1
|
Identification of candidate enhancers controlling the transcriptome during the formation of interphalangeal joints. Sci Rep 2022; 12:12835. [PMID: 35896673 PMCID: PMC9329285 DOI: 10.1038/s41598-022-16951-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
The formation of the synovial joint begins with the visible emergence of a stripe of densely packed mesenchymal cells located between distal ends of the developing skeletal anlagen called the interzone. Recently the transcriptome of the early synovial joint was reported. Knowledge about enhancers would complement these data and lead to a better understanding of the control of gene transcription at the onset of joint development. Using ChIP-sequencing we have mapped the H3-signatures H3K27ac and H3K4me1 to locate regulatory elements specific for the interzone and adjacent phalange, respectively. This one-stage atlas of candidate enhancers (CEs) was used to map the association between these respective joint tissue specific CEs and biological processes. Subsequently, integrative analysis of transcriptomic data and CEs identified new putative regulatory elements of genes expressed in interzone (e.g., GDF5, BMP2 and DACT2) and phalange (e.g., MATN1, HAPLN1 and SNAI1). We also linked such CEs to genes known as crucial in synovial joint hypermobility and osteoarthritis, as well as phalange malformations. These analyses show that the CE atlas can serve as resource for identifying, and as starting point for experimentally validating, putative disease-causing genomic regulatory regions in patients with synovial joint dysfunctions and/or phalange disorders, and enhancer-controlled synovial joint and phalange formation.
Collapse
|
2
|
Abstract
Synovial joints enable movement and protect the integrity of the articular cartilage. Joints form within skeletal condensations destined to undergo chondrogenesis. The suppression of this chondrogenic program in the interzone is the first morphological sign of joint formation. While we have a fairly good understanding of the essential roles of BMP and TGFβ family members in promoting chondrogenic differentiation in developing skeletal elements, we know very little about how BMP activity is suppressed specifically within the interzone, a crucial step in joint development. The function of the BMP ligand Gdf5 has been especially difficult to decipher. On the one hand, Gdf5 is required to promote chondrogenesis of articular elements. On the other hand, Gdf5 is highly expressed in the joint interzone where chondrogenesis must be suppressed for the formation of many joints. Here we review the evidence that BMP signaling must be suppressed within the joint interzone for joint morphogenesis to progress, and consider how Gdf5 exerts its divergent effects on chondrogenesis and joint formation. We also consider how TGFβ signaling impacts formation of the interzone. Finally, we propose a model whereby Gdf5 exerts distinct effects in the interzone vs. surrounding cartilage based on the repertoire of BMP receptors available in these tissues. Understanding how BMP antagonists and counteracting TGFβ signals intersect with Gdf5 to sculpt the joint interzone is essential for understanding the origin of osteoarthritis and other diseases of joint tissues.
Collapse
Affiliation(s)
- Karen M Lyons
- Department of Orthopaedic Surgery, Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States.
| |
Collapse
|
3
|
The Elution Kinetics of BMP-2, BMP-4, and BMP-7 From a Commercial Human Demineralized Bone Matrix Putty. J Craniofac Surg 2017; 28:2183-2188. [DOI: 10.1097/scs.0000000000004016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
4
|
Hudnall AM, Arthur JW, Lowery JW. Clinical Relevance and Mechanisms of Antagonism Between the BMP and Activin/TGF-β Signaling Pathways. J Osteopath Med 2017; 116:452-61. [PMID: 27367950 DOI: 10.7556/jaoa.2016.089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The transforming growth factor β (TGF-β) superfamily is a large group of signaling molecules that participate in embryogenesis, organogenesis, and tissue homeostasis. These molecules are present in all animal genomes. Dysfunction in the regulation or activity of this superfamily's components underlies numerous human diseases and developmental defects. There are 2 distinct arms downstream of the TGF-β superfamily ligands-the bone morphogenetic protein (BMP) and activin/TGF-β signaling pathways-and these 2 responses can oppose one another's effects, most notably in disease states. However, studies have commonly focused on a single arm of the TGF-β superfamily, and the antagonism between these pathways is unknown in most physiologic and pathologic contexts. In this review, the authors summarize the clinically relevant scenarios in which the BMP and activin/TGF-β pathways reportedly oppose one another and identify several molecular mechanisms proposed to mediate this interaction. Particular attention is paid to experimental findings that may be informative to human pathology to highlight potential therapeutic approaches for future investigation.
