1
|
Jaabar IL, Foley B, Mezzetti A, Pillier F, Berenbaum F, Landoulsi J, Houard X. Unraveling the Mechanisms of Hypertrophy-Induced Matrix Mineralization and Modifications in Articular Chondrocytes. Calcif Tissue Int 2024; 115:269-282. [PMID: 38918254 DOI: 10.1007/s00223-024-01229-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/12/2024] [Indexed: 06/27/2024]
Abstract
Chondrocyte hypertrophic differentiation is a main event leading to articular cartilage degradation in osteoarthritis. It is associated with matrix remodeling and mineralization, the dynamics of which is not well characterized during chondrocyte hypertrophic differentiation in articular cartilage. Based on an in vitro model of progressive differentiation of immature murine articular chondrocytes (iMACs) into prehypertrophic (Prehyp) and hypertrophic (Hyp) chondrocytes, we performed kinetics of chondrocyte differentiation from Prehyp to Hyp to follow matrix mineralization and remodeling by immunofluorescence, biochemical, molecular, and physicochemical approaches, including atomic force microscopy, scanning electron microscopy associated with energy-dispersive X-ray spectroscopy (SEM-EDS), attenuated total reflection infrared analyses, and X-ray diffraction. Chondrocyte apoptosis was determined by TUNEL assay. The results show the formation of a mineral phase 7 days after Hyp induction, which spreads within the matrices to form poorly crystalline carbonate-substituted hydroxyapatite after 14 days, then the proportions of crystalline relative to amorphous content increases over time. Hyp differentiation also induced a matrix turnover that occurs over the first 7 days, characterized by a decrease in type II collagen and aggrecan and the concomitant appearance of type X collagen. This is accompanied by an increase in the enzymatic activity of MMP-13, the main collagenase in cartilage. The number of apoptotic chondrocytes slightly increased with Hyp differentiation and SEM-EDS analyses detected phosphorus-rich structures that could correspond to apoptotic bodies. Our findings highlight the mechanisms of matrix remodeling events leading to the mineralization of articular cartilage that may occur in osteoarthritis.
Collapse
Affiliation(s)
- Ilhem Lilia Jaabar
- Laboratoire de Réactivité de Surface, LRS, CNRS, Sorbonne Université, 4, Place Jussieu, 75005, Paris, France
- INSERM, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, 34 Rue Crozatier, 75012, Paris, France
| | - Brittany Foley
- Laboratoire de Réactivité de Surface, LRS, CNRS, Sorbonne Université, 4, Place Jussieu, 75005, Paris, France
- Laboratoire de Biomécanique & Bioingénierie, CNRS, Université de Technologie de Compiègne, BP 20529, 60205, Compiègne Cedex, France
| | - Alberto Mezzetti
- Laboratoire de Réactivité de Surface, LRS, CNRS, Sorbonne Université, 4, Place Jussieu, 75005, Paris, France
| | - Françoise Pillier
- Laboratoire Interfaces et Systèmes Electrochimiques, LISE, CNRS,, Sorbonne Université, 75012, Paris, France
| | - Francis Berenbaum
- INSERM, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, 34 Rue Crozatier, 75012, Paris, France
- Rheumatology Department, AP-HP Saint-Antoine Hospital, 184, Rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Jessem Landoulsi
- Laboratoire de Réactivité de Surface, LRS, CNRS, Sorbonne Université, 4, Place Jussieu, 75005, Paris, France.
| | - Xavier Houard
- INSERM, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, 34 Rue Crozatier, 75012, Paris, France.
| |
Collapse
|
2
|
Zhang Q, Li Q, Wang Y, Zhang Y, Peng R, Wang Z, Zhu B, Xu L, Gao X, Chen Y, Gao H, Hu J, Qian C, Ma M, Duan R, Li J, Zhang L. Characterization of Chromatin Accessibility in Fetal Bovine Chondrocytes. Animals (Basel) 2023; 13:1875. [PMID: 37889831 PMCID: PMC10251841 DOI: 10.3390/ani13111875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 10/29/2023] Open
Abstract
Despite significant advances of the bovine epigenome investigation, new evidence for the epigenetic basis of fetal cartilage development remains lacking. In this study, the chondrocytes were isolated from long bone tissues of bovine fetuses at 90 days. The Assay for Transposase-Accessible Chromatin with high throughput sequencing (ATAC-seq) and transcriptome sequencing (RNA-seq) were used to characterize gene expression and chromatin accessibility profile in bovine chondrocytes. A total of 9686 open chromatin regions in bovine fetal chondrocytes were identified and 45% of the peaks were enriched in the promoter regions. Then, all peaks were annotated to the nearest gene for Gene Ontology (GO) and Kyoto Encylopaedia of Genes and Genomes (KEGG) analysis. Growth and development-related processes such as amide biosynthesis process (GO: 0043604) and translation regulation (GO: 006417) were enriched in the GO analysis. The KEGG analysis enriched endoplasmic reticulum protein processing signal pathway, TGF-β signaling pathway and cell cycle pathway, which are closely related to protein synthesis and processing during cell proliferation. Active transcription factors (TFs) were enriched by ATAC-seq, and were fully verified with gene expression levels obtained by RNA-seq. Among the top50 TFs from footprint analysis, known or potential cartilage development-related transcription factors FOS, FOSL2 and NFY were found. Overall, our data provide a theoretical basis for further determining the regulatory mechanism of cartilage development in bovine.
Collapse
Affiliation(s)
- Qi Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Qian Li
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Yahui Wang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Yapeng Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Ruiqi Peng
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Zezhao Wang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Bo Zhu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Lingyang Xu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Xue Gao
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Yan Chen
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Huijiang Gao
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Junwei Hu
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Cong Qian
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Minghao Ma
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Rui Duan
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Junya Li
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Lupei Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| |
Collapse
|
3
|
Ge J, Cheng X, Yan Q, Wu C, Wang Y, Yu H, Yang H, Zhou F, Zou J. Calcitonin inhibits intervertebral disc degeneration by regulating protein kinase C. J Cell Mol Med 2020; 24:8650-8661. [PMID: 32564456 PMCID: PMC7412402 DOI: 10.1111/jcmm.15496] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 04/29/2020] [Accepted: 05/24/2020] [Indexed: 01/18/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) is the most critical factor that causes low back pain. Molecular biotherapy is a fundamental strategy for IVDD treatment. Calcitonin can promote the proliferation of chondrocytes, stimulate the synthesis of matrix and prevent cartilage degeneration. However, its effect and the underlying mechanism for IVDD have not been fully revealed. Chondrogenic specific matrix components’ mRNA expression of nucleus pulposus cell (NPC) was determined by qPCR. Protein expression of NPC matrix components and protein kinase C was determined by Western blotting. A rat caudal intervertebral disc degeneration model was established and tested for calcitonin in vivo. IL‐1 induced NPC change via decreasing protein kinase C (PKC)‐ε phosphorylation, while increasing PKC‐δ phosphorylation. Calcitonin treatment could prevent or reverse IL‐1‐induced cellular change on PKC signalling associated with degeneration. The positive effect of calcitonin on IVDD in vivo was verified on a rat caudal model. In summary, this study, for the first time, elucidated the important role of calcitonin in the regulation of matrix components in the nucleus of the intervertebral disc. Calcitonin can delay degeneration of the intervertebral disc nucleus by activating the PKC‐ε pathway and inhibiting the PKC‐δ pathway.
Collapse
Affiliation(s)
- Jun Ge
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoqiang Cheng
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Yan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cenhao Wu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yingjie Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Yu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Gonzalez-Fernandez T, Rathan S, Hobbs C, Pitacco P, Freeman FE, Cunniffe GM, Dunne NJ, McCarthy HO, Nicolosi V, O'Brien FJ, Kelly DJ. Pore-forming bioinks to enable spatio-temporally defined gene delivery in bioprinted tissues. J Control Release 2019; 301:13-27. [PMID: 30853527 DOI: 10.1016/j.jconrel.2019.03.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 12/17/2022]
Abstract
The regeneration of complex tissues and organs remains a major clinical challenge. With a view towards bioprinting such tissues, we developed a new class of pore-forming bioink to spatially and temporally control the presentation of therapeutic genes within bioprinted tissues. By blending sacrificial and stable hydrogels, we were able to produce bioinks whose porosity increased with time following printing. When combined with amphipathic peptide-based plasmid DNA delivery, these bioinks supported enhanced non-viral gene transfer to stem cells in vitro. By modulating the porosity of these bioinks, it was possible to direct either rapid and transient (pore-forming bioinks), or slower and more sustained (solid bioinks) transfection of host or transplanted cells in vivo. To demonstrate the utility of these bioinks for the bioprinting of spatially complex tissues, they were next used to zonally position stem cells and plasmids encoding for either osteogenic (BMP2) or chondrogenic (combination of TGF-β3, BMP2 and SOX9) genes within networks of 3D printed thermoplastic fibers to produce mechanically reinforced, gene activated constructs. In vivo, these bioprinted tissues supported the development of a vascularised, bony tissue overlaid by a layer of stable cartilage. When combined with multiple-tool biofabrication strategies, these gene activated bioinks can enable the bioprinting of a wide range of spatially complex tissues.
Collapse
Affiliation(s)
- T Gonzalez-Fernandez
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland
| | - S Rathan
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - C Hobbs
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; School of Physics, Trinity College Dublin, Ireland; Centre for Research of Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Ireland
| | - P Pitacco
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - F E Freeman
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - G M Cunniffe
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - N J Dunne
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Ireland; School of Mechanical and Manufacturing Engineering, Dublin City University, Ireland; School of Pharmacy, Queen's University Belfast, UK
| | - H O McCarthy
- School of Pharmacy, Queen's University Belfast, UK
| | - V Nicolosi
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; School of Physics, Trinity College Dublin, Ireland; Centre for Research of Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Ireland
| | - F J O'Brien
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in, Ireland
| | - D J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in, Ireland.
