can be generated from intramembranous osteoblasts.
Collapse
Affiliation(s)
- Yinxiang Wang
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jessica Aijia Liu
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Keith K. H. Leung
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Mai Har Sham
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Danny Chan
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kathryn S. E. Cheah
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Martin Cheung
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
179
|
Bernhard JC, Hulphers E, Rieder B, Ferguson J, Rünzler D, Nau T, Redl H, Vunjak-Novakovic G. Perfusion Enhances Hypertrophic Chondrocyte Matrix Deposition, But Not the Bone Formation. Tissue Eng Part A 2018; 24:1022-1033. [PMID: 29373945 DOI: 10.1089/ten.tea.2017.0356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Perfusion bioreactors have been an effective tool in bone tissue engineering. Improved nutrient delivery and the application of shear forces have stimulated osteoblast differentiation and matrix production, allowing for generation of large, clinically sized constructs. Differentiation of hypertrophic chondrocytes has been considered an alternative strategy for bone tissue engineering. We studied the effects of perfusion on hypertrophic chondrocyte differentiation, matrix production, and subsequent bone formation. Hypertrophic constructs were created by differentiation in chondrogenic medium (2 weeks) and maturation in hypertrophic medium (3 weeks). Bioreactors were customized to study a range of flow rates (0-1200 μm/s). During chondrogenic differentiation, increased flow rates correlated with cartilage matrix deposition and the presence of collagen type X. During induced hypertrophic maturation, increased flow rates correlated with bone template deposition and the increased secretion of chondroprotective cytokines. Following an 8-week implantation into the critical-size femoral defect in nude rats, nonperfused constructs displayed larger bone volume, more compact mineralized matrix, and better integration with the adjacent native bone. Therefore, although medium perfusion stimulated the formation of bone template in vitro, it failed to enhance bone regeneration in vivo. However, the promising results of the less developed template in the critical-sized defect warrant further investigation, beyond interstitial flow, into the specific environment needed to optimize hypertrophic chondrocyte-based constructs for bone repair.
Collapse
Affiliation(s)
- Jonathan C Bernhard
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Elizabeth Hulphers
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Bernhard Rieder
- 2 Department of Biochemical Engineering, University of Applied Sciences Technikum Wien , Austrian Cluster for Tissue Regeneration Vienna, Vienna, Austria
| | - James Ferguson
- 3 Ludwig Boltzmann Institute of Experimental and Clinical Traumatology , University of Vienna, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Dominik Rünzler
- 2 Department of Biochemical Engineering, University of Applied Sciences Technikum Wien , Austrian Cluster for Tissue Regeneration Vienna, Vienna, Austria
| | - Thomas Nau
- 3 Ludwig Boltzmann Institute of Experimental and Clinical Traumatology , University of Vienna, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Heinz Redl
- 3 Ludwig Boltzmann Institute of Experimental and Clinical Traumatology , University of Vienna, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Gordana Vunjak-Novakovic
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
- 4 Department of Medicine, Columbia University , New York, New York
| |
Collapse
|
180
|
Ding M, Jin L, Xie L, Park SH, Tong Y, Wu D, Chhabra AB, Fu Z, Li X. A Murine Model for Human ECO Syndrome Reveals a Critical Role of Intestinal Cell Kinase in Skeletal Development. Calcif Tissue Int 2018; 102:348-357. [PMID: 29098359 PMCID: PMC5820141 DOI: 10.1007/s00223-017-0355-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023]
Abstract
An autosomal-recessive inactivating mutation R272Q in the human intestinal cell kinase (ICK) gene caused profound multiplex developmental defects in human endocrine-cerebro-osteodysplasia (ECO) syndrome. ECO patients exhibited a wide variety of skeletal abnormalities, yet the underlying mechanisms by which ICK regulates skeletal development remained largely unknown. The goal of this study was to understand the structural and mechanistic basis underlying skeletal anomalies caused by ICK dysfunction. Ick R272Q knock-in transgenic mouse model not only recapitulated major ECO skeletal defects such as short limbs and polydactyly but also revealed a deformed spine with defective intervertebral disk. Loss of ICK function markedly reduced mineralization in the spinal column, ribs, and long bones. Ick mutants showed a significant decrease in the proliferation zone of long bones and the number of type X collagen-expressing hypertrophic chondrocytes in the spinal column and the growth plate of long bones. These results implicate that ICK plays an important role in bone and cartilage development by promoting chondrocyte proliferation and maturation. Our findings provided new mechanistic insights into the skeletal phenotype of human ECO and ECO-like syndromes.
Collapse
Affiliation(s)
- Mengmeng Ding
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Jin
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
| | - Lin Xie
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
- Department of Orthopaedic Surgery, Wuhan Orthopaedic Hospital, Huazhong University of Science & Technology, Hubei, 430030, China
| | - So Hyun Park
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
| | - Yixin Tong
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
- The Gastrointestinal Surgery Center, Tongji Hospital, Huazhong University of Science & Technology, Hubei, 430030, China
| | - Di Wu
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA
| | - A Bobby Chhabra
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA
| | - Zheng Fu
- Department of Pharmacology, University of Virginia, PO Box 800735, 1340 Jefferson Park Avenue, Charlottesville, VA, 22908, USA.
| | - Xudong Li
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr., Charlottesville, VA, 22908, USA.