Collapse
|
5
|
Miyamoto Y, Kanzaki H, Wada S, Tsuruoka S, Itohiya K, Kumagai K, Hamada Y, Nakamura Y. Asporin stably expressed in the surface layer of mandibular condylar cartilage and augmented in the deeper layer with age. Bone Rep 2017; 7:41-50. [PMID: 28875156 PMCID: PMC5574816 DOI: 10.1016/j.bonr.2017.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/30/2017] [Accepted: 07/20/2017] [Indexed: 12/29/2022] Open
Abstract
Mandibular condylar cartilage (MCC) exhibits dual roles both articular cartilage and growth center. Of many growth factors, TGF-β has been implicated in the growth of articular cartilage including MCC. Recently, Asporin, decoy to TGF-β, was discovered and it blocks TGF-β signaling. Asporin is expressed in a variety of tissues including osteoarthritic articular cartilage, though there was no report of Asporin expression in MCC. In the present study, we investigated the temporal and spatial expression of Asporin in MCC. Gene expression profile of MCC and epiphyseal cartilage in tibia of 5 weeks old ICR mice were firstly compared with microarray analysis using the laser capture microdissected samples. Variance of gene expression was further confirmed by real-time RT-PCR and immunohistochemical staining at 1,3,10, and 20 weeks old. TGF-β and its signaling molecule, phosphorylated Smad-2/3 (p-Smad2/3), were also examined by immunohistochemical staining. Microarray analysis revealed that Asporin was highly expressed in MCC. Real-time RT-PCR analysis confirmed that the fibrous layer of MCC exhibited stable higher Asporin expression at any time points as compared to epiphyseal cartilage. This was also observed in immunohistochemical staining. Deeper layer in MCC augmented Asporin expression with age. Whereas, TGF-β was stably highly observed in the layer. The fibrous layer of MCC exhibited weak staining of p-Smad2/3, though the proliferating layer of MCC was strongly stained as compared to epiphyseal cartilage of tibia at early time point. Consistent with the increase of Asporin expression in the deeper layer of MCC, the intensity of p-Smad-2/3 staining was decreased with age. In conclusion, we discovered that Asporin was stably expressed at the fibrous layer of MCC, which makes it possible to manage both articular cartilage and growth center at the same time. Asporin gene and protein were highly expressed in mandibular condylar cartilage as compared to tibial epiphyseal cartilage. Asporin in mandibular condylar cartilage was augmented with age. TGF-β signaling is suppressed by augmented Asporin and decreased TGF-β production in mandibular condylar cartilage.