| |
Collapse
|
5
|
Taylor RW, Mitchell GK, Andrade JL, Svoboda KK. Expression of Collagen Types I, II, IX, and X in the Mineralizing Turkey Gastrocnemius Tendon. Anat Rec (Hoboken) 2019; 303:1664-1669. [DOI: 10.1002/ar.24091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 12/07/2018] [Accepted: 12/11/2018] [Indexed: 11/10/2022]
Affiliation(s)
| | - Georgia K. Mitchell
- Roseman University of Health Sciences, College of Dental Medicine Henderson Nevada
| | - Jon Lee Andrade
- Carnegie Mellon University Mellon College of Science Pittsburgh Pennsylvania
| | - Kathy K. Svoboda
- Texas A&M College of Dentistry, Biomedical Sciences Dallas Texas
| |
Collapse
|
6
|
Gonzalez-Fernandez T, Sathy B, Hobbs C, Cunniffe G, McCarthy H, Dunne N, Nicolosi V, O'Brien F, Kelly D. Mesenchymal stem cell fate following non-viral gene transfection strongly depends on the choice of delivery vector. Acta Biomater 2017; 55:226-238. [PMID: 28363788 DOI: 10.1016/j.actbio.2017.03.044] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/06/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022]
Abstract
Controlling the phenotype of mesenchymal stem cells (MSCs) through the delivery of regulatory genes is a promising strategy in tissue engineering (TE). Essential to effective gene delivery is the choice of gene carrier. Non-viral delivery vectors have been extensively used in TE, however their intrinsic effects on MSC differentiation remain poorly understood. The objective of this study was to investigate the influence of three different classes of non-viral gene delivery vectors: (1) cationic polymers (polyethylenimine, PEI), (2) inorganic nanoparticles (nanohydroxyapatite, nHA) and (3) amphipathic peptides (RALA peptide) on modulating stem cell fate after reporter and therapeutic gene delivery. Despite facilitating similar reporter gene transfection efficiencies, these nanoparticle-based vectors had dramatically different effects on MSC viability, cytoskeletal morphology and differentiation. After reporter gene delivery (pGFP or pLUC), the nHA and RALA vectors supported an elongated MSC morphology, actin stress fibre formation and the development of mature focal adhesions, while cells appeared rounded and less tense following PEI transfection. These changes in MSC morphology correlated with enhanced osteogenesis following nHA and RALA transfection and adipogenesis following PEI transfection. When therapeutic genes encoding for transforming growth factor beta 3 (TGF-β3) and/or bone morphogenic protein 2 (BMP2) were delivered to MSCs, nHA promoted osteogenesis in 2D culture and the development of an endochondral phenotype in 3D culture, while RALA was less osteogenic and appeared to promote a more stable hyaline cartilage-like phenotype. In contrast, PEI failed to induce robust osteogenesis or chondrogenesis of MSCs, despite effective therapeutic protein production. Taken together, these results demonstrate that the differentiation of MSCs through the application of non-viral gene delivery strategies depends not only on the gene delivered, but also on the gene carrier itself. STATEMENT OF SIGNIFICANCE Nanoparticle-based non-viral gene delivery vectors have been extensively used in regenerative medicine, however their intrinsic effects on mesenchymal stem cell (MSC) differentiation remain poorly understood. This paper demonstrates that different classes of commonly used non-viral vectors are not inert and they have a strong effect on cell morphology, stress fiber formation and gene transcription in MSCs, which in turn modulates their capacity to differentiate towards osteogenic, adipogenic and chondrogenic lineages. These results also point to the need for careful and tissue-specific selection of nanoparticle-based delivery vectors to prevent undesired phenotypic changes and off-target effects when delivering therapeutic genes to damaged or diseased tissues.
Collapse
|
7
|
Weiss-Bilka HE, McGann ME, Meagher MJ, Roeder RK, Wagner DR. Ectopic models for endochondral ossification: comparing pellet and alginate bead culture methods. J Tissue Eng Regen Med 2017; 12:e541-e549. [PMID: 27690279 DOI: 10.1002/term.2324] [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: 05/28/2015] [Revised: 07/11/2016] [Accepted: 09/26/2016] [Indexed: 01/13/2023]
Abstract
Key aspects of native endochondral bone development and fracture healing can be mimicked in mesenchymal stem cells (MSCs) through standard in vitro chondrogenic induction. Exploiting this phenomenon has recently emerged as an attractive technique to engineer bone tissue, however, relatively little is known about the best conditions for doing so. The objective of the present study was to compare the bone-forming capacity and angiogenic induction of hypertrophic cell constructs containing human adipose-derived stem cells (hASCs) primed for chondrogenesis in two different culture systems: high-density pellets and alginate bead hydrogels. The hASC constructs were subjected to 4 weeks of identical chondrogenic induction in vitro, encapsulated in an agarose carrier, and then implanted subcutaneously in immune-compromised mice for 8 weeks to evaluate their endochondral potential. At the time of implantation, both pellets and beads expressed aggrecan and type II collagen, as well as alkaline phosphatase (ALP) and type X collagen. Interestingly, ASCs in pellets formed a matrix containing higher glycosaminoglycan and collagen contents than that in beads, and ALP activity per cell was higher in pellets. However, after 8 weeks in vivo, pellets and beads induced an equivalent volume of mineralized tissue and a comparable level of vascularization. Although osteocalcin and osteopontin-positive osteogenic tissue and new vascular growth was found within both types of constructs, all appeared to be better distributed throughout the hydrogel beads. The results of this ectopic model indicate that hydrogel culture may be an attractive alternative to cell pellets for bone tissue engineering via the endochondral pathway. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Holly E Weiss-Bilka
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA.,Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Megan E McGann
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Matthew J Meagher
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
| | - Ryan K Roeder
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Diane R Wagner
- Department of Mechanical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
8
|
Lee SH, Manandhar S, Lee YM. Roles of RUNX in Hypoxia-Induced Responses and Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 962:449-469. [PMID: 28299673 DOI: 10.1007/978-981-10-3233-2_27] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During the past two decades, Runt domain transcription factors (RUNX1, 2, and 3) have been investigated in regard to their function, structural elements, genetic variants, and roles in normal development and pathological conditions. The Runt family proteins are evolutionarily conserved from Drosophila to mammals, emphasizing their physiological importance. A hypoxic microenvironment caused by insufficient blood supply is frequently observed in developing organs, growing tumors, and tissues that become ischemic due to impairment or blockage of blood vessels. During embryonic development and tumor growth, hypoxia triggers a stress response that overcomes low-oxygen conditions by increasing erythropoiesis and angiogenesis and triggering metabolic changes. This review briefly introduces hypoxic conditions and cellular responses, as well as angiogenesis and its related signaling pathways, and then describes our current knowledge on the functions and molecular mechanisms of Runx family proteins in hypoxic responses, especially in angiogenesis.
Collapse
Affiliation(s)
- Sun Hee Lee
- National Basic Research Laboratory of Vascular Homeostasis Regulation, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, 41566, South Korea
| | - Sarala Manandhar
- National Basic Research Laboratory of Vascular Homeostasis Regulation, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, 41566, South Korea
| | - You Mie Lee
- National Basic Research Laboratory of Vascular Homeostasis Regulation, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
9
|
Bone response of broiler chickens (Gallus gallus domesticus) induced by corticosterone. Comp Biochem Physiol A Mol Integr Physiol 2013; 164:410-6. [DOI: 10.1016/j.cbpa.2012.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/28/2012] [Accepted: 12/01/2012] [Indexed: 11/22/2022]
|
10
|
Sun H, Yang F, Chu W, Zhao H, McMahon C, Li C. Lentiviral-mediated RNAi knockdown of Cbfa1 gene inhibits endochondral ossification of antler stem cells in micromass culture. PLoS One 2012; 7:e47367. [PMID: 23056636 PMCID: PMC3467256 DOI: 10.1371/journal.pone.0047367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 09/12/2012] [Indexed: 01/02/2023] Open
Abstract
Articular cartilage (AC) lacks ability to repair defects due to its avascular nature as healing process relies on cells being brought in by blood vessels. Multiple approaches have been taken to facilitate cartilage repair in clinics, to date there is no effective treatment available that can restores the AC lesion to a normally functioning level over extended periods. In this regard, antler cartilage is unique in being richly vascularised and hence can effectively repair and regenerate. Interestingly, antler stem cells, from which the vascularised cartilage is derived, can form avascular cartilage when taken away from their original niche, suggesting that the vascular or avascular state of antler cartilage is controlled by extrinsic factors. Understanding the mechanisms underlying this phenotype switch may help us to devise a way to trigger the effective intrinsic repair of AC. However, adoption of antler cartilage model for AC repair requires the demonstration that the cartilage specific signalling pathways also prevail in antler chondrogenesis. To achieve this, in the present study we silenced expression of Cbfa1, a key factor regulatingendochondral ossification, using RNAi, and showed that expression of the downstream genes type I collagen and osteocalcin were suppressed which, in turn, inhibited endochondral ossification process taking place in the antler stem cell-formed nodules. Therefore, we provided further evidence at molecular level that antler could be developed as novel model for the study of AC repair. The eventual identification of the extrinsic factors dictating the phenotype switch between the vascular and avascular state of antler cartilage will open up a new avenue for the cure of osteoarthritis.
Collapse
Affiliation(s)
- Hongmei Sun
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun, Jilin, P. R. China
- State Key Laboratory for Molecular Biology of Special Economic Animals, Jilin, P. R. China
| | - Fuhe Yang
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun, Jilin, P. R. China
- State Key Laboratory for Molecular Biology of Special Economic Animals, Jilin, P. R. China
| | - Wenhui Chu
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun, Jilin, P. R. China
- State Key Laboratory for Molecular Biology of Special Economic Animals, Jilin, P. R. China
| | - Haiping Zhao
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun, Jilin, P. R. China
- State Key Laboratory for Molecular Biology of Special Economic Animals, Jilin, P. R. China
| | - Chris McMahon
- AgResearch, Invermay Agricultural Centre, Mosgiel, New Zealand
| | - Chunyi Li
- AgResearch, Invermay Agricultural Centre, Mosgiel, New Zealand
- State Key Laboratory for Molecular Biology of Special Economic Animals, Jilin, P. R. China
- * E-mail:
| |
Collapse
|
11
|
Lee SH, Che X, Jeong JH, Choi JY, Lee YJ, Lee YH, Bae SC, Lee YM. Runx2 protein stabilizes hypoxia-inducible factor-1α through competition with von Hippel-Lindau protein (pVHL) and stimulates angiogenesis in growth plate hypertrophic chondrocytes. J Biol Chem 2012; 287:14760-71. [PMID: 22351759 DOI: 10.1074/jbc.m112.340232] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The regulation of hypoxia-inducible factor-1α (HIF-1α) during endochondral bone formation is not fully understood. Here, we investigated the cross-talk between HIF-1α and Runt-related transcription factor 2 (Runx2) in the growth plate. Runx2 caused the accumulation of HIF-1α protein in ATDC5 chondrocytes and HEK293 cells under normoxic conditions. Runx2 also increased the nuclear translocation of HIF-1α when coexpressed in HEK293 cells and interacted with HIF-1α at the oxygen-dependent degradation domain (ODDD). In addition, Runx2 competed with von Hippel-Lindau tumor suppressor protein by directly binding to ODDD-HIF-1α and significantly inhibited the ubiquitination of HIF-1α, even though Runx2 did not change the hydroxylation status of HIF-1α. Furthermore, overexpression of Runx2 resulted in the significant enhancement of vascular endothelial growth factor (VEGF) promoter reporter activity and protein secretion. Runx2 significantly increased angiogenic activity in human umbilical vein endothelial cells in vitro. In wild-type mice, HIF-1α and Runx2 were colocalized in hypertrophic chondrocytes in which the cluster of differentiation 31 (CD31) protein was expressed at embryonic day 15.5 (E15.5). In contrast, the expression of HIF-1α was markedly reduced in areas of CD31 expression in Runx2(-/-) mice. These results suggest that Runx2 stabilizes HIF-1α by binding to ODDD to block the interaction between von Hippel-Lindau protein and HIF-1α. In conclusion, Runx2, HIF-1α, and VEGF may regulate vascular angiogenesis spatially and temporally in the hypertrophic zone of the growth plate during endochondral bone formation.
Collapse
Affiliation(s)
- Sun-Hee Lee
- School of Life Sciences and Biotechnology, College of Natural Sciences, Daegu 702-701, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Cartilaginous repair of full-thickness articular cartilage defects is induced by the intermittent activation of PTH/PTHrP signaling. Osteoarthritis Cartilage 2011; 19:886-94. [PMID: 21571083 DOI: 10.1016/j.joca.2011.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/05/2011] [Accepted: 04/16/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We studied the effects of the transient activation of parathyroid hormone (PTH)/PTH-related peptide (PTHrP) signaling during the repair of 5-mm-diameter full-thickness defects of articular cartilage in the rabbit. MATERIALS AND METHODS Cylindrical full-thickness articular cartilage defects of 5mm in diameter were artificially created in the femoral trochlea of male adolescent Japanese white rabbits using a hand-drill. Recombinant human PTH(1-84) was then administered into the joint cavity continuously or intermittently for 2 weeks post-injury. The reparative tissues were histologically examined at 2, 4, and 8 weeks, and were also immunohistochemically examined for type II collagen. Double immunostaining analysis was also performed for the PTH/PTHrP receptor and proliferating cell nuclear antigen (PCNA) in the regenerating tissues. RESULTS No evidence of cartilage formation was evident throughout the period of the experiments in injured animals administered saline alone. In contrast, cartilage formation occurred at 4 weeks in both the continuous and intermittent PTH-treated defects. At 8 weeks post-injury, for the intermittently treated defects, the regenerated cartilage successfully resurfaced the defects and the original bone-articular cartilage junction was recovered. In contrast, the defects were covered with fibrous or fibrocartilaginous tissues in the continuously administered group. PCNA and PTH/PTHrP receptor-double positive mesenchymal cells were significantly increased in both the continuous and intermittent PTH-treated defects at 2 weeks post-injury. CONCLUSIONS The present results suggest that the transient activation and release from PTH/PTHrP signaling during the early stages of the cartilage repair process facilitates the induction of regenerative chondrogenesis in full-thickness articular cartilage defects.