| |
Collapse
|
181
|
Yoo JI, Ha YC. Review of Epidemiology, Diagnosis, and Treatment of Osteosarcopenia in Korea. J Bone Metab 2018; 25:1-7. [PMID: 29564300 PMCID: PMC5854818 DOI: 10.11005/jbm.2018.25.1.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia was listed in the International Classification of Diseases, Tenth Revision, Clinical Modification (ICD-10-CM) as M62.84, on October 1, 2016. Sarcopenia is primarily associated with metabolic diseases, such as diabetes, obesity, and cachexia, as well as chronic renal failure, congestive heart failure, and chronic obstructive pulmonary disease. Sarcopenia is also significantly associated with osteoporosis in elderly populations and the combined disease is defined as osteosarcopenia. Several studies have confirmed that sarcopenia and osteoporosis (osteosarcopenia) share common risk factors and biological pathways. Osteosarcopenia is associated with significant physical disability, representing a significant threat to the loss of independence in later life. However, the pathophysiology and diagnosis of osteosarcopenia are not fully defined. Additionally, pharmacologic and hormonal treatments for sarcopenia are undergoing clinical trials. This review summarizes the epidemiology, pathophysiology, diagnosis, and treatment of osteosarcopenia, and includes Korean data.
Collapse
Affiliation(s)
- Jun-Il Yoo
- Department of Orthopaedic Surgery, Gyeongsang National University Hospital, Jinju, Korea
| | - Yong-Chan Ha
- Department of Orthopaedic Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
182
|
Chai M, Sanosaka T, Okuno H, Zhou Z, Koya I, Banno S, Andoh-Noda T, Tabata Y, Shimamura R, Hayashi T, Ebisawa M, Sasagawa Y, Nikaido I, Okano H, Kohyama J. Chromatin remodeler CHD7 regulates the stem cell identity of human neural progenitors. Genes Dev 2018; 32:165-180. [PMID: 29440260 PMCID: PMC5830929 DOI: 10.1101/gad.301887.117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022]
Abstract
Multiple congenital disorders often present complex phenotypes, but how the mutation of individual genetic factors can lead to multiple defects remains poorly understood. In the present study, we used human neuroepithelial (NE) cells and CHARGE patient-derived cells as an in vitro model system to identify the function of chromodomain helicase DNA-binding 7 (CHD7) in NE-neural crest bifurcation, thus revealing an etiological link between the central nervous system (CNS) and craniofacial anomalies observed in CHARGE syndrome. We found that CHD7 is required for epigenetic activation of superenhancers and CNS-specific enhancers, which support the maintenance of the NE and CNS lineage identities. Furthermore, we found that BRN2 and SOX21 are downstream effectors of CHD7, which shapes cellular identities by enhancing a CNS-specific cellular program and indirectly repressing non-CNS-specific cellular programs. Based on our results, CHD7, through its interactions with superenhancer elements, acts as a regulatory hub in the orchestration of the spatiotemporal dynamics of transcription factors to regulate NE and CNS lineage identities.
Collapse
Affiliation(s)
- MuhChyi Chai
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan.,Gene Regulation Research, Nara Institute of Science and Technology, Ikoma, Nara 630-0101, Japan
| | - Tsukasa Sanosaka
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hironobu Okuno
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Zhi Zhou
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ikuko Koya
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Satoe Banno
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tomoko Andoh-Noda
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yoshikuni Tabata
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan.,E-WAY Research Laboratory, Discovery, Medicine Creation, Neurology Business Group, Tsukuba, Ibaraki 300-2635, Japan
| | - Rieko Shimamura
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tetsutaro Hayashi
- Bioinformatics Research Unit, Advanced Center for Computing and Communication, RIKEN, Wako, Saitama 351-0198, Japan
| | - Masashi Ebisawa
- Bioinformatics Research Unit, Advanced Center for Computing and Communication, RIKEN, Wako, Saitama 351-0198, Japan
| | - Yohei Sasagawa
- Bioinformatics Research Unit, Advanced Center for Computing and Communication, RIKEN, Wako, Saitama 351-0198, Japan
| | - Itoshi Nikaido
- Bioinformatics Research Unit, Advanced Center for Computing and Communication, RIKEN, Wako, Saitama 351-0198, Japan.,Single-Cell Omics Research Unit, RIKEN Center for Developmental Biology, Wako, Saitama 351-0198, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Jun Kohyama
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
183
|
Hernandez-Muñoz I, Figuerola E, Sanchez-Molina S, Rodriguez E, Fernández-Mariño AI, Pardo-Pastor C, Bahamonde MI, Fernández-Fernández JM, García-Domínguez DJ, Hontecillas-Prieto L, Lavarino C, Carcaboso AM, de Torres C, Tirado OM, de Alava E, Mora J. RING1B contributes to Ewing sarcoma development by repressing the NaV1.6 sodium channel and the NF-κB pathway, independently of the fusion oncoprotein. Oncotarget 2018; 7:46283-46300. [PMID: 27317769 PMCID: PMC5216798 DOI: 10.18632/oncotarget.10092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/28/2016] [Indexed: 11/25/2022] Open
Abstract
Ewing sarcoma (ES) is an aggressive tumor defined by EWSR1 gene fusions that behave as an oncogene. Here we demonstrate that RING1B is highly expressed in primary ES tumors, and its expression is independent of the fusion oncogene. RING1B-depleted ES cells display an expression profile enriched in genes functionally involved in hematological development but RING1B depletion does not induce cellular differentiation. In ES cells, RING1B directly binds the SCN8A sodium channel promoter and its depletion results in enhanced Nav1.6 expression and function. The signaling pathway most significantly modulated by RING1B is NF-κB. RING1B depletion results in enhanced p105/p50 expression, which sensitizes ES cells to apoptosis by FGFR/SHP2/STAT3 blockade. Reduced NaV1.6 function protects ES cells from apoptotic cell death by maintaining low NF-κB levels. Our findings identify RING1B as a trait of the cell-of-origin and provide a potential targetable vulnerability.