Collapse
Affiliation(s)
- Yutaka Miyamoto
- Department of orthodontics, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| | - Hiroyuki Kanzaki
- Department of orthodontics, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| | - Satoshi Wada
- Department of orthodontics, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| | - Sari Tsuruoka
- Department of orthodontics, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| | - Kanako Itohiya
- Department of orthodontics, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| | - Kenichi Kumagai
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| | - Yoshiki Hamada
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| | - Yoshiki Nakamura
- Department of orthodontics, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Kanagawa Pref., Japan
| |
Collapse
|
6
|
Study on the effects of gradient mechanical pressures on the proliferation, apoptosis, chondrogenesis and hypertrophy of mandibular condylar chondrocytes in vitro. Arch Oral Biol 2017; 73:186-192. [DOI: 10.1016/j.archoralbio.2016.10.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 09/03/2016] [Accepted: 10/15/2016] [Indexed: 11/23/2022]
|
7
|
Miyatake K, Iwasa K, McNary SM, Peng G, Reddi AH. Modulation of Superficial Zone Protein/Lubricin/PRG4 by Kartogenin and Transforming Growth Factor-β1 in Surface Zone Chondrocytes in Bovine Articular Cartilage. Cartilage 2016; 7:388-97. [PMID: 27688846 PMCID: PMC5029568 DOI: 10.1177/1947603516630789] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Superficial zone protein (SZP)/lubricin/PRG4 functions as a boundary lubricant in articular cartilage to decrease friction and wear. As articular cartilage lubrication is critical for normal joint function, the accumulation of SZP at the surface of cartilage is important for joint homeostasis. Recently, a heterocyclic compound called kartogenin (KGN) was found to induce chondrogenic differentiation and enhance mRNA expression of lubricin. The objective of this study was to determine whether KGN can stimulate synthesis of SZP in superficial zone, articular chondrocytes. DESIGN We investigated the effects of KGN and transforming growth factor-β1 (TGF-β1) on articular cartilage and synovium of the bovine knee joint by evaluating SZP secretion by enzyme-linked immunosorbent assay analysis. Monolayer, micromass, and explant cultures of articular cartilage, and monolayer culture of synoviocytes, were treated with KGN. SZP accumulation in the medium was evaluated and mRNA expression was measured through quantitative polymerase chain reaction. RESULTS TGF-β1 stimulated SZP secretion by superficial zone chondrocytes in monolayer, explant, and micromass cultures as expected. In addition, SZP secretion was inhibited by IL-1β in explant cultures, and enhanced by TGF-β1 in synoviocyte monolayer cultures. Although KGN elicited a 1.2-fold increase in SZP mRNA expression in combination with TGF-β1, KGN neither stimulated any significant increases in SZP synthesis nor prevented catabolic decreases in SZP production from IL-1β. CONCLUSIONS These data suggest that the chondrogenic effects of KGN depend on cellular phenotype and differentiation status, as KGN did not alter SZP synthesis in differentiated, superficial zone articular chondrocytes.
Collapse
Affiliation(s)
- Kazumasa Miyatake
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Kenjiro Iwasa
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Sean M. McNary
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Gordon Peng
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - A. Hari Reddi
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California, Davis, Sacramento, CA, USA,A. Hari Reddi, Department of Orthopaedic Surgery, School of Medicine, University of California, Davis, Research Building I, Room 2000, Sacramento, CA 95817, USA.
| |
Collapse
|
8
|
Peeters M, Detiger SEL, Karfeld-Sulzer LS, Smit TH, Yayon A, Weber FE, Helder MN. BMP-2 and BMP-2/7 Heterodimers Conjugated to a Fibrin/Hyaluronic Acid Hydrogel in a Large Animal Model of Mild Intervertebral Disc Degeneration. Biores Open Access 2015; 4:398-406. [PMID: 26543683 PMCID: PMC4623986 DOI: 10.1089/biores.2015.0025] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is etiologically associated with low back pain and is currently only treated in severe cases with spinal fusion. Regenerative medicine attempts to restore degenerated tissue by means of cells, hydrogels, and/or growth factors and can therefore be used to slow, halt, or reverse the degeneration of the IVD in a minimally invasive manner. Previously, the growth factors bone morphogenetic proteins 2 and 7 (BMP-2, -7) were shown to enhance disc regeneration, in vitro and in vivo. Since BMPs have only a short in vivo half-life, and to prevent heterotopic ossification, we evaluated the use of a slow release system for BMP-2 homodimers and BMP-2/7 heterodimers for IVD regeneration. BMP growth factors were conjugated to a fibrin/hyaluronic acid (FB/HA) hydrogel and intradiscally injected in a goat model of mild IVD degeneration to study safety and efficacy. Mild degeneration was induced in five lumbar discs of seven adult Dutch milk goats, by injections with the enzyme chondroitinase ABC. After 12 weeks, discs were treated with either FB/HA-hydrogel only or supplemented with 1 or 5 μg/mL of BMP-2 or BMP-2/7. BMPs were linked to the FB/HA hydrogels using a transglutaminase moiety, to be released through an incorporated plasmin cleavage site. After another 12 weeks, goats were sacrificed and discs were assessed using radiography, MRI T2* mapping, and biochemical and histological analyses. All animals maintained weight throughout the study and no heterotopic bone formation or other adverse effects were noted during follow-up. Radiographs showed significant disc height loss upon induction of mild degeneration. MRI T2* mapping showed strong and significant correlations with biochemistry and histology as shown before. Surprisingly, no differences could be demonstrated in any parameter between intervention groups. To our knowledge, this is the first large animal study evaluating BMPs conjugated to an FB/HA-hydrogel for the treatment of mild IVD degeneration. The conjugated BMP-2 and BMP-2/7 appeared safe, but no disc regeneration was observed. Possible explanations include too low dosages, short follow-up time, and/or insufficient release of the conjugated BMPs. These aspects should be addressed in future studies.