Collapse
|
13
|
Maye P, Fu Y, Butler DL, Chokalingam K, Liu Y, Floret J, Stover ML, Wenstrup R, Jiang X, Gooch C, Rowe D. Generation and characterization of Col10a1-mcherry reporter mice. Genesis 2011; 49:410-8. [PMID: 21328521 PMCID: PMC5638041 DOI: 10.1002/dvg.20733] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We report here on the generation of a new fluorescent protein reporter transgenic mouse line, Col10a1-mCherry, which can be used as a tool to study chondrocyte biology and pathology. Collagen, Type X, alpha 1 (Col10a1) is highly expressed in hypertrophic chondrocytes and commonly used as a gene marker for this cell population. The Col10a1-mCherry reporter line was generated using a bacterial recombination strategy with the mouse BAC clone RP23-192A7. To aid in the characterization of this animal model, we intercrossed Col10a1-mCherry mice with Collagen, Type II, alpha 1 (Col2a1) enhanced cyan fluorescent protein (ECFP) reporter mice and characterized the expression of both chondrocyte reporters during embryonic skeletal development from days E10.5 to E17.5. Additionally, at postnatal day 0, Col10a1-mCherry reporter expression was compared to endogenous Col10a1 mRNA expression in long bones and revealed that mCherry fluorescence extended past the Col10a1 expression domain. However, in situ hybridization for mCherry was consistent with the zone of Col10a1 mRNA expression, indicating that the persistent detection of mCherry fluorescence was a result of the long protein half life of mCherry in conjunction with a very rapid phase of skeletal growth and not due to aberrant transcriptional regulation. Taking advantage of the continued fluorescence of hypertrophic chondrocytes at the chondro-osseus junction, we intercrossed Col10a1-mCherry mice with two different Collagen, Type 1, alpha 1, (Col1a1) osteoblast reporter mice, pOBCol3.6-Topaz and pOBCol2.3-Emerald to investigate the possibility that hypertrophic chondrocytes transdifferentiate into osteoblasts. Evaluation of long bones at birth suggests that residual hypertrophic chondrocytes and osteoblasts in the trabecular zone exist as two completely distinct cell populations. genesis 49:410-418, 2011.
Collapse
Affiliation(s)
- Peter Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut 06030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wang S, Qiu Y, Ma Z, Xia C, Zhu F, Zhu Z. Expression of Runx2 and type X collagen in vertebral growth plate of patients with adolescent idiopathic scoliosis. Connect Tissue Res 2010; 51:188-96. [PMID: 20073986 DOI: 10.3109/03008200903215590] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The different expression of type X collagen and Runx2 between the convex and concave side of vertebral growth plate in scoliosis may help to improve our understanding of the role that growth plate tissue play in the development or progression of idiopathic scoliosis. In this investigation, there were significant differences of the total expression of type X collagen, Runx2 protein, and Runx2 mRNA between convex side and concave side growth plates of the apex vertebrae (p < 0.05). The total expression of type X collagen in the concave side growth plates of the lower end vertebrae was higher than that in the same side growth plates of apex (p < 0.05). The total expression of Runx2 in the concave side growth plates in the upper and lower end vertebrae were higher than that in the concave side growth plates of apex (p < 0.05). The expression of type X collagen, Runx2, and Runx2 mRNA, the cell density of type X collagen and Runx2 positive chondrocytes, and histological changes between convex side and concave side of the vertebral growth plate indicated that the vertebral growth plate was affected by mechanical forces, which was a secondary change and could contribute to progression of adolescent idiopathic scoliosis.
Collapse
Affiliation(s)
- Shoufeng Wang
- Spine Surgery, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | | | | | | | | | | |
Collapse
|
15
|
Dreier R, Günther BK, Mainz T, Nemere I, Bruckner P. Terminal differentiation of chick embryo chondrocytes requires shedding of a cell surface protein that binds 1,25-dihydroxyvitamin D3. J Biol Chem 2007; 283:1104-12. [PMID: 17984094 DOI: 10.1074/jbc.m703336200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Endochondral ossification comprises a cascade of cell differentiation culminating in chondrocyte hypertrophy and is negatively controlled by soluble environmental mediators at several checkpoints. Proteinases modulate this control by processing protein signals and/or their receptors. Here, we show that insulin-like growth factor I can trigger hypertrophic development by stimulating production and/or activation of proteinases in some populations of chick embryo chondrocytes. Cell surface targets of the enzymes include 1,25-dihydroxyvitamin D3 membrane-associated rapid response steroid receptor (1,25 D3 MARRS receptor), also known as ERp57/GRp58/ERp60. This protein is anchored to the outer surface of plasma membranes and inhibits late chondrocyte differentiation after binding of 1,25-dihydroxyvitamin D3. Upon treatment with insulin-like growth factor I, 1,25 D3 MARRS receptor is cleaved into two fragments of approximately 30 and 22 kDa. This process is abrogated along with hypertrophic development by E-64 or cystatin C, inhibitors of cysteine proteinases. Cell differentiation is enhanced by treatment with antibodies to 1,25 D3 MARRS receptor that either block binding of the inhibitory ligand 1,25-dihydroxyvitamin D3 or inactivate 1,25 D3 MARRS receptor left intact after treatment with proteinase inhibitors. Therefore, proteolytic shedding of 1,25 D3 MARRS receptor constitutes a molecular mechanism eliminating the 1,25-dihydroxyvitamin D3-induced barrier against late cartilage differentiation and is a potentially important step during endochondral ossification or cartilage degeneration in osteoarthritis.
Collapse
Affiliation(s)
- Rita Dreier
- Institute for Physiological Chemistry and Pathobiochemistry, University Hospital of Münster, 48149 Münster, Germany
| | | | | | | | | |
Collapse
|
16
|
Abzhanov A, Rodda SJ, McMahon AP, Tabin CJ. Regulation of skeletogenic differentiation in cranial dermal bone. Development 2007; 134:3133-44. [PMID: 17670790 DOI: 10.1242/dev.002709] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although endochondral ossification of the limb and axial skeleton is relatively well-understood, the development of dermal (intramembranous) bone featured by many craniofacial skeletal elements is not nearly as well-characterized. We analyzed the expression domains of a number of markers that have previously been associated with endochondral skeleton development to define the cellular transitions involved in the dermal ossification process in both chick and mouse. This led to the recognition of a series of distinct steps in the dermal differentiation pathways, including a unique cell type characterized by the expression of both osteogenic and chondrogenic markers. Several signaling molecules previously implicated in endochondrial development were found to be expressed during specific stages of dermal bone formation. Three of these were studied functionally using retroviral misexpression. We found that activity of bone morphogenic proteins (BMPs) is required for neural crest-derived mesenchyme to commit to the osteogenic pathway and that both Indian hedgehog (IHH) and parathyroid hormone-related protein (PTHrP, PTHLH)negatively regulate the transition from preosteoblastic progenitors to osteoblasts. These results provide a framework for understanding dermal bone development with an aim of bringing it closer to the molecular and cellular resolution available for the endochondral bone development.
Collapse
Affiliation(s)
- Arhat Abzhanov
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
17
|
Nakaoka R, Hsiong SX, Mooney DJ. Regulation of chondrocyte differentiation level via co-culture with osteoblasts. TISSUE ENGINEERING 2006; 12:2425-33. [PMID: 16995776 DOI: 10.1089/ten.2006.12.2425] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The close apposition of osteoblasts and chondrocytes in bone and their interaction during bone development and regeneration suggest that they may each regulate the other's growth and differentiation. In these studies, osteoblasts and chondrocytes were co-cultured in vitro, with both direct and indirect contact. Proliferation of the co-cultured chondrocytes was enhanced using soluble factors produced from the osteoblasts, and the differentiation level of the osteoblasts influenced the differentiation level of the chondrocytes. In addition, the chondrocytes regulated differentiation of the co-cultured osteoblasts using soluble factors and direct contact. These data support the possibility of direct, reciprocal instructive interactions between chondrocytes and osteoblasts in a variety of normal processes and further suggest that it may be necessary to account for this signaling in the regeneration of complex tissues comprising cartilage and mineralized tissue.
Collapse
Affiliation(s)
- Ryusuke Nakaoka
- Division of Medical Devices, National Institute of Health Sciences, Tokyo, Japan.
| | | | | |
Collapse
|
18
|
Mizuta H, Kudo S, Nakamura E, Takagi K, Hiraki Y. Expression of the PTH/PTHrP receptor in chondrogenic cells during the repair of full-thickness defects of articular cartilage. Osteoarthritis Cartilage 2006; 14:944-52. [PMID: 16644246 DOI: 10.1016/j.joca.2006.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2005] [Accepted: 03/11/2006] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We studied the accumulation of parathyroid hormone (PTH)/PTHrP receptor-positive mesenchymal cells using double immunohistochemistry and examined whether this correlated with the subsequent regeneration of 3-mm-diameter full-thickness defects of articular cartilage. MATERIALS AND METHODS Cylindrical full-thickness articular cartilage defects (3 mm) were artificially created in the femoral trochlea of male adolescent Japanese white rabbits (n = 210) with a hand-drill. Recombinant human PTH(1-84) was then administered into the defect cavities with an osmotic pump for either 2 or 4 weeks post-injury. Following PTH treatment, the repair processes in the cartilage defects were histologically examined. Double immunostaining analyses for the PTH/PTH-related peptide (PTHrP) receptor and proliferating cell nuclear antigen (PCNA) in the regenerating tissues were then performed. RESULTS Activation of PTH/PTHrP receptor signaling by hPTH(1-84) results in the inhibition of chondrogenic differentiation in full-thickness articular cartilage defects. At the conclusion of the 2-week PTH treatment, the defect cavities were filled with undifferentiated mesenchymal cells, which were similar to the controls. In addition, almost all of these cells localized at the center of the injuries were both PTH/PTHrP receptor- and PCNA-positive. In contrast, after prolonged PTH treatment for 4 weeks, there was no indication of a cartilaginous repair response and cells that had migrated to the defect cavities were found to have irreversibly lost expression of the PTH/PTHrP receptor. CONCLUSIONS The chondrogenic capacity of cells that had migrated to the area of these defect cavities is closely associated with their ability to express the PTH/PTHrP receptor. Moreover, these cells maintain their chondrogenic potential within only a limited time-span of 2 weeks.