Collapse
Affiliation(s)
| | - Elisabeth Figuerola
- Developmental Tumor Biology Laboratory, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950-Barcelona, Spain
| | - Sara Sanchez-Molina
- Developmental Tumor Biology Laboratory, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950-Barcelona, Spain
| | - Eva Rodriguez
- Developmental Tumor Biology Laboratory, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950-Barcelona, Spain
| | - Ana Isabel Fernández-Mariño
- Laboratori de Fisiologia Molecular, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08003-Barcelona, Spain.,Present Affiliation: Department of Neuroscience and Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin, Madison-53705, USA
| | - Carlos Pardo-Pastor
- Laboratori de Fisiologia Molecular, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08003-Barcelona, Spain
| | - María Isabel Bahamonde
- Laboratori de Fisiologia Molecular, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08003-Barcelona, Spain
| | - José M Fernández-Fernández
- Laboratori de Fisiologia Molecular, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08003-Barcelona, Spain
| | - Daniel J García-Domínguez
- Department of Pediatric Hematology and Oncology, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, 41013-Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Pediatric Hematology and Oncology, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, 41013-Seville, Spain
| | - Cinzia Lavarino
- Developmental Tumor Biology Laboratory, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950-Barcelona, Spain
| | - Angel M Carcaboso
- Developmental Tumor Biology Laboratory, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950-Barcelona, Spain
| | - Carmen de Torres
- Developmental Tumor Biology Laboratory, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950-Barcelona, Spain
| | - Oscar M Tirado
- Sarcoma Research Group, Laboratori d'Oncología Molecular, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908-Barcelona, Spain
| | - Enrique de Alava
- Department of Pediatric Hematology and Oncology, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, 41013-Seville, Spain
| | - Jaume Mora
- Developmental Tumor Biology Laboratory, Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950-Barcelona, Spain
| |
Collapse
|
184
|
Kraus T, Fischerauer S, Treichler S, Martinelli E, Eichler J, Myrissa A, Zötsch S, Uggowitzer PJ, Löffler JF, Weinberg AM. The influence of biodegradable magnesium implants on the growth plate. Acta Biomater 2018; 66:109-117. [PMID: 29174472 DOI: 10.1016/j.actbio.2017.11.031] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/18/2017] [Accepted: 11/20/2017] [Indexed: 11/17/2022]
Abstract
Mg-based biodegradable materials are considered promising candidates in the paediatric field due to their favourable mechanical and biological properties and their biodegrading potential that makes a second surgery for implant removal unnecessary. In many cases the surgical fixation technique requires a crossing of the growth plate by the implant in order to achieve an adequate fragment replacement or fracture stabilisation. This study investigates the kinetics of slowly and rapidly degrading Mg alloys in a transphyseal rat model, and also reports on their dynamics in the context of the physis and consecutive bone growth. Twenty-six male Sprague-Dawley rats received either a rapidly degrading (ZX50; n = 13) or a slowly degrading (WZ21; n = 13) Mg alloy, implanted transphyseal into the distal femur. The contralateral leg was drilled in the same manner and served as a direct sham specimen. Degradation behaviour, gas formation, and leg length were measured by continuous in vivo micro CT for up to 52 weeks, and additional high-resolution µCT (HRS) scans and histomorphological analyses of the growth plate were performed. The growth plate was locally destroyed and bone growth was significantly diminished by the fast degradation of ZX50 implants and the accompanying release of large amounts of hydrogen gas. In contrast, WZ21 implants showed homogenous and moderate degradation performance, and the effect on bone growth did not differ significantly from a single drill-hole defect. STATEMENT OF SIGNIFICANCE This study is the first that reports on the effects of degrading magnesium implants on the growth plate in a living animal model. The results show that high evolution of hydrogen gas due to rapid Mg degradation can damage the growth plate substantially. Slow degradation, however, such as seen for WZ21 alloys, does not affect the growth plate more than drilling alone, thus meeting one important prerequisite for deployment in paediatric osteosynthesis.
Collapse
Affiliation(s)
- Tanja Kraus
- Department of Paediatric Orthopaedics, Medical University Graz, 8036 Graz, Austria.