Collapse
Affiliation(s)
- Mirte Peeters
- Department of Orthopaedic Surgery, VU University Medical Center , Amsterdam, The Netherlands . ; Center for Translational Regenerative Medicine (CTRM), MOVE Research Institute Amsterdam , Amsterdam, The Netherlands
| | - Suzanne E L Detiger
- Department of Orthopaedic Surgery, VU University Medical Center , Amsterdam, The Netherlands . ; Center for Translational Regenerative Medicine (CTRM), MOVE Research Institute Amsterdam , Amsterdam, The Netherlands
| | | | - Theo H Smit
- Department of Orthopaedic Surgery, VU University Medical Center , Amsterdam, The Netherlands . ; Center for Translational Regenerative Medicine (CTRM), MOVE Research Institute Amsterdam , Amsterdam, The Netherlands
| | - Avner Yayon
- ProCore Biomed Ltd. , Weizman Science Park, Nes Ziona, Israel
| | - Franz E Weber
- University Hospital , Cranio-Maxillofacial and Oral Surgery/Bioengineering, Zürich, Switzerland
| | - Marco N Helder
- Department of Orthopaedic Surgery, VU University Medical Center , Amsterdam, The Netherlands . ; Center for Translational Regenerative Medicine (CTRM), MOVE Research Institute Amsterdam , Amsterdam, The Netherlands
| |
Collapse
|
9
|
Killian MS, Schmuki P. Influence of bioactive linker molecules on protein adsorption. SURF INTERFACE ANAL 2014. [DOI: 10.1002/sia.5497] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Manuela S. Killian
- Department of Materials Science and Engineering, WW4-LKO; University of Erlangen-Nuremberg; Germany
| | - Patrik Schmuki
- Department of Materials Science and Engineering, WW4-LKO; University of Erlangen-Nuremberg; Germany
- Department of Chemistry; King Abdulaziz University; Jeddah Saudi Arabia
| |
Collapse
|
10
|
Diverse effects of lead nitrate on the proliferation, differentiation, and gene expression of stem cells isolated from a dental origin. ScientificWorldJournal 2014; 2014:235941. [PMID: 24616615 PMCID: PMC3927845 DOI: 10.1155/2014/235941] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/27/2013] [Indexed: 11/30/2022] Open
Abstract
Lead (Pb2+) exposure continues to be a significant public health problem. Therefore, it is vital to have a continuous epidemiological dataset for a better understanding of Pb2+ toxicity. In the present study, we have exposed stem cells isolated from deciduous and permanent teeth, periodontal ligament, and bone marrow to five different types of Pb2+ concentrations (160, 80, 40, 20, and 10 µM) for 24 hours to identify the adverse effects of Pb2+ on the proliferation, differentiation, and gene expression on these cell lines. We found that Pb2+ treatment altered the morphology and adhesion of the cells in a dose-dependent manner. There were no significant changes in terms of cell surface phenotypes. Cells exposed to Pb2+ continued to differentiate into chondrogenesis and adipogenesis, and a severe downregulation was observed in osteogenesis. Gene expression studies revealed a constant expression of key markers associated with stemness (Oct 4, Rex 1) and DNA repair enzyme markers, but downregulation occurred with some ectoderm and endoderm markers, demonstrating an irregular and untimely differentiation trail. Our study revealed for the first time that Pb2+ exposure not only affects the phenotypic characteristics but also induces significant alteration in the differentiation and gene expression in the cells.