Collapse
Affiliation(s)
- H Mizuta
- Department of Orthopaedic and Neuro-Musculoskeletal Surgery, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | | | | | | | | |
Collapse
|
19
|
Singh MK, Petry M, Haenig B, Lescher B, Leitges M, Kispert A. The T-box transcription factor Tbx15 is required for skeletal development. Mech Dev 2005; 122:131-44. [PMID: 15652702 DOI: 10.1016/j.mod.2004.10.011] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 10/24/2004] [Accepted: 10/25/2004] [Indexed: 11/27/2022]
Abstract
During early limb development several signaling centers coordinate limb bud outgrowth as well as patterning. Members of the T-box gene family of transcriptional regulators are crucial players in these processes by activating and interpreting these signaling pathways. Here, we show that Tbx15, a member of this gene family, is expressed during limb development, first in the mesenchyme of the early limb bud, then during early endochondral bone development in prehypertrophic chondrocytes of cartilaginous templates. Expression is also found in mesenchymal precursor cells and prehypertrophic chondrocytes, respectively, during development of skeletal elements of the vertebral column and the head. Analysis of Tbx15 null mutant mice indicates a role of Tbx15 in the development of skeletal elements throughout the body. Mutants display a general reduction of bone size and changes of bone shape. In the forelimb skeleton, the scapula lacks the central region of the blade. Cartilaginous templates are already reduced in size and show a transient delay in ossification in mutant embryos. Mutants show a significantly reduced proliferation of prehypertrophic chondrocytes as well as of mesenchymal precursor cells. These data suggest that Tbx15 plays an important role in the development of the skeleton of the limb, vertebral column and head by controlling the number of mesenchymal precursor cells and chondrocytes.
Collapse
Affiliation(s)
- Manvendra K Singh
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Shimo T, Koyama E, Sugito H, Wu C, Shimo S, Pacifici M. Retinoid signaling regulates CTGF expression in hypertrophic chondrocytes with differential involvement of MAP kinases. J Bone Miner Res 2005; 20:867-77. [PMID: 15824860 DOI: 10.1359/jbmr.041235] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Revised: 12/21/2004] [Accepted: 12/21/2004] [Indexed: 02/02/2023]
Abstract
UNLABELLED Retinoids are important for growth plate chondrocyte maturation, but their downstream effectors remain unclear. Recently, CTGF (CCN2) was found to regulate chondrocyte function, particularly in the hypertrophic zone. The goal of the study was to determine whether CTGF is a retinoid signaling effector molecule, how it is regulated, and how it acts. INTRODUCTION Using a combination of in vivo and in vitro approaches, we carried out a series of studies at the cellular, biochemical, and molecular level to determine whether and how retinoid signaling is related to expression and function of connective tissue growth factor (CTGF) in chondrocyte maturation and endochondral ossification. MATERIALS AND METHODS Limbs of chick embryos in ovo were implanted with retinoic pan-antagonist RO 41-5253-filled beads, and phenotypic changes were assessed by in situ hybridization. CTGF gene expression and roles were tested in primary cultures of immature and hypertrophic chondrocytes. Cross-talk between retinoid signaling and other pathways was tested by determining endogenous levels of active ERK1/2 and p38 MAP kinases and phenotypic modulations exerted by specific antagonists of mitogen-activated protein (MAP) kinases and BMP signaling (Noggin). RESULTS Interference with retinoid signaling blocked expression of CTGF and other posthypertrophic markers in long bone anlagen in vivo and hypertrophic chondrocyte cultures, whereas all-trans-retinoic acid (RA) boosted CTGF expression and even induced it in immature proliferating cultures. Exogenous recombinant CTGF stimulated chondrocyte maturation, but failed to do so in presence of retinoid antagonists. Immunoblots showed that hypertrophic chondrocytes contained sizable levels of phosphorylated ERK1/2 and p38 MAP kinases that were dose- and time-dependently increased by RA treatment. Experimental ERK1/2 inhibition led to a severe drop in baseline and RA-stimulated CTGF expression, whereas p38 inhibition increased it markedly. These responses were gene-specific, because the opposite was seen with other hypertrophic chondrocyte genes such as collagen X and RA receptor gamma (RARgamma). Tests with Noggin showed that RA induction of CTGF expression was negatively influenced by BMP signaling, whereas induction of collagen X expression was BMP-dependent. CONCLUSIONS Retinoids appear to have a preeminent role in controlling expression and function of CTGF in hypertrophic and posthypertrophic chondrocytes and do so with differential cooperation and intervention of MAP kinases and BMP signaling.
Collapse
Affiliation(s)
- Tsuyoshi Shimo
- Department of Orthopedic Surgery, Thomas Jefferson University College of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Heng BC, Cao T, Stanton LW, Robson P, Olsen B. Strategies for directing the differentiation of stem cells into the osteogenic lineage in vitro. J Bone Miner Res 2004; 19:1379-94. [PMID: 15312238 DOI: 10.1359/jbmr.040714] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 03/02/2004] [Accepted: 05/18/2004] [Indexed: 01/29/2023]
Abstract
A major area in regenerative medicine is the application of stem cells in bone reconstruction and bone tissue engineering. This will require well-defined and efficient protocols for directing the differentiation of stem cells into the osteogenic lineage, followed by their selective purification and proliferation in vitro. The development of such protocols would reduce the likelihood of spontaneous differentiation of stem cells into divergent lineages on transplantation, as well as reduce the risk of teratoma formation in the case of embryonic stem cells. Additionally, such protocols could provide useful in vitro models for studying osteogenesis and bone development, and facilitate the genetic manipulation of stem cells for therapeutic applications. The development of pharmokinetic and cytotoxicity/genotoxicity screening tests for bone-related biomaterials and drugs could also use protocols developed for the osteogenic differentiation of stem cells. This review critically examines the various strategies that could be used to direct the differentiation of stem cells into the osteogenic lineage in vitro.
Collapse
Affiliation(s)
- Boon Chin Heng
- Faculty of Dentistry, National University of Singapore, 119074 Singapore
| | | | | | | | | |
Collapse
|
22
|
Mizuta H, Kudo S, Nakamura E, Otsuka Y, Takagi K, Hiraki Y. Active proliferation of mesenchymal cells prior to the chondrogenic repair response in rabbit full-thickness defects of articular cartilage. Osteoarthritis Cartilage 2004; 12:586-96. [PMID: 15219574 DOI: 10.1016/j.joca.2004.04.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Accepted: 04/09/2004] [Indexed: 02/02/2023]
Abstract
OBJECTIVES In full-thickness articular defects, fibroblast growth factor-2 (FGF-2) participates in the chondrogenic repair response which occurs in a defect-size dependent manner. Here we demonstrate that FGF-2 plays a critical role in the proliferation of pre-chondrogenic mesenchymal cells during chondrogenic induction. METHODS Three-millimeter- or 5-mm-diameter cylindrical defects were created in the femoral trochlea of the rabbit knee. The defects received sterile saline or FGF-2 (50 pg/h) via an osmotic pump for the initial 2 weeks. We assessed the proliferative capacity of undifferentiated mesenchymal cells in the reparative tissue with the anti-proliferating cell nuclear antigen (PCNA) monoclonal antibody. Using a total of 180 rabbits, we performed three sets of experiments. RESULTS In the 3-mm-diameter defects, undifferentiated mesenchymal cells spontaneously initiated chondrogenic differentiation within 2 weeks, resulting in the regeneration of surfacing articular cartilage concomitantly with the repair of subchondral bone. No evidence of chondrogenesis was seen in the 5-mm-diameter defects, whereas application of FGF-2 promoted successful regeneration of articular cartilage. In the 3-mm-diameter defects and in the FGF-2-treated 5-mm defects, PCNA immunoreactivity was widely detected in undifferentiated cells in the reparative tissue at 1 and 2 weeks after creation of the defects. In contrast, in the 5-mm-diameter defects without FGF-2 treatment, the PCNA-positive cells were found at a significantly lower incidence. CONCLUSIONS Active expansion of undifferentiated cell population mediated by FGF-2 is required to initiate and support a chondrogenic repair response in full-thickness defects of articular cartilage. Endogenous FGF-2 could not meet the requirements of growth signaling in the center of larger sized defects.
Collapse
Affiliation(s)
- Hiroshi Mizuta
- Department of Orthopaedic and Neuro-Musculoskeletal Surgery, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | | | | | | | | | | |
Collapse
|
23
|
Kuorilehto T, Nissinen M, Koivunen J, Benson MD, Peltonen J. NF1 tumor suppressor protein and mRNA in skeletal tissues of developing and adult normal mouse and NF1-deficient embryos. J Bone Miner Res 2004; 19:983-9. [PMID: 15125795 DOI: 10.1359/jbmr.040130] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2003] [Revised: 11/03/2003] [Accepted: 01/23/2004] [Indexed: 11/18/2022]
Abstract
UNLABELLED NF1 is a heritable disease with multiple osseous lesions. The expression of the NF1 gene was studied in embryonic and adult rodent skeleton and in NF1-deficient embryos. The NF1 gene was expressed intensely in the cartilage and the periosteum. Impaired NF1 expression may lead to inappropriate development and dynamics of bones and ultimately to the osseous manifestations of the disease. INTRODUCTION Neurofibromatosis type 1 is caused by mutations in the NF1 gene encoding the Ras GTPase activating protein (Ras-GAP) neurofibromin. Skeletal ailments such as short stature, kyphoscoliosis, and tibial bowing and pseudarthrosis are common osseous manifestations of NF1. These symptoms are congenital, implying a role for neurofibromin in proper bone growth. However, little is known about its expression in skeletal tissues during their development. MATERIALS AND METHODS The expression of the NF1 gene was studied in normal and NF1+/- mouse fetuses at embryonic days 12.5-15.5 and in skeletal tissues of adult mice and rats. In situ hybridization, immunohistochemistry, and Western blot analysis were used to identify the NF1 gene expression profile. RESULTS NF1 mRNA and protein were elevated in resting, maturation, and hypertrophic chondrocytes at the growth plate. Parallel studies on NF1+/- embryos showed expression patterns identical to wildtype. The periosteum, including osteoblasts and osteoclasts, and osteocytes of the cortical bone of adult mice were also intensely labeled for NF1 protein and mRNA. Western transfer analysis detected NF1 protein in the respective rat tissues. Phosphorylation of p42 and p44 MAP kinases, the downstream consequence of Ras activation, was elevated in hypertrophic chondrocytes of NF1+/- embryos. CONCLUSIONS The results suggest that neurofibromin may act as a Ras-GAP in skeletal cells to attenuate Ras transduced growth signals and thus play a role during ossification and dynamics of bone. Loss of NF1 function may therefore lead to dysplastic bone growth, thereby causing the debilitating osseous symptoms of NF1.
Collapse
Affiliation(s)
- Tommi Kuorilehto
- Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland
| | | | | | | | | |
Collapse
|
24
|
Abstract
Manipulation of the mouse genome through mis-expressing, knocking out, and introducing mutations into genes of interest has provided important insights into the genetic pathways responsible for human skeletal development. These pathways contribute to the sequential phases of skeletal morphogenesis that include patterning, condensation, and overt organogenesis of the membranous and endochondral embryonic skeletons and to subsequent linear growth. Disturbances in these pathways account for many developmental syndromes and disorders of the human skeleton. Recurrent themes include establishment of interlocking regulatory circuits involving growth factors, receptors, signalling pathways, and transcription factors that control cellular programmes such as migration, adhesion, proliferation, differentiation, and apoptosis, and use of common molecules for different purposes. Technical advances suggest that genetic engineering in mice will continue to be highly instructive in the field of skeletal biology.