| | - Stefan Fischerauer
- Department of Trauma Surgery, Medical University Graz, 8036 Graz, Austria
| | - Stefan Treichler
- Department of Orthopaedics, Medical University Graz, 8036 Graz, Austria
| | | | - Johannes Eichler
- Department of Orthopaedics, Medical University Graz, 8036 Graz, Austria
| | - Anastasia Myrissa
- Department of Orthopaedics, Medical University Graz, 8036 Graz, Austria
| | - Silvia Zötsch
- Department of Paediatric and Adolescent Surgery, Medical University Graz, 8036 Graz, Austria
| | - Peter J Uggowitzer
- Laboratory of Metal Physics and Technology, Department of Materials, ETH Zurich, 8093 Zurich, Switzerland
| | - Jörg F Löffler
- Laboratory of Metal Physics and Technology, Department of Materials, ETH Zurich, 8093 Zurich, Switzerland
| | | |
Collapse
|
185
|
Corciulo C, Irrera N, Cronstein BN. Adenosine Receptors Regulate Bone Remodeling and Cartilage Physiology. THE ADENOSINE RECEPTORS 2018:515-527. [DOI: 10.1007/978-3-319-90808-3_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
186
|
Protein and Peptides for Elderly Health. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 112:265-308. [DOI: 10.1016/bs.apcsb.2018.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
187
|
Bonyadi Rad E, Musumeci G, Pichler K, Heidary M, Szychlinska MA, Castrogiovanni P, Marth E, Böhm C, Srinivasaiah S, Krönke G, Weinberg A, Schäfer U. Runx2 mediated Induction of Novel Targets ST2 and Runx3 Leads to Cooperative Regulation of Hypertrophic Differentiation in ATDC5 Chondrocytes. Sci Rep 2017; 7:17947. [PMID: 29263341 PMCID: PMC5738421 DOI: 10.1038/s41598-017-18044-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/04/2017] [Indexed: 11/17/2022] Open
Abstract
Knowledge concerning expression and function of Suppression of Tumorigenicity 2 (ST2) in chondrocytes is at present, limited. Analysis of murine growth plates and ATDC5 chondrocytes indicated peak expression of the ST2 transmembrane receptor (ST2L) and soluble (sST2) isoforms during the hypertrophic differentiation concomitant with the expression of the hypertrophic markers Collagen X (Col X), Runx2 and MMP-13. Gain- and loss-of-function experiments in ATDC5 and primary human growth plate chondrocytes (PHCs), confirmed regulation of ST2 by the key transcription factor Runx2, indicating ST2 to be a novel Runx2 target. ST2 knock-out mice (ST2−/−) exhibited noticeable hypertrophic zone (HZ) reduction in murine growth plates, accompanied by lower expression of Col X and Osteocalcin (OSC) compared to wild-type (WT) mice. Likewise, ST2 knockdown resulted in decreased Col X expression and downregulation of OSC and Vascular Endothelial Growth Factor (VEGF) in ATDC5 cells. The ST2 suppression was also associated with upregulation of the proliferative stage markers Sox9 and Collagen II (Col II), indicating ST2 to be a new regulator of ATDC5 chondrocyte differentiation. Runx3 was, furthermore, identified as a novel Runx2 target in chondrocytes. This study suggests that Runx2 mediates ST2 and Runx3 induction to cooperatively regulate hypertrophic differentiation of ATDC5 chondrocytes.
Collapse
Affiliation(s)
- Ehsan Bonyadi Rad
- Department of Orthopedics and Trauma Surgery, Medical University Graz, Graz, Austria.
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Karin Pichler
- Department of Children and Adolescent Medicine, Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria.,Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Maryam Heidary
- Translational Research Department, Institute Curie, Paris, France
| | - Marta Anna Szychlinska
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Egon Marth
- Institute of Hygiene, Microbiology and Environmental Medicine, Medical University Graz, Graz, Austria
| | - Christina Böhm
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sriveena Srinivasaiah
- Department of Orthopedics and Trauma Surgery, Medical University Graz, Graz, Austria
| | - Gerhard Krönke
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Annelie Weinberg
- Department of Orthopedics and Trauma Surgery, Medical University Graz, Graz, Austria
| | - Ute Schäfer
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| |
Collapse
|
188
|
Hu X, Li L, Yu X, Zhang R, Yan S, Zeng Z, Shu Y, Zhao C, Wu X, Lei J, Li Y, Zhang W, Yang C, Wu K, Wu Y, An L, Huang S, Ji X, Gong C, Yuan C, Zhang L, Liu W, Huang B, Feng Y, Zhang B, Haydon RC, Luu HH, Reid RR, Lee MJ, Wolf JM, Yu Z, He TC. CRISPR/Cas9-mediated reversibly immortalized mouse bone marrow stromal stem cells (BMSCs) retain multipotent features of mesenchymal stem cells (MSCs). Oncotarget 2017; 8:111847-111865. [PMID: 29340096 PMCID: PMC5762364 DOI: 10.18632/oncotarget.22915] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells that can undergo self-renewal and differentiate into multi-lineages. Bone marrow stromal stem cells (BMSCs) represent one of the most commonly-used MSCs. In order to overcome the technical challenge of maintaining primary BMSCs in long-term culture, here we seek to establish reversibly immortalized mouse BMSCs (imBMSCs). By exploiting CRISPR/Cas9-based homology-directed-repair (HDR) mechanism, we target SV40T to mouse Rosa26 locus and efficiently immortalize mouse BMSCs (i.e., imBMSCs). We also immortalize BMSCs with retroviral vector SSR #41 and establish imBMSC41 as a control line. Both imBMSCs and imBMSC41 exhibit long-term proliferative capability although imBMSC41 cells have a higher proliferation rate. SV40T mRNA expression is 130% higher in imBMSC41 than that in imBMSCs. However, FLP expression leads to 86% reduction of SV40T expression in imBMSCs, compared with 63% in imBMSC41 cells. Quantitative genomic PCR analysis indicates that the average copy number of SV40T and hygromycin is 1.05 for imBMSCs and 2.07 for imBMSC41, respectively. Moreover, FLP expression removes 92% of SV40T in imBMSCs at the genome DNA level, compared with 58% of that in imBMSC41 cells, indicating CRISPR/Cas9 HDR-mediated immortalization of BMSCs can be more effectively reversed than that of retrovirus-mediated random integrations. Nonetheless, both imBMSCs and imBMSC41 lines express MSC markers and are highly responsive to BMP9-induced osteogenic, chondrogenic and adipogenic differentiation in vitro and in vivo. Thus, the engineered imBMSCs can be used as a promising alternative source of primary MSCs for basic and translational research in the fields of MSC biology and regenerative medicine.