Collapse
|
11
|
|
12
|
Karl A, Olbrich N, Pfeifer C, Berner A, Zellner J, Kujat R, Angele P, Nerlich M, Mueller MB. Thyroid hormone-induced hypertrophy in mesenchymal stem cell chondrogenesis is mediated by bone morphogenetic protein-4. Tissue Eng Part A 2013; 20:178-88. [PMID: 23937304 DOI: 10.1089/ten.tea.2013.0023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chondrogenic differentiating mesenchymal stem cells (MSCs) express markers of hypertrophic growth plate chondrocytes. As hypertrophic cartilage undergoes ossification, this is a concern for the application of MSCs in articular cartilage tissue engineering. To identify mechanisms that elicit this phenomenon, we used an in vitro hypertrophy model of chondrifying MSCs for differential gene expression analysis and functional experiments with the focus on bone morphogenetic protein (BMP) signaling. Hypertrophy was induced in chondrogenic MSC pellet cultures by transforming growth factor β (TGFβ) and dexamethasone withdrawal and addition of triiodothyronine. Differential gene expression analysis of BMPs and their receptors was performed. Based on these results, the in vitro hypertrophy model was used to investigate the effect of recombinant BMP4 and the BMP inhibitor Noggin. The enhancement of hypertrophy could be shown clearly by an increased cell size, alkaline phosphatase activity, and collagen type X deposition. Upon induction of hypertrophy, BMP4 and the BMP receptor 1B were upregulated. Addition of BMP4 further enhanced hypertrophy in the absence, but not in the presence of TGFβ and dexamethasone. Thyroid hormone induced hypertrophy by upregulation of BMP4 and this induced enhancement of hypertrophy could be blocked by the BMP antagonist Noggin. BMP signaling is an important modulator of the late differentiation stages in MSC chondrogenesis and the thyroid hormone induces this pathway. As cartilage tissue engineering constructs will be exposed to this factor in vivo, this study provides important insight into the biology of MSC-based cartilage. Furthermore, the possibility to engineer hypertrophic cartilage may be helpful for critical bone defect repair.
Collapse
Affiliation(s)
- Alexandra Karl
- Department of Trauma Surgery, University of Regensburg Medical Center , Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Development of collagen/demineralized bone powder scaffolds and periosteum-derived cells for bone tissue engineering application. Int J Mol Sci 2013; 14:2056-71. [PMID: 23337204 PMCID: PMC3565365 DOI: 10.3390/ijms14012056] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 01/11/2013] [Accepted: 01/14/2013] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to investigate physical and biological properties of collagen (COL) and demineralized bone powder (DBP) scaffolds for bone tissue engineering. DBP was prepared and divided into three groups, based on various particle sizes: 75-125 µm, 125-250 µm, and 250-500 µm. DBP was homogeneously mixed with type I collagen and three-dimensional scaffolds were constructed, applying chemical crosslinking and lyophilization. Upon culture with human periosteum-derived cells (PD cells), osteogenic differentiation of PD cells was investigated using alkaline phosphatase (ALP) activity and calcium assay kits. The physical properties of the COL/DBP scaffolds were obviously different from COL scaffolds, irrespective of the size of DBP. In addition, PD cells cultured with COL scaffolds showed significantly higher cell adhesion and proliferation than those with COL/DBP scaffolds. In contrast, COL/DBP scaffolds exhibited greater osteoinductive potential than COL scaffolds. The PD cells with COL/DBP scaffolds possessed higher ALP activity than those with COL scaffolds. PD cells cultured with COL/DBP scaffolds with 250-500 mm particle size yielded the maximum calcium deposition. In conclusion, PD cells cultured on the scaffolds could exhibit osteoinductive potential. The composite scaffold of COL/DBP with 250-500 mm particle size could be considered a potential bone tissue engineering implant.