Collapse
Affiliation(s)
- William A Horton
- Shriners Hospital for Children, Oregon Health and Science University, 3101 Sam Jackson Park Road, Portland, OR 97239-3009, USA.
| |
Collapse
|
25
|
Naumann A, Dennis JE, Awadallah A, Carrino DA, Mansour JM, Kastenbauer E, Caplan AI. Immunochemical and mechanical characterization of cartilage subtypes in rabbit. J Histochem Cytochem 2002; 50:1049-58. [PMID: 12133908 DOI: 10.1177/002215540205000807] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cartilage is categorized into three general subgroups, hyaline, elastic, and fibrocartilage, based primarily on morphologic criteria and secondarily on collagen (Types I and II) and elastin content. To more precisely define the different cartilage subtypes, rabbit cartilage isolated from joint, nose, auricle, epiglottis, and meniscus was characterized by immunohistochemical (IHC) localization of elastin and of collagen Types I, II, V, VI, and X, by biochemical analysis of total glycosaminoglycan (GAG) content, and by biomechanical indentation assay. Toluidine blue staining and safranin-O staining were used for morphological assessment of the cartilage subtypes. IHC staining of the cartilage samples showed a characteristic pattern of staining for the collagen antibodies that varied in both location and intensity. Auricular cartilage is discriminated from other subtypes by interterritorial elastin staining and no staining for Type VI collagen. Epiglottal cartilage is characterized by positive elastin staining and intense staining for Type VI collagen. The unique pattern for nasal cartilage is intense staining for Type V collagen and collagen X, whereas articular cartilage is negative for elastin (interterritorially) and only weakly positive for collagen Types V and VI. Meniscal cartilage shows the greatest intensity of staining for Type I collagen, weak staining for collagens V and VI, and no staining with antibody to collagen Type X. Matching cartilage samples were categorized by total GAG content, which showed increasing total GAG content from elastic cartilage (auricle, epiglottis) to fibrocartilage (meniscus) to hyaline cartilage (nose, knee joint). Analysis of aggregate modulus showed nasal and auricular cartilage to have the greatest stiffness, epiglottal and meniscal tissue the lowest, and articular cartilage intermediate. This study illustrates the differences and identifies unique characteristics of the different cartilage subtypes in rabbits. The results provide a baseline of data for generating and evaluating engineered repair cartilage tissue synthesized in vitro or for post-implantation analysis.
Collapse
Affiliation(s)
- Andreas Naumann
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
26
|
Ohbayashi N, Shibayama M, Kurotaki Y, Imanishi M, Fujimori T, Itoh N, Takada S. FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis. Genes Dev 2002; 16:870-9. [PMID: 11937494 PMCID: PMC186331 DOI: 10.1101/gad.965702] [Citation(s) in RCA: 340] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Fibroblast growth factor (FGF) signaling is involved in skeletal development of the vertebrate. Gain-of-function mutations of FGF receptors (FGFR) cause craniosynostosis, premature fusion of the skull, and dwarfism syndromes. Disruption of Fgfr3 results in prolonged growth of long bones and vertebrae. However, the role that FGFs actually play in skeletal development in the embryo remains unclear. Here we show that Fgf18 is expressed in and required for osteogenesis and chondrogenesis in the mouse embryo. Fgf18 is expressed in both osteogenic mesenchymal cells and differentiating osteoblasts during calvarial bone development. In addition, Fgf18 is expressed in the perichondrium and joints of developing long bones. In calvarial bone development of Fgf18-deficient mice generated by gene targeting, the progress of suture closure is delayed. Furthermore, proliferation of calvarial osteogenic mesenchymal cells is decreased, and terminal differentiation to calvarial osteoblasts is specifically delayed. Delay of osteogenic differentiation is also observed in the developing long bones of this mutant. Conversely, chondrocyte proliferation and the number of differentiated chondrocytes are increased. Therefore, FGF18 appears to regulate cell proliferation and differentiation positively in osteogenesis and negatively in chondrogenesis.
Collapse
Affiliation(s)
- Norihiko Ohbayashi
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Zelzer E, Glotzer DJ, Hartmann C, Thomas D, Fukai N, Soker S, Olsen BR. Tissue specific regulation of VEGF expression during bone development requires Cbfa1/Runx2. Mech Dev 2001; 106:97-106. [PMID: 11472838 DOI: 10.1016/s0925-4773(01)00428-2] [Citation(s) in RCA: 249] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a critical regulator of angiogenesis during development, but little is known about the factors that control its expression. We provide the first example of tissue specific loss of VEGF expression as a result of targeting a single gene, Cbfa1/Runx2. During endochondral bone formation, invasion of blood vessels into cartilage is associated with upregulation of VEGF in hypertrophic chondrocytes and increased expression of VEGF receptors in the perichondrium. This upregulation is lacking in Cbfa1 deficient mice, and cartilage angiogenesis does not occur. Finally, over-expression of Cbfa1 in fibroblasts induces an increase in their VEGF mRNA level and protein production by stimulating VEGF transcription. The results demonstrate that Cbfa1 is a necessary component of a tissue specific genetic program that regulates VEGF during endochondral bone formation.
Collapse
Affiliation(s)
- E Zelzer
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Masuda I, Iyama KI, Halligan BD, Barbieri JT, Haas AL, McCarty DJ, Ryan LM. Variations in site and levels of expression of chondrocyte nucleotide pyrophosphohydrolase with aging. J Bone Miner Res 2001; 16:868-75. [PMID: 11341331 DOI: 10.1359/jbmr.2001.16.5.868] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The aim of this study was to identify changes in cartilage intermediate layer protein/nucleotide pyrophosphohydrolase (CILP/NTPPH) expression in articular cartilage during aging. Adult (3-4 years old) and young (7-10 days old) porcine articular hyaline cartilage and fibrocartilage were studied by Northern blot analysis, in situ hybridization, and immunohistochemistry using a complementary DNA (cDNA) probe encoding porcine CILP/NTPPH and antibody to a synthetic peptide corresponding to a CILP/NTPPH sequence. Northern blot analysis of chondrocytes showed lower expression of CILP/NTPPH messenger RNA (mRNA) in young cartilage than in adult cartilage. In adult cartilage, extracellular matrix from the surface to the middeep zone was immunoreactive for CILP/NTPPH, especially in the pericellular matrix surrounding the middeep zone chondrocytes. In young cartilage, chondrocytes were moderately immunoreactive for CILP/NTPPH throughout all zones except the calcified zone. The matrix of young cartilage was negative except in the superficial zone. In young cartilage, CILP/NTPPH mRNA expression was undetectable. In adult cartilage, chondrocytes showed strong mRNA expression for CILP/NTPPH throughout middeep zones. Protein and mRNA signals were not detectable below the tidemark. CILP/NTPPH secretion into matrix around chondrocytes increases with aging. In this extracellular site it may generate inorganic pyrophosphate and contribute to age-related calcium pyrophosphate dihydrate crystal deposition disease.
Collapse
Affiliation(s)
- I Masuda
- Department of Medicine, Medical College of Wisconsin, Milwaukee 53226, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Early development of the vertebrate skeleton depends on genes that pattern the distribution and proliferation of cells from cranial neural crest, sclerotomes, and lateral plate mesoderm into mesenchymal condensations at sites of future skeletal elements. Within these condensations, cells differentiate to chondrocytes or osteoblasts and form cartilages and bones under the control of various transcription factors. In most of the skeleton, organogenesis results in cartilage models of future bones; in these models cartilage is replaced by bone by the process of endochondral ossification. Lastly, through a controlled process of bone growth and remodeling the final skeleton is shaped and molded. Significant and exciting insights into all aspects of vertebrate skeletal development have been obtained through molecular and genetic studies of animal models and humans with inherited disorders of skeletal morphogenesis, organogenesis, and growth.
Collapse
Affiliation(s)
- B R Olsen
- Harvard Medical School, Department of Cell Biology, 240 Longwood Avenue, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
30
|
Yamasaki A, Itabashi M, Sakai Y, Ito H, Ishiwari Y, Nagatsuka H, Nagai N. Expression of type I, type II, and type X collagen genes during altered endochondral ossification in the femoral epiphysis of osteosclerotic (oc/oc) mice. Calcif Tissue Int 2001; 68:53-60. [PMID: 12037624 DOI: 10.1007/bf02685003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2000] [Accepted: 08/15/2000] [Indexed: 10/22/2022]
Abstract
The osteosclerotic (oc/oc) mouse, a genetically distinct murine mutation that has a functional defect in its osteoclasts, also has rickets and shows an altered endochondral ossification in the epiphyseal growth plate. The disorder is morphologically characterized by an abnormal extension of hypertrophic cartilage at 10 days after birth, which is later (21 days after birth) incorporated into the metaphyseal woven bone without breakdown of the cartilage matrix following vascular invasion of chondrocyte lacunae. In situ hybridization revealed that the extending hypertrophic chondrocytes expressed type I and type II collagen mRNA, as well as that of type X collagen and that the osteoblasts in the metaphysis expressed type II and type X collagen mRNA, in addition to type I collagen mRNA. The topographic distribution of the signals suggests a possible co-expression of each collagen gene in the individual cells. Immunohistochemically, an overlapping deposition of type I, type II, and type X collagen was observed in both the extending cartilage and metaphyseal bony trabeculae. Such aberrant gene expression and synthesis of collagen indicate that pathologic ossification takes place in the epiphyseal/metaphyseal junction of oc/oc mouse femur in different way than in normal endochondral ossification. This abnormality is probably not due to a developmental disorder in the epiphyseal plate but to the failure in conversion of cartilage into bone, since the epiphyseal plate otherwise appeared normal, showing orderly stratified zones with a proper expression of cartilage-specific genes.
Collapse
Affiliation(s)
- A Yamasaki
- Department of Oral Pathology, Ohu University School of Dentistry, 31-1 Misumido, Tomita-machi, Koriyama, Fukushima 963-8611, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
Kudo S, Mizuta H, Otsuka Y, Takagi K, Hiraki Y. Inhibition of chondrogenesis by parathyroid hormone in vivo during repair of full-thickness defects of articular cartilage. J Bone Miner Res 2000; 15:253-60. [PMID: 10703926 DOI: 10.1359/jbmr.2000.15.2.253] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We studied the effects of parathyroid hormone (PTH) on differentiation of chondroprogenitor cells during the repair of full-thickness articular cartilage defects. Three-millimeter cylindrical full-thickness articular cartilage defects, which are small enough to be resurfaced spontaneously by hyaline cartilage, were created in the femoral trochlea of the rabbit knee. Recombinant human PTH(1-84) (hPTH[1-84]) (25 ng/h) then was administered into the joint cavity with an osmotic pump, or in control animals, saline alone was administered. The animals were killed at 1, 2, 4, and 8 weeks. At 1 week, the defects were filled with undifferentiated cells, regardless of the PTH treatments. By 8 weeks, well-developed cartilage covered the defects with reconstitution of subchondral bone up to the original bone-articular cartilage junction. In contrast, no evidence of chondrogenic differentiation was seen at any time during the experimental period in the defects treated with PTH. The reparative tissues also were examined immunohistochemically using anti-proliferating cell nuclear antigen (PCNA) and anti-PTH/PTH-related peptide (PTHrP) receptor antibodies. Interestingly, the chondroprogenitor cells that filled the defects expressed PTH/PTHrP receptor, suggesting that these cells are capable of responding to PTH/PTHrP signaling before overt chondrogenesis. Application of PTH did not interfere with proliferation but inhibited chondrogenic differentiation of the cells resulting in the formation of fibrous tissue that lost the expression of PTH/PTHrP receptor within 4 weeks.