Collapse
Affiliation(s)
- Xue Hu
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Li Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Biomedical Engineering, School of Biomedical Engineering, Chongqing University, Chongqing 400044, China
| | - Xinyi Yu
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ruyi Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shujuan Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
- The Children’s Hospital, Chongqing Medical University, Chongqing 400014, China
| | - Chen Zhao
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xingye Wu
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jiayan Lei
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yasha Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- The Children’s Hospital, Chongqing Medical University, Chongqing 400014, China
| | - Wenwen Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Laboratory Medicine and Clinical Diagnostics, The Affiliated Yantai Hospital, Binzhou Medical University, Yantai 264100, China
| | - Chao Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- The Children’s Hospital, Chongqing Medical University, Chongqing 400014, China
| | - Ke Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ying Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Immunology and Microbiology, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Liping An
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Orthopaedic Surgery of Gansu Province and The Department of Orthopaedic Surgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Shifeng Huang
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaojuan Ji
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- The Children’s Hospital, Chongqing Medical University, Chongqing 400014, China
| | - Cheng Gong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Surgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Chengfu Yuan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Biochemistry and Molecular Biology, China Three Gorges University School of Medicine, Yichang 443002, China
| | - Linghuan Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- The Children’s Hospital, Chongqing Medical University, Chongqing 400014, China
| | - Wei Liu
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bo Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yixiao Feng
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bo Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Key Laboratory of Orthopaedic Surgery of Gansu Province and The Department of Orthopaedic Surgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zebo Yu
- Departments of Blood Transfusion, Nephrology, Orthopaedic Surgery, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
189
|
Geng X, Liu S, Yuan Z, Jiang Y, Zhi D, Liu Z. A Genome-Wide Association Study Reveals That Genes with Functions for Bone Development Are Associated with Body Conformation in Catfish. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2017; 19:570-578. [PMID: 28971324 DOI: 10.1007/s10126-017-9775-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 08/08/2017] [Indexed: 06/07/2023]
Abstract
Body conformation is of great scientific and commercial interest for aquaculture fish species because it affects biological adaptation of the organism to environments, and is of economic importance to the aquaculture industry considering its direct effect on fillet yield. Catfish is the primary aquaculture species in the USA. Two major species used in the aquaculture industry, channel catfish and blue catfish, differ in body shape and therefore the backcross progenies serve as a good model for quantitative trait locus (QTL) analysis. Here, a genome-wide association study (GWAS) with hybrid catfish was conducted to identify the QTL for body conformation, including deheaded body length (DBL), body length (BL), body depth (BD), and body breadth (BB), which were all standardized by cubic root of body weight. Overall, the results indicate that the traits are polygenic. For DBL, linkage group (LG) 2 and LG 24 contain significant QTL, and LG 13 and LG 26 contain suggestively associated QTL (-log10(P value) > 4.5). Compared with DBL, additional SNPs were identified to be associated with body length on LG 2, LG 7, and LG 18. Although no significant QTL for body depth was found, three suggestively associated QTLs were identified on LG 5, LG 13, and LG 14. No SNP for body breadth reached the threshold for suggestive association. Genes close to the associated SNPs were determined, many of which are known to be involved in bone development. This work therefore provides the basis for future identification of causal genes for the control of body conformation.
Collapse
Affiliation(s)
- Xin Geng
- Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Shikai Liu
- Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Zihao Yuan
- Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Yanliang Jiang
- Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Degui Zhi
- School of Public Health and School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhanjiang Liu
- Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
190
|
Live Fluorescent Staining Platform for Drug-Screening and Mechanism-Analysis in Zebrafish for Bone Mineralization. Molecules 2017; 22:molecules22122068. [PMID: 29186901 PMCID: PMC6149919 DOI: 10.3390/molecules22122068] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/10/2017] [Accepted: 11/22/2017] [Indexed: 11/21/2022] Open
Abstract
Currently, drug screening relies on cell-based experiments or on animal models to confirm biological effects. The mammalian system is considered too time-consuming, expensive and complex to perform high-throughput drug screening. There is a gap between in vitro cell-based models and the in vivo mammalian models. The zebrafish is an ideal model that could link preclinical toxicity screening with the drug development pipeline. Taking advantage of a highly conservative genomic, rapid development, large number of offspring, low cost and easy manipulation, zebrafish has been considered an excellent animal model for disease-based drug screening. In this study, zebrafish embryos were incubated with small molecular compounds that potentially affected bone mineralization in microplates. Two compounds of alendronate and dorsomorphin were used as positive and negative controls, respectively. The level of osteogenic mineralization was measured and quantified by using ImageJ software with fluorescent calcein-staining images. Among twenty-four tested compounds from the kinase inhibitor library, we identified two compounds, pentamidine and BML-267, which showed increased embryonic mineralization; while six compounds, RWJ-60475, levamisole HCL, tetramisole HCL, fenvalerate, NSC-663284, and BML-267ester, were inhibitory to bone mineralization. In addition, real time quantitative PCR (RT-qPCR) was performed to evaluate the biological pathways involved in bone metabolism at the molecular level. We confirmed that alendronate enhanced the level of bone mineralization by inhibiting osteoclast-related genes. In summary, our research established a simple method to screen potential bone metabolic drugs and to perform mechanism analysis for bone mineralization in vivo.
Collapse
|
191
|
Doro DH, Grigoriadis AE, Liu KJ. Calvarial Suture-Derived Stem Cells and Their Contribution to Cranial Bone Repair. Front Physiol 2017; 8:956. [PMID: 29230181 PMCID: PMC5712071 DOI: 10.3389/fphys.2017.00956] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/10/2017] [Indexed: 12/13/2022] Open
Abstract
In addition to the natural turnover during life, the bones in the skeleton possess the ability to self-repair in response to injury or disease-related bone loss. Based on studies of bone defect models, both processes are largely supported by resident stem cells. In the long bones, the source of skeletal stem cells has been widely investigated over the years, where the major stem cell population is thought to reside in the perivascular niche of the bone marrow. In contrast, we have very limited knowledge about the stem cells contributing to the repair of calvarial bones. In fact, until recently, the presence of specific stem cells in adult craniofacial bones was uncertain. These flat bones are mainly formed via intramembranous rather than endochondral ossification and thus contain minimal bone marrow space. It has been previously proposed that the overlying periosteum and underlying dura mater provide osteoprogenitors for calvarial bone repair. Nonetheless, recent studies have identified a major stem cell population within the suture mesenchyme with multiple differentiation abilities and intrinsic reparative potential. Here we provide an updated review of calvarial stem cells and potential mechanisms of regulation in the context of skull injury repair.