Collapse
|
14
|
Mifune Y, Matsumoto T, Takayama K, Ota S, Li H, Meszaros LB, Usas A, Nagamune K, Gharaibeh B, Fu FH, Huard J. The effect of platelet-rich plasma on the regenerative therapy of muscle derived stem cells for articular cartilage repair. Osteoarthritis Cartilage 2013; 21:175-85. [PMID: 23041435 DOI: 10.1016/j.joca.2012.09.018] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 09/10/2012] [Accepted: 09/24/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Platelet-rich plasma (PRP) is reported to promote collagen synthesis and cell proliferation as well as enhance cartilage repair. Our previous study revealed that the intracapsular injection of muscle derived stem cells (MDSCs) expressing bone morphogenetic protein 4 (BMP-4) combined with soluble Flt-1 (sFlt1) was effective for repairing articular cartilage (AC) after osteoarthritis (OA) induction. The current study was undertaken to investigate whether PRP could further enhance the therapeutic effect of MDSC therapy for the OA treatment. METHODS MDSCs expressing BMP-4 and sFlt1 were mixed with PRP and injected into the knees of immunodeficient rats with chemically induced OA. Histological assessments were performed 4 and 12 weeks after cell transplantation. Moreover, to elucidate the repair mechanisms, we performed in vitro assays to assess cell proliferation, adhesion, migration and mixed pellet co-culture of MDSCs and OA chondrocytes. RESULTS The addition of PRP to MDSCs expressing BMP-4 and sFlt1 significantly improved AC repair histologically at week 4 compared to MDSCs expressing BMP-4 and sFlt1 alone. Higher numbers of cells producing type II collagen and lower levels of chondrocyte apoptosis were observed by MDSCs expressing BMP-4 and sFlt1 and mixed with PRP. In the in vitro experiments, the addition of PRP promoted proliferation, adhesion and migration of the MDSCs. During chondrogenic pellet culture, PRP tended to increase the number of type II collagen producing cells and in contrast to the in vivo data, it increased cell apoptosis. CONCLUSIONS Our findings indicate that PRP can promote the therapeutic potential of MDSCs expressing BMP-4 and sFlt1 for AC repair (4 weeks post-treatment) by promoting collagen synthesis, suppressing chondrocyte apoptosis and finally by enhancing the integration of the transplanted cells in the repair process.
Collapse
Affiliation(s)
- Y Mifune
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
BACKGROUND Progress made by the scientific community in the understanding of cell receptors and metabolic pathways has led to discovery of chemical and protein agents which act as delivery vectors to specific tissues. Conjugating these agents to noble-metal nanoparticles allows for subsequent accumulation on or within targeted cells. Utilizing the unique light absorption properties of these nanoparticles then allows for photothermal heating of the particles and surrounding tissue. DISCUSSION The heat equations are solved for the case of gold nanoparticles in biological hard tissues, such as bone, for applications to two future cancer therapies: nanophotothermolysis and nanophotohyperthermia. CONCLUSIONS A survey of recent research in bone-targeting bioconjugates and simulations of nanoparticle thermal fields shows promise for these therapies in the near future.
Collapse
|
16
|
CHIANG HONGSEN, HUANG YIYOU, JIANG CHINGCHUAN. REPAIR OF ARTICULAR CARTILAGE INJURY. BIOMEDICAL ENGINEERING-APPLICATIONS BASIS COMMUNICATIONS 2012. [DOI: 10.4015/s1016237205000366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Articular cartilage defects heal poorly and lead to consequences as osteoarthritis. Clinical experience has indicated that no existing medication would substantially promote the healing process, and the cartilage defect requires surgical replacement. Allograft decays quickly for multiple reasons including the preparation process and immune reaction, and the outcome is disappointing. The extreme shortage of sparing in articular cartilage has much discouraged the use of autograft, which requires modification. The concept that constructs a chondral or osteochondral construct for the replacement of