Collapse
Affiliation(s)
- S Kudo
- Department of Orthopaedic Surgery, Kumamoto University School of Medicine, Japan
| | | | | | | | | |
Collapse
|
32
|
Yagami K, Suh JY, Enomoto-Iwamoto M, Koyama E, Abrams WR, Shapiro IM, Pacifici M, Iwamoto M. Matrix GLA protein is a developmental regulator of chondrocyte mineralization and, when constitutively expressed, blocks endochondral and intramembranous ossification in the limb. J Cell Biol 1999; 147:1097-108. [PMID: 10579728 PMCID: PMC2169349 DOI: 10.1083/jcb.147.5.1097] [Citation(s) in RCA: 137] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/1999] [Accepted: 10/20/1999] [Indexed: 11/22/2022] Open
Abstract
Matrix GLA protein (MGP), a gamma-carboxyglutamic acid (GLA)-rich, vitamin K-dependent and apatite-binding protein, is a regulator of hypertrophic cartilage mineralization during development. However, MGP is produced by both hypertrophic and immature chondrocytes, suggesting that MGP's role in mineralization is cell stage-dependent, and that MGP may have other roles in immature cells. It is also unclear whether MGP regulates the quantity of mineral or mineral nature and quality as well. To address these issues, we determined the effects of manipulations of MGP synthesis and expression in (a) immature and hypertrophic chondrocyte cultures and (b) the chick limb bud in vivo. The two chondrocyte cultures displayed comparable levels of MGP gene expression. Yet, treatment with warfarin, a gamma-carboxylase inhibitor and vitamin K antagonist, triggered mineralization in hypertrophic but not immature cultures. Warfarin effects on mineralization were highly selective, were accompanied by no appreciable changes in MGP expression, alkaline phosphatase activity, or cell number, and were counteracted by vitamin K cotreatment. Scanning electron microscopy, x-ray microanalysis, and Fourier-transform infrared spectroscopy revealed that mineral forming in control and warfarin-treated hypertrophic cell cultures was similar and represented stoichiometric apatite. Virally driven MGP overexpression in cultured chondrocytes greatly decreased mineralization. Surprisingly, MGP overexpression in the developing limb not only inhibited cartilage mineralization, but also delayed chondrocyte maturation and blocked endochondral ossification and formation of a diaphyseal intramembranous bone collar. The results show that MGP is a powerful but developmentally regulated inhibitor of cartilage mineralization, controls mineral quantity but not type, and appears to have a previously unsuspected role in regulating chondrocyte maturation and ossification processes.
Collapse
Affiliation(s)
- K Yagami
- Department of Oral Surgery, Showa University, Dental School, Ohta-Ku, Tokyo 145, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Akiyama H, Hiraki Y, Noda M, Shigeno C, Ito H, Nakamura T. Molecular cloning and biological activity of a novel Ha-Ras suppressor gene predominantly expressed in skeletal muscle, heart, brain, and bone marrow by differential display using clonal mouse EC cells, ATDC5. J Biol Chem 1999; 274:32192-7. [PMID: 10542256 DOI: 10.1074/jbc.274.45.32192] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We cloned a cDNA encoding a novel mouse protein, named A-C1, by differential display between two mouse cell lines: embryonic fibroblast C3H10T1/2 and chondrogenic ATDC5. The deduced amino acid sequence of A-C1 consists of 167 amino acids and shows 46% identity with that of a ras-responsive gene, rat Ha-rev107. Northern blot analysis showed a distinct hybridization band of 3.2 kilobases. Expression of A-C1 mRNA was detected in undifferentiated ATDC5 cells and myoblastic C2C12 cells, while none of C3H10T1/2 cells, NIH3T3 fibroblasts, Balb/c 3T3 fibroblasts, osteoblastic MC3T3-E1 cells, and ST2 bone marrow stromal cells expressed A-C1 mRNA in vitro. Moreover, A-C1 mRNA was expressed in skeletal muscle, heart, brain, and bone marrow in adult mice. By in situ hybridization, A-C1 gene expression was localized in hippocampus as well as bone marrow cells. By immunocytochemistry, A-C1 protein was detected in the cytoplasm as well as perinuclear region of the cells. Transfection of A-C1 cDNA into Ha-ras-transformed NIH3T3 cell line caused increase in the number of flat colonies and inhibition of cell growth. Our data indicate that A-C1 is expressed in some specific tissues in vivo and modulates Ha-ras-mediated signaling pathway.
Collapse
Affiliation(s)
- H Akiyama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto 606-8507, Japan.
| | | | | | | | | | | |
Collapse
|
34
|
Fukada K, Shibata S, Suzuki S, Ohya K, Kuroda T. In situ hybridisation study of type I, II, X collagens and aggrecan mRNas in the developing condylar cartilage of fetal mouse mandible. J Anat 1999; 195 ( Pt 3):321-9. [PMID: 10580848 PMCID: PMC1468002 DOI: 10.1046/j.1469-7580.1999.19530321.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to investigate the developmental characteristics of the mandibular condyle in sequential phases at the gene level using in situ hybridisation. At d 14.5 of gestation, although no expression of type II collagen mRNA was observed, aggrecan mRNA was detected with type I collagen mRNA in the posterior region of the mesenchymal cell aggregation continuous with the ossifying mandibular bone anlage prior to chondrogenesis. At d 15.0 of gestation, the first cartilaginous tissue appeared at the posterior edge of the ossifying mandibular bone anlage. The primarily formed chondrocytes in the cartilage matrix had already shown the appearance of hypertrophy and expressed types I, II and X collagens and aggrecan mRNAs simultaneously. At d 16.0 of gestation, the condylar cartilage increased in size due to accumulation of hypertrophic chondrocytes characterised by the expression of type X collagen mRNA, whereas the expression of type I collagen mRNA had been reduced in the hypertrophic chondrocytes and was confined to the periosteal osteogenic cells surrounding the cartilaginous tissue. At d 18.0 of gestation before birth, cartilage-characteristic gene expression had been reduced in the chondrocytes of the lower half of the hypertrophic cell layer. The present findings demonstrate that the initial chondrogenesis for the mandibular condyle starts continuous with the posterior edge of the mandibular periosteum and that chondroprogenitor cells for the condylar cartilage rapidly differentiate into hypertrophic chondrocytes. Further, it is indicated that sequential rapid changes and reductions of each mRNA might be closely related to the construction of the temporal mandibular ramus in the fetal stage.
Collapse
Affiliation(s)
- K Fukada
- 2nd Department of Orthodontics, School of Dentistry, Tokyo Medical and Dental University, Japan.
| | | | | | | | | |
Collapse
|
35
|
Abstract
OBJECTIVE To determine whether human osteoarthritic (OA) cartilage loses its ability to remain avascular when placed into the in-vivo model of angiogenesis, the chick embryo chorio-allantoic membrane (CAM), and to determine specific changes that occur in the cartilage matrix when the cartilage is exposed to an active vasculature. DESIGN Articular cartilage from OA and non-OA joints was grafted onto the CAM for up to 5 days before fixing and processing for histological, histochemical and immunological examination for specific changes in proteoglycan and collagen. RESULTS OA cartilage, but not non-OA cartilage, showed invasion of its matrix by blood vessels from the CAM to various extents. Associated with these blood vessels was a loss of staining for proteoglycans and cartilage specific glycosaminoglycans (GAG). A deposition of collagen types I and X was also visualized around the invasive vessels. CONCLUSIONS OA cartilage loses or has already lost its ability to remain avascular when placed onto the chick CAM. Changes occur in the matrix around the invasive blood vessels, specifically a loss of proteoglycan and GAG, and the deposition of new collagen types, notably I and X.
Collapse
Affiliation(s)
- S A Fenwick
- Glenfield Hospital NHS Trust, University of Leicester, Groby Road, Leicester, LE3 9QP
| | | | | |
Collapse
|
36
|
Ito H, Akiyama H, Shigeno C, Iyama K, Matsuoka H, Nakamura T. Hedgehog signaling molecules in bone marrow cells at the initial stage of fracture repair. Biochem Biophys Res Commun 1999; 262:443-51. [PMID: 10462495 DOI: 10.1006/bbrc.1999.1197] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ihh is a secreted protein expressed in chondrocytes in cartilaginous soft callus and thought to be involved in regulation of chondrogenic differentiation in fracture repair processes. However, gene expression and function of Ihh and its signaling molecules, Ptc and Smo, at the initial stage of fracture repair remain unknown. In the present study, we showed by RT-PCR of mouse rib fractures that the upregulation of Ihh mRNA occurred within hours after fracture, immediately followed by that of Ptc mRNA, and that both Ihh and Ptc mRNAs exhibited the time course similar to those of OP and OC mRNAs at the initial stage of fracture repair. The transcript level of Smo mRNA gradually increased within hours after fracture and was continuously maintained throughout the subsequent fracture repair processes. By in situ hybridization analysis, the transcripts of Ptc and Smo genes localized in bone marrow of unfractured ribs, and those of Ihh, Ptc, and Smo were expressed in the vicinity of the fracture site at 8 h after fracture. Furthermore, in adherent bone marrow cells in culture, mrIhh-N upregulated the gene expression of TGF-beta(1) as well as OPGL, a potent stimulator of osteoclastogenesis and osteoclast activity. These observations suggest that Ihh may play roles in the initial stage of fracture repair via TGF-beta(1) and OPGL.
Collapse
Affiliation(s)
- H Ito
- Department of Nuclear Medicine and Diagnostic Imaging, Graduate School of Medicine, Sakyo, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Akiyama H, Shigeno C, Iyama K, Ito H, Hiraki Y, Konishi J, Nakamura T. Indian hedgehog in the late-phase differentiation in mouse chondrogenic EC cells, ATDC5: upregulation of type X collagen and osteoprotegerin ligand mRNAs. Biochem Biophys Res Commun 1999; 257:814-20. [PMID: 10208865 DOI: 10.1006/bbrc.1999.0494] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endochondral bone formation includes a cascade of cellular events such as proliferation, maturation, hypertrophic conversion and calcification of chondrocytes and the cartilage replacement by bone. During these processes, hypertrophic conversion and calcification of chondrocytes (the late-phase differentiation) is a crucial process of chondrogenic differentiation. Indian hedgehog (Ihh), a secreted protein expressed in early hypertrophic chondrocytes, is thought to be involved in regulation of hypertrophic conversion via a feedback loop through the perichondrium. In the present study, we showed by Northern analysis and in situ hybridization that Smoothened (Smo), a key component in hedgehog signal transduction, was expressed in chondrocytes in both adult mice and mouse embryos at 16 days post-coitum in vivo, suggesting that Ihh directly acts on chondrocytes. We previously reported that Ihh, Patched and Smo were all expressed in differentiated ATDC5 cells. Exogenously administered mouse recombinant N-terminal protein of Ihh (mrIhh-N) upregulated the gene expression of type X collagen, a phenotypic marker of hypertrophic chondrocytes, as well as osteoprotegerin ligand (OPGL), a potent stimulator of osteoclastogenesis and osteoclast activity, while it did not modulate the expression of Ihh itself, bone morphogenetic protein (BMP)-4, BMP-6, transforming growth factor (TGF)-beta1 and TGF-beta2 in differentiated ATDC5 cells. Moreover, when added to the osteoclast cultures, mrIhh-N markedly stimulated the formation of resorption pits on dentine slices. Our data support the hypothesis that Ihh stimulated the late-phase chondrogenic differentiation in differentiated ATDC5 cells and upregulated the gene expression of OPGL in these cells.