Collapse
Affiliation(s)
- Daniel H Doro
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - Agamemnon E Grigoriadis
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
192
|
DLX3 promotes bone marrow mesenchymal stem cell proliferation through H19/miR-675 axis. Clin Sci (Lond) 2017; 131:2721-2735. [PMID: 28963438 DOI: 10.1042/cs20171231] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/03/2017] [Accepted: 09/27/2017] [Indexed: 11/17/2022]
Abstract
The underlying molecular mechanism of the increased bone mass phenotype in Tricho-dento-osseous (TDO) syndrome remains largely unknown. Our previous study has shown that the TDO point mutation c.533A>G, Q178R in DLX3 could increase bone density in a TDO patient and transgenic mice partially through delaying senescence in bone marrow mesenchymal stem cells (BMSCs). In the present study, we provided a new complementary explanation for TDO syndrome: the DLX3 (Q178R) mutation increased BMSCs proliferation through H19/miR-675 axis. We found that BMSCs derived from the TDO patient (TDO-BMSCs) had stronger proliferation ability than controls by clonogenic and CCK-8 assays. Next, experiments of overexpression and knockdown of wild-type DLX3 via lentiviruses in normal BMSCs confirmed the results by showing its negative role in cell proliferation. Through validated high-throughput data, we found that the DLX3 mutation reduced the expression of H19 and its coexpression product miR-675 in BMSCs. Function and rescue assays suggested that DLX3, long noncoding RNA H19, and miR-675 are negative factors in modulation of BMSCs proliferation as well as NOMO1 expression. The original higher proliferation rate and the expression of NOMO1 in TDO-BMSCs were suppressed after H19 restoration. Collectively, it indicates that DLX3 regulates BMSCs proliferation through H19/miR-675 axis. Moreover, the increased expression of NOMO1 and decreased H19/miR-675 expression in DLX3 (Q178R) transgenic mice, accompanying with accrual bone mass and density detected by micro-CT, further confirmed our hypothesis. In summary, we, for the first time, demonstrate that DLX3 mutation interferes with bone formation partially through H19/miR-675/NOMO1 axis in TDO syndrome.
Collapse
|
193
|
Oura P, Paananen M, Niinimäki J, Tammelin T, Auvinen J, Korpelainen R, Karppinen J, Junno JA. High-impact exercise in adulthood and vertebral dimensions in midlife - the Northern Finland Birth Cohort 1966 study. BMC Musculoskelet Disord 2017; 18:433. [PMID: 29110646 PMCID: PMC5674855 DOI: 10.1186/s12891-017-1794-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 10/31/2017] [Indexed: 11/10/2022] Open
Abstract
Background Vertebral size and especially cross-sectional area (CSA) are independently associated with vertebral fracture risk. Previous studies have suggested that physical activity and especially high-impact exercise may affect vertebral strength. We aimed to investigate the association between high-impact exercise at 31 and 46 years of age and vertebral dimensions in midlife. Methods We used a subsample of 1023 individuals from the Northern Finland Birth Cohort 1966 study with records of self-reported sports participation from 31 and 46 years and MRI-derived data on vertebral dimensions from 46 years. Based on the sports participation data, we constructed three impact categories (high, mixed, low) that represented longitudinal high-impact exercise activity in adulthood. We used linear regression and generalized estimating equation (GEE) models to analyse the association between high-impact exercise and vertebral CSA, with adjustments for vertebral height and body mass index. Results Participation in high-impact sports was associated with large vertebral CSA among women but not men. The women in the 'mixed' group had 36.8 (95% confidence interval 11.2–62.5) mm2 larger CSA and the women in the 'high' group 43.2 (15.2–71.1) mm2 larger CSA than the 'low' group. Conclusions We suggest that participation (≥ 1/week) in one or more high-impact sports in adulthood is associated with larger vertebral size, and thus increased vertebral strength, among middle-aged women.
Collapse
Affiliation(s)
- Petteri Oura
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5000, 90014, Oulu, Finland. .,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland. .,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.
| | - Markus Paananen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - Jaakko Niinimäki
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - Tuija Tammelin
- LIKES-Research Center for Sport and Health Sciences, Rautpohjankatu 8, 40700, Jyväskylä, Finland
| | - Juha Auvinen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - Raija Korpelainen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Department of Sports and Exercise Medicine, Oulu Deaconess Institute, Albertinkatu 18A, 90100, Oulu, Finland
| | - Jaro Karppinen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Finnish Institute of Occupational Health, Kastelli Research Center, Aapistie 1, 90220, Oulu, Finland
| | - Juho-Antti Junno
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Department of Archaeology, Faculty of Humanities, University of Oulu, P.O. Box 8000, 90014, Oulu, Finland
| |
Collapse
|
194
|
Marcucio RS, Qin L, Alsberg E, Boerckel JD. Reverse engineering development: Crosstalk opportunities between developmental biology and tissue engineering. J Orthop Res 2017; 35:2356-2368. [PMID: 28660712 DOI: 10.1002/jor.23636] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/12/2017] [Indexed: 02/04/2023]
Abstract
The fields of developmental biology and tissue engineering have been revolutionized in recent years by technological advancements, expanded understanding, and biomaterials design, leading to the emerging paradigm of "developmental" or "biomimetic" tissue engineering. While developmental biology and tissue engineering have long overlapping histories, the fields have largely diverged in recent years at the same time that crosstalk opportunities for mutual benefit are more salient than ever. In this perspective article, we will use musculoskeletal development and tissue engineering as a platform on which to discuss these emerging crosstalk opportunities and will present our opinions on the bright future of these overlapping spheres of influence. The multicellular programs that control musculoskeletal development are rapidly becoming clarified, represented by shifting paradigms in our understanding of cellular function, identity, and lineage specification during development. Simultaneously, advancements in bioartificial matrices that replicate the biochemical, microstructural, and mechanical properties of developing tissues present new tools and approaches for recapitulating development in tissue engineering. Here, we introduce concepts and experimental approaches in musculoskeletal developmental biology and biomaterials design and discuss applications in tissue engineering as well as opportunities for tissue engineering approaches to inform our understanding of fundamental biology. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2356-2368, 2017.