Collapse
Affiliation(s)
- H Akiyama
- Department of Nuclear Medicine and Diagnostic Imaging, Graduate School of Medicine, Kyoto University, Kyoto, Sakyo, 606-8507, Japan.
| | | | | | | | | | | | | |
Collapse
|
38
|
Koyama E, Golden EB, Kirsch T, Adams SL, Chandraratna RA, Michaille JJ, Pacifici M. Retinoid signaling is required for chondrocyte maturation and endochondral bone formation during limb skeletogenesis. Dev Biol 1999; 208:375-91. [PMID: 10191052 DOI: 10.1006/dbio.1999.9207] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Retinoids have long been known to influence skeletogenesis but the specific roles played by these effectors and their nuclear receptors remain unclear. Thus, it is not known whether endogenous retinoids are present in developing skeletal elements, whether expression of the retinoic acid receptor (RAR) genes alpha, beta, and gamma changes during chondrocyte maturation, or how interference with retinoid signaling affects skeletogenesis. We found that immature chondrocytes present in stage 27 (Day 5.5) chick embryo humerus exhibited low and diffuse expression of RARalpha and gamma, while RARbeta expression was strong in perichondrium. Emergence of hypertrophic chondrocytes in Day 8-10 embryo limbs was accompanied by a marked and selective up-regulation of RARgamma gene expression. The RARgamma-rich type X collagen-expressing hypertrophic chondrocytes lay below metaphyseal prehypertrophic chondrocytes expressing Indian hedgehog (Ihh) and were followed by mineralizing chondrocytes undergoing endochondral ossification. Bioassays revealed that cartilaginous elements in Day 5.5, 8.5, and 10 chick embryo limbs all contained endogenous retinoids; strikingly, the perichondrial tissues surrounding the cartilages contained very large amounts of retinoids. Implantation of beads filled with retinoid antagonist Ro 41-5253 or AGN 193109 near the humeral anlagens in stage 21 (Day 3.5) or stage 27 chick embryos severely affected humerus development. In comparison to their normal counterparts, antagonist-treated humeri in Day 8.5-10 chick embryos were significantly shorter and abnormally bent; their diaphyseal chondrocytes had remained prehypertrophic Ihh-expressing cells, did not express RARgamma, and were not undergoing endochondral ossification. Interestingly, formation of an intramembranous bony collar around the diaphysis was not affected by antagonist treatment. Using chondrocyte cultures, we found that the antagonists effectively interfered with the ability of all-trans-retinoic acid to induce terminal cell maturation. The results provide clear evidence that retinoid-dependent and RAR-mediated mechanisms are required for completion of the chondrocyte maturation process and endochondral ossification in the developing limb. These mechanisms may be positively influenced by cooperative interactions between the chondrocytes and their retinoid-rich perichondrial tissues.
Collapse
Affiliation(s)
- E Koyama
- Department of Anatomy and Histology, School of Dental Medicine, Philadelphia, Pennsylvania, 19104, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Hernández-Vidal G, Jeffcott LB, Davies ME. Immunolocalization of cathepsin B in equine dyschondroplastic articular cartilage. Vet J 1998; 156:193-201. [PMID: 9883087 DOI: 10.1016/s1090-0233(98)80122-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A polyclonal antiserum raised in sheep against human cathepsin B was tested for specificity and cross-reactivity with the horse homologue by SDS-PAGE and Western blotting, prior to being used for immunolocalization of the enzyme in equine articular cartilage. In Western blots, the antiserum recognized the 30 kDa single chain and 25 kDa heavy chain of the mature enzyme in purified bovine cathepsin B, and corresponding bands at 32 and 27 kDa in equine chondrocyte and fibroblast lysates. This antiserum was then used to compare the expression and distribution of cathepsin B in normal and dyschondroplastic cartilage of young horses. In normal articular cartilage (n = 6 animals), significant amounts of enzyme were detected only in hypertrophic chondrocytes in the deep zone. The enzyme was intracellular, located in the lysosomal granules. No extracellular matrix staining was observed. Levels of cathepsin B were increased slightly above normal in the deep zone in age-matched dyschondroplastic cartilage (n = 5 animals). The most striking finding, however, was the abundance of the enzyme in chondrocyte clonal clusters associated with the lesions. Cathepsin B levels were low in chondrocytes isolated from normal cartilage (n = 6), but increased progressively during serial subculture, reaching a maximum at passage 5-6. In contrast, primary cultures of dyschondroplastic chondrocytes (n = 3) expressed abundant cathepsin B.
Collapse
|
40
|
Abstract
Genetic studies of humans and mice continue to highlight the nonredundant mechanical role of components in complexes that anchor cells to extracellular matrices. At the same time, recent data provide exciting insights into nonredundant, critical roles of transcription factors in regulating differentiation and function of matrix-producing cells.
Collapse
Affiliation(s)
- J P Gorski
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri Kansas City, MO 64110, USA.
| | | |
Collapse
|
41
|
Lammi P, Inkinen RI, von der Mark K, Puustjärvi K, Arokoski J, Hyttinen MM, Lammi MJ. Localization of type X collagen in the intervertebral disc of mature beagle dogs. Matrix Biol 1998; 17:449-53. [PMID: 9840446 DOI: 10.1016/s0945-053x(98)90104-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Type X collagen expression in intervertebral disc of young adult beagle dogs (n = 10) was studied. Type X collagen was immunostained mainly pericellularly in the central area of the vertebral endplate, but interterritorial staining there was also present. Annulus fibrosus and nucleus pulposus did not usually stain for type X collagen. However, immunostaining of nucleus pulposus for type X collagen with a simultaneous expression of collagen alpha1(X) mRNA was observed in one dog. A weak staining was observed in two other animals with a weak collagen alpha1(X) mRNA signal. In annulus fibrosus, lamellar staining was observed in two dogs. In three animals, type X collagen mRNAs were observed in the outer edge of the annulus fibrosus, but immunohistochemical staining did not always correlate with in situ hybridization signals. In conclusion, intervertebral disc type X collagen was mainly expressed in the cartilaginous endplate. In some apparently healthy animals there was type X collagen expression in the nucleus pulposus and also in the annulus fibrosus.
Collapse
Affiliation(s)
- P Lammi
- Department of Clinical Chemistry, Kuopio University Hospital, Finland
| | | | | | | | | | | | | |
Collapse
|
42
|
Sugahara K, Iyama K, Kuroda MJ, Sano K. Double intratracheal instillation of keratinocyte growth factor prevents bleomycin-induced lung fibrosis in rats. J Pathol 1998; 186:90-8. [PMID: 9875145 DOI: 10.1002/(sici)1096-9896(199809)186:1<90::aid-path137>3.0.co;2-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Alveolar re-epithelialization is necessary in the repair of damaged alveolar epithelium after lung injury. Keratinocyte growth factor (KGF) has been shown to be a potent proliferation and differentiation factor for rat alveolar type II cells. The present study examined whether KGF would prevent bleomycin-induced lung fibrosis. Adult rats were anaesthetized and recombinant human KGF (rhKGF) (150 micrograms/kg) or saline was injected intratracheally at 48 h before and 24 h after bleomycin (Bleo, 5 mg/kg) instillation. Seven and 14 days after the last administration, rat lungs were processed for lung physiology, immunohistochemistry, and in situ hybridization. Double instillation of KGF prevented the loss of body weight and reduction in total lung capacity (TLC) due to Bleo, and markedly attenuated the protein accumulation and mRNA expression of collagen types I and III and the decreased expression of surfactant protein mRNAs in the fibrotic lesions of Bleo-treated rats. KGF may play an important role in maintaining alveolar epithelium and repairing the damaged epithelium after lung injury.
Collapse
Affiliation(s)
- K Sugahara
- Department of Anaesthesiology, Kumamoto University School of Medicine, Japan
| | | | | | | |
Collapse
|
43
|
Linsenmayer TF, Long F, Nurminskaya M, Chen Q, Schmid TM. Type X collagen and other up-regulated components of the avian hypertrophic cartilage program. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 1998; 60:79-109. [PMID: 9594572 DOI: 10.1016/s0079-6603(08)60890-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elucidating the cellular and molecular processes involved in growth and remodeling of skeletal elements is important for our understanding of congenital limb deformities. These processes can be advantageously studied in the epiphyseal growth zone, the region in which all of the increase in length of a developing long bone is achieved. Here, young chondrocytes divide, mature, become hypertrophic, and ultimately are removed. During cartilage hypertrophy, a number of changes occur, including the acquisition of synthesis of new components, the most studied being type X collagen. In this review, which is based largely on our own work, we will first examine the structure and properties of the type X collagen molecule. We then will describe the supramolecular forms into which the molecule becomes assembled within tissues, and how this changes its physical properties, such as thermal stability. Certain of these studies involve a novel, immunohistochemical approach that utilizes an antitype X collagen monoclonal antibody that detects the native conformation of the molecule. We describe the developmental acquisition of the molecule, and its transcriptional regulation as deduced by in vivo footprinting, transient transfection, and gel-shift assays. We provide evidence that the molecule has unique diffusion and regulatory properties that combine to alter the hypertrophic cartilage matrix. These conclusions are derived from an in vitro system in which exogenously added type X collagen moves rapidly through the cartilage matrix and subsequently produces certain changes mimicking ones that have been shown normally to occur in vivo. These include altering the cartilage collagen fibrils and effecting changes in proteoglycans. Last, we describe the subtractive hybridization, isolation, and characterization of other genes up-regulated during cartilage hypertrophy, with specific emphasis on one of these--transglutaminase.
Collapse
Affiliation(s)
- T F Linsenmayer
- Department of Anatomy and Cellular Biology, Tufts University Medical School, Boston, Massachusetts 02111, USA
| | | | | | | | | |
Collapse
|
44
|
Frischholz S, Beier F, Girkontaite I, Wagner K, Pöschl E, Turnay J, Mayer U, von der Mark K. Characterization of human type X procollagen and its NC-1 domain expressed as recombinant proteins in HEK293 cells. J Biol Chem 1998; 273:4547-55. [PMID: 9468510 DOI: 10.1074/jbc.273.8.4547] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type X collagen is a short-chain, network-forming collagen found in hypertrophic cartilage in the growth zones of long bones, vertebrae, and ribs. To obtain information about the structure and assembly of mammalian type X collagen, we generated recombinant human type collagen X by stable expression of full-length human alpha1(X) cDNA in the human embryonal kidney cell line HEK293 and the fibrosarcoma cell line HT1080. Stable clones were obtained secreting recombinant human type X collagen (hrColX) in amounts of 50 microg/ml with alpha1(X)-chains of apparent molecular mass of 75 kDa. Pepsin digestion converted the native protein to a molecule migrating as one band at 65 kDa, while bands of 55 and 43 kDa were generated by trypsin digestion. Polyclonal antibodies prepared against purified hrColX reacted specifically with type X collagen in sections of human fetal growth cartilage. Circular dichroism spectra and trypsin/chymotrypsin digestion experiments of hrColX at increasing temperatures indicated triple helical molecules with a reduced melting temperature of 31 degrees C as a result of partial underhydroxylation. Ultrastructural analysis of hrColX by rotary shadowing demonstrated rodlike molecules with a length of 130 nm, assembling into aggregates via the globular noncollagenous (NC)-1 domains as reported for chick type X collagen. NC-1 domains generated by collagenase digestion of hrColX migrated as multimers of apparent mass of 40 kDa on SDS-polyacrylamide gel electrophoresis, even after reduction and heat denaturation, and gave rise to monomers of 18-20 kDa after treatment with trichloroacetic acid. The NC-1 domains prepared by collagenase digestion comigrated with NC-1 domains prepared as recombinant protein in HEK293 cells, both in the multimeric and monomeric form. These studies demonstrate the potential of the pCMVsis expression system to produce recombinant triple helical type X collagens in amounts sufficient for further studies on its structural and functional domains.
Collapse
Affiliation(s)
- S Frischholz
- Institute of Experimental Medicine, Friedrich Alexander University, Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hiraki Y, Inoue H, Iyama K, Kamizono A, Ochiai M, Shukunami C, Iijima S, Suzuki F, Kondo J. Identification of chondromodulin I as a novel endothelial cell growth inhibitor. Purification and its localization in the avascular zone of epiphyseal cartilage. J Biol Chem 1997; 272:32419-26. [PMID: 9405451 DOI: 10.1074/jbc.272.51.32419] [Citation(s) in RCA: 152] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cartilage is unique among tissues of mesenchymal origin in that it is resistant to vascular invasion due to an intrinsic angiogenic inhibitor. During endochondral bone formation, however, calcified cartilage formed in the center of the cartilaginous bone rudiment allows vascular invasion, which initiates the replacement of cartilage by bone. The transition of cartilage from the angioresistant to the angiogenic status thus plays a key role in bone formation. However, the molecular basis of this phenotypic transition of cartilage has been obscure. We report here purification of an endothelial cell growth inhibitor from a guanidine extract of bovine epiphyseal cartilage. The N-terminal amino acid sequence indicated that the inhibitor was identical to chondromodulin I (ChM-I), a cartilage-specific growth-modulating factor. Purified ChM-I inhibited DNA synthesis and proliferation of vascular endothelial cells as well as tube morphogenesis in vitro. Expression of ChM-I cDNA in COS7 cells indicated that mature ChM-I molecules were secreted from the cells after post-translational modifications and cleavage from the transmembrane precursor at the predicted processing signal. Recombinant ChM-I stimulated DNA synthesis and proteoglycan synthesis of cultured growth plate chondrocytes, but inhibited tube morphogenesis of endothelial cells. In situ hybridization and immunohistochemical studies indicated that ChM-I is specifically expressed in the avascular zone of cartilage in developing bone, but not present in calcifying cartilage. These results suggest a regulatory role of ChM-I in vascular invasion during endochondral bone formation.
Collapse
Affiliation(s)
- Y Hiraki
- Department of Biochemistry, Osaka University Faculty of Dentistry, Osaka 565, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Shibata S, Fukada K, Suzuki S, Yamashita Y. Immunohistochemistry of collagen types II and X, and enzyme-histochemistry of alkaline phosphatase in the developing condylar cartilage of the fetal mouse mandible. J Anat 1997; 191 ( Pt 4):561-70. [PMID: 9449075 PMCID: PMC1467723 DOI: 10.1046/j.1469-7580.1997.19140561.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We investigated the immunohistochemical localisation of types II and X collagen as well as the cytochemical localisation of alkaline phosphatase in the developing condylar cartilage of the fetal mouse mandible on d 14-16 of pregnancy. On d 14 of pregnancy, although no immunostaining for types II and X collagen was observed, alkaline phosphatase activity was detected in all cells in the anlage of the future condylar process. On d 15 of pregnancy, immunostaining for both collagen types was simultaneously detected in the primarily formed condylar cartilage. Alkaline phosphatase activity was also detected in chondrocytes at this stage. By d 16 of pregnancy, the hypertrophic cell zone rapidly increased in size. These findings strongly support a periosteal origin for the condylar cartilage of the fetal mouse mandible, and show that progenitor cells for condylar cartilage rapidly or directly differentiate into hypertrophic chondrocytes.
Collapse
Affiliation(s)
- S Shibata
- 1st Department of Oral Anatomy, School of Dentistry, Tokyo Medical and Dental University, Japan.
| | | | | | | |
Collapse
|
47
|
Shukunami C, Ishizeki K, Atsumi T, Ohta Y, Suzuki F, Hiraki Y. Cellular hypertrophy and calcification of embryonal carcinoma-derived chondrogenic cell line ATDC5 in vitro. J Bone Miner Res 1997; 12:1174-88. [PMID: 9258747 DOI: 10.1359/jbmr.1997.12.8.1174] [Citation(s) in RCA: 242] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
During the process of endochondral bone formation, proliferating chondrocytes give rise to hypertrophic cells, which then deposit a mineralized matrix to form calcified cartilage prior to replacement by bone. Previously, we reported that a clonal cell line, ATDC5, undergoes efficient chondrogenic differentiation through a cellular condensation stage. Here we report that the differentiated ATDC5 cells became hypertrophic at the center of cartilage nodules, when the cells ceased to grow. Formation of hypertrophic chondrocytes took place in association with type X collagen gene expression and a dramatic elevation of alkaline phosphate (ALPase) activity. After 5 weeks of culture, mineralization of the culture could be discerned as Alizarin red-positive spots, which spread throughout the nodules even in the absence of beta-glycerophosphate. Electron microscopy and electron probe microanalysis revealed that calcification was first initiated at matrix vesicles in the territorial matrix and that it advanced progressively along the collagen fibers in a manner similar to that which occurs in vivo. The infrared spectrum of the mineralized nodules indicated two absorption doublets around 1030 cm-1 and 600 cm-1, which are characteristic of apatitic mineral. Calcifying cultures of ATDC5 cells retained responsiveness to parathyroid hormone (PTH): PTH markedly inhibited elevation of ALPase activity and calcification in the culture in a dose-dependent manner. Thus, we demonstrated that ATDC5 cells keep track of the multistep differentiation process encompassing the stages from mesenchymal condensation to calcification in vitro. ATDC5 cells provide an excellent model to study the molecular mechanism underlying regulation of cartilage differentiation during endochondral bone formation.
Collapse
Affiliation(s)
- C Shukunami
- Department of Biochemistry, Osaka University Faculty of Dentistry, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Otsuka Y, Mizuta H, Takagi K, Iyama K, Yoshitake Y, Nishikawa K, Suzuki F, Hiraki Y. Requirement of fibroblast growth factor signaling for regeneration of epiphyseal morphology in rabbit full-thickness defects of articular cartilage. Dev Growth Differ 1997; 39:143-56. [PMID: 9108328 DOI: 10.1046/j.1440-169x.1997.t01-1-00003.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The involvement of fibroblast growth factor-2 (FGF-2) during the repair process in rabbit full-thickness defects of articular cartilage was studied. Fibroblast growth factor-2 (50 pg/h) was administered for 2 weeks in a 5 mm defect of articular cartilage, which is large enough not to repair spontaneously. The administration of FGF-2 resulted in the regeneration of the articular cartilage and the subchondral bone within 8 weeks. In these defects, undifferentiated mesenchymal cells initiated chondrogenic differentiation coupled with replacement by subchondral bone, resulting in the resurfacing of the defects with hyaline cartilage and the recovery of subchondral bone up to the original bone-articular cartilage junction. In rabbits, full-thickness defects are capable of regenerating articular cartilage as long as the defect size is limited to < or = 3 mm in diameter. In the defects, strong immunoreactivity for FGF-2 was observed in the granulation tissue filling the defects in the early stage of repair, in association with the expression of FGF-2 mRNA shown by in situ hybridization. Once the undifferentiated mesenchymal cells had differentiated into chondrocytes, both the immunoreactivity and the in situ hybridization signal declined significantly. Upon the local administration of a monoclonal antibody against FGF-2 (bFM-1, 50 ng/h), the defects were filled with fibrous tissue and no resurfacing hyaline cartilage was formed. Compared to the non-treated defects, there were marked increases in FGF-2 immunoreactivity and the overexpression of FGF-2 mRNA in the reparative tissue in the bFM-1-treated defects. This rebound phenomenon indicates that the autocrine FGF-2 signaling is critically important for the regeneration of articular cartilage.
Collapse
Affiliation(s)
- Y Otsuka
- Department of Orthopaedic Surgery, Kumamoto University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Barber RE, Kwan AP. Partial characterization of the C-terminal non-collagenous domain (NC1) of collagen type X. Biochem J 1996; 320 ( Pt 2):479-85. [PMID: 8973556 PMCID: PMC1217955 DOI: 10.1042/bj3200479] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Collagen type X is composed of three identical alpha 1(X) chains of 59 kDa, each containing a triple-helical region of 45 kDa flanked by a short N-terminal sequence and a larger non-collagenous C-terminal (NC1) domain of approx. 15 kDa. Collagen type X molecules can associate via their C-termini to form a regular hexagonal lattice in vitro, which in vivo may provide a modified extracellular matrix for the events of endochondral ossification. The NC1 domain of chick collagen type X was isolated and purified from a highly purified bacterial collagenase digest of hypertrophic chondrocyte medium proteins. The structure and aggregation properties of the NC1 domain of collagen X were investigated, independently of the triple helix. A trimer, a dimer and a monomer of the individual alpha-chain NC1 polypeptides were identified from a bacterial collagenase digest of cartilage collagens using [14C]tyrosine labelling, N-chlorosuccinimide peptide mapping and N-terminal sequencing. The trimer (50 kDa) remained intact in Laemmli sample buffer unless boiled, upon which it dissociated into the dimer (38 kDa) and the monomer (20 kDa). The dimer persisted even after prolonged periods of heating or reduction with beta-mercaptoethanol, and in preparations obtained from chondrocyte cultures treated with beta-aminoproprionitrile, indicating the presence of non-reducible, non-lysine-derived, covalent cross-links. Hexamers of the individual C-termini were observed in rotary-shadowed preparations of purified NC1 domain, reflecting the ability of collagen type X to self-assemble via its C-termini under appropriate conditions.
Collapse
Affiliation(s)
- R E Barber
- School of Biological Sciences, University of Manchester, U.K
| | | |
Collapse
|
50
|
Kitaoka M, Iyama K, Ushijima T, Mimata C, Hori H, Abe N, Yoshioka H. Differential expressions of collagen types IV, III, and I during the development of invasive trophoblasts in rats. Dev Dyn 1996; 207:319-31. [PMID: 8922531 DOI: 10.1002/(sici)1097-0177(199611)207:3<319::aid-aja9>3.0.co;2-k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We examined the differential expressions of collagen types IV, III and I in the developing feto-maternal placental tissue of pregnant rats by a combination of in situ hybridization and immunohistochemistry. At day 9.5 of gestation, polygonal invasive cytotrophoblasts from the ectoplacental cone, which was modifying the maternal central artery, revealed intensely expressed alpha 1 (IV) and alpha 1 (III) collagen mRNAs. The localization patterns of these translated products, collagen type IV and procollagen type III, were slightly different in the invasive cytotrophoblasts. Collagen type IV densely deposited intracellularly and intercellularly in the maternal central artery and in the thickened basement membranes of the cytotrophoblasts. However, expression of alpha 1 (I) collagen mRnA and procollagen type I was hardly detectable in the cytotrophoblasts. At day 13 of gestation, a high level of alpha 1 (IV) collagen mRNA was expressed in the cytotrophoblastic cell layer (trophospongium) and in the invasive large cytotrophoblasts. A moderate level of alpha 1 (III) collagen mRNA was also expressed mainly in the cytotrophoblasts, while alpha 1 (I) collagen mRNA was expressed at very low levels. Interestingly, procollagen type III failed to show linear immunoreactivity in the subepithelial extracellular matrix beneath the maternal artery with the invasive cytotrophoblasts. Additional quantitative analyses of these type IV, III, and I collagen mRNA levels in in situ hybridization experiments between several cell types also revealed significant differences individually. Electron-microscopic study detected no cross-striated collagen fibers in the thickened basement membrane-like structures adjacent to the invasive cytotrophoblasts. Fibrillar and basement membrane collagen gene expressions, their protein syntheses, and the processing of these procollagens seems to be developmentally regulated in the invasive cytotrophoblasts during the organization of feto-maternal placental tissue. The remodeling of the maternal central artery by the invasive cytotrophoblasts is important for ensuring the adequate blood supply to the developing placenta and fetus.
Collapse
Affiliation(s)
- M Kitaoka
- Division of Pathology, Kumamoto Chuo Hospital, Japan
| | | | | | | | | | | | | |
Collapse
|