Collapse
Affiliation(s)
- Ralph S Marcucio
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania
| | - Eben Alsberg
- Departments of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, Cleveland, Ohio
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania.,Department of Bioengineering, University of Pennslyvania, Philadelphia, Pennsylvania.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
195
|
Kim KM, Jang WG. Zaluzanin C (ZC) induces osteoblast differentiation through regulating of osteogenic genes expressions in early stage of differentiation. Bioorg Med Chem Lett 2017; 27:4789-4793. [DOI: 10.1016/j.bmcl.2017.09.061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 11/24/2022]
|
196
|
Walmsley GG, Ransom RC, Zielins ER, Leavitt T, Flacco JS, Hu MS, Lee AS, Longaker MT, Wan DC. Stem Cells in Bone Regeneration. Stem Cell Rev Rep 2017; 12:524-529. [PMID: 27250635 DOI: 10.1007/s12015-016-9665-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Bone has the capacity to regenerate and repair itself. However, this capacity may be impaired or lost depending on the size of the defect or the presence of certain disease states. In this review, we discuss the key principles underlying bone healing, efforts to characterize bone stem and progenitor cell populations, and the current status of translational and clinical studies in cell-based bone tissue engineering. Though barriers to clinical implementation still exist, the application of stem and progenitor cell populations to bone engineering strategies has the potential to profoundly impact regenerative medicine.
Collapse
Affiliation(s)
- Graham G Walmsley
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Hagey Building, 257 Campus Dr., Stanford, CA, 94305, USA
| | - Ryan C Ransom
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Hagey Building, 257 Campus Dr., Stanford, CA, 94305, USA
| | - Elizabeth R Zielins
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA
| | - Tripp Leavitt
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA
| | - John S Flacco
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA
| | - Michael S Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Hagey Building, 257 Campus Dr., Stanford, CA, 94305, USA.,Department of Surgery, John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawai'i, USA
| | - Andrew S Lee
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Hagey Building, 257 Campus Dr., Stanford, CA, 94305, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Hagey Building, 257 Campus Dr., Stanford, CA, 94305, USA
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive Room GK106, Stanford, CA, 94305-5461, USA.
| |
Collapse
|
197
|
Wang L, Huang J, Moore DC, Zuo C, Wu Q, Xie L, von der Mark K, Yuan X, Chen D, Warman ML, Ehrlich MG, Yang W. SHP2 Regulates the Osteogenic Fate of Growth Plate Hypertrophic Chondrocytes. Sci Rep 2017; 7:12699. [PMID: 28983104 PMCID: PMC5629218 DOI: 10.1038/s41598-017-12767-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 09/14/2017] [Indexed: 02/07/2023] Open
Abstract
Transdifferentiation of hypertrophic chondrocytes into bone-forming osteoblasts has been reported, yet the underlying molecular mechanism remains incompletely understood. SHP2 is an ubiquitously expressed cytoplasmic protein tyrosine phosphatase. SHP2 loss-of-function mutations in chondroid cells are linked to metachondromatosis in humans and mice, suggesting a crucial role for SHP2 in the skeleton. However, the specific role of SHP2 in skeletal cells has not been elucidated. To approach this question, we ablated SHP2 in collagen 2α1(Col2α1)-Cre- and collagen 10α1(Col10α1)-Cre-expressing cells, predominantly proliferating and hypertrophic chondrocytes, using "Cre-loxP"-mediated gene excision. Mice lacking SHP2 in Col2α1-Cre-expressing cells die at mid-gestation. Postnatal SHP2 ablation in the same cell population caused dwarfism, chondrodysplasia and exostoses. In contrast, mice in which SHP2 was ablated in the Col10α1-Cre-expressing cells appeared normal but were osteopenic. Further mechanistic studies revealed that SHP2 exerted its influence partly by regulating the abundance of SOX9 in chondrocytes. Elevated and sustained SOX9 in SHP2-deficient hypertrophic chondrocytes impaired their differentiation to osteoblasts and impaired endochondral ossification. Our study uncovered an important role of SHP2 in bone development and cartilage homeostasis by influencing the osteogenic differentiation of hypertrophic chondrocytes and provided insight into the pathogenesis and potential treatment of skeletal diseases, such as osteopenia and osteoporosis.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Jiahui Huang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Douglas C Moore
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Chunlin Zuo
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P.R. China
| | - Qian Wu
- Department of Pathology and Laboratory Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Liqin Xie
- Regeneron Pharmaceuticals, Tarrytown, NY, 10591, USA
| | - Klaus von der Mark
- Department of Experimental Medicine, University of Erlangen-Nürnberg, Gluckstrasse 6, 91054, Erlangen, Germany
| | - Xin Yuan
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Di Chen
- Department of Biochemistry, Rush University, 600 S. Paulina St., Chicago, IL, 60612, USA
| | - Matthew L Warman
- Orthopaedic Research Laboratories and Howard Hughes Medical Institute, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Michael G Ehrlich
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Wentian Yang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA.
| |
Collapse
|
198
|
Choi YJ, Jeong S, Yoon KA, Sung HJ, Cho HS, Kim DW, Cho JY. Deficiency of DGCR8 increases bone formation through downregulation of miR-22 expression. Bone 2017; 103:287-294. [PMID: 28739418 DOI: 10.1016/j.bone.2017.07.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/15/2017] [Accepted: 07/19/2017] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNA) significantly contribute to bone formation by post-transcriptional regulation of gene expression. Mature miRNAs are generated following sequential cleavage by DROSHA/DGCR8 and DICER. However, recent studies have identified that some miRNAs require only one of these enzymes. Most studies seeking to clarify the role of miRNA during bone formation have been performed using DICER deletion strategies, but little is known regarding the role of DGCR8. To study the function of DGCR8 in osteogenesis, we generated mice in which Dgcr8 is conditionally deleted in osteoprogenitor cells by Col1a1-Cre. Dgcr8-cKO mice showed increased bone volume (BV/TV), trabecular number (Tb/N), and trabecular thickness (Tb.Th), but decreased trabecular separation (Tb.Sp) in the femur. Von Kossa, tartrate-resistant acid phosphatase staining, and calcein double labeling identified that osteoblast activity is increased in Dgcr8-cKO mice. In an effort to elucidate a detailed cellular mechanism, we found that miR-22 was downregulated in Dgcr8-cKO mice, leading to upregulation of the osteocalcin transcript, a key marker of osteoblasts. Interestingly, the mRNA expression level of Dgcr8 was decreased during osteoblast differentiation. Taken together, these results strongly indicate that DGCR8-dependent generation of miR-22 is essential for bone formation and that miR-22 could be a therapeutic target for individuals with bone disease.
Collapse
Affiliation(s)
- Young-Jin Choi
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Sujin Jeong
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kyung-Ae Yoon
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hye-Jin Sung
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hye-Sim Cho
- ProtAnBio, Seoul National University, Seoul, South Korea
| | - Dong Wook Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
199
|
|
200
|
Liao J, Yu X, Hu X, Fan J, Wang J, Zhang Z, Zhao C, Zeng Z, Shu Y, Zhang R, Yan S, Li Y, Zhang W, Cui J, Ma C, Li L, Yu Y, Wu T, Wu X, Lei J, Wang J, Yang C, Wu K, Wu Y, Tang J, He BC, Deng ZL, Luu HH, Haydon RC, Reid RR, Lee MJ, Wolf JM, Huang W, He TC. lncRNA H19 mediates BMP9-induced osteogenic differentiation of mesenchymal stem cells (MSCs) through Notch signaling. Oncotarget 2017; 8:53581-53601. [PMID: 28881833 PMCID: PMC5581132 DOI: 10.18632/oncotarget.18655] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/23/2017] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitor cells that can undergo self-renewal and differentiate into multiple lineages. Osteogenic differentiation from MSCs is a well-orchestrated process and regulated by multiple signaling pathways. We previously demonstrated that BMP9 is one of the most potent osteogenic factors. However, molecular mechanism through which BMP9 governs osteoblastic differentiation remains to be fully understood. Increasing evidence indicates noncoding RNAs (ncRNAs) may play important regulatory roles in many physiological and/or pathologic processes. In this study, we investigate the role of lncRNA H19 in BMP9-regulated osteogenic differentiation of MSCs. We demonstrated that H19 was sharply upregulated at the early stage of BMP9 stimulation of MSCs, followed by a rapid decease and gradual return to basal level. This process was correlated with BMP9-induced expression of osteogenic markers. Interestingly, either constitutive H19 expression or silencing H19 expression in MSCs significantly impaired BMP9-induced osteogenic differentiation in vitro and in vivo, which was effectively rescued by the activation of Notch signaling. Either constitutive H19 expression or silencing H19 expression led to the increased expression of a group of miRNAs that are predicted to target Notch ligands and receptors. Thus, these results indicate that lncRNA H19 functions as an important mediator of BMP9 signaling by modulating Notch signaling-targeting miRNAs. Our findings suggest that the well-coordinated biphasic expression of lncRNA H19 may be essential in BMP9-induced osteogenic differentiation of MSCs, and that dysregulated H19 expression may impair normal osteogenesis, leading to pathogenic processes, such as bone tumor development.
Collapse
Affiliation(s)
- Junyi Liao
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Xinyi Yu
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Xue Hu
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Zhicai Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Chen Zhao
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Ruyi Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Shujuan Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yasha Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Wenwen Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Department of Laboratory Medicine and Clinical Diagnostics, The Affiliated Yantai Hospital, Binzhou Medical University, Yantai, China
| | - Jing Cui
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Chao Ma
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Departments of Neurosurgery, and Otolaryngology-Head & Neck Surgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Li Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Department of Biomedical Engineering, School of Bioengineering, Chongqing University, Chongqing, China
| | - Yichun Yu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Department of Emergency Medicine, Beijing Hospital, Beijing, China
| | - Tingting Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Departments of Neurosurgery, and Otolaryngology-Head & Neck Surgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xingye Wu
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Jiayan Lei
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Jia Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Chao Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Ke Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Ying Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Department of Immunology and Microbiology, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Tang
- Cytate Institute for Precision Medicine & Innovation, Guangzhou Cytate Biomedical Technologies Inc., Guangzhou, China
| | - Bai-Cheng He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Zhong-Liang Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Wei Huang
- Departments of Orthopaedic Surgery, Blood Transfusion, Nephrology, and General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and The Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| |
Collapse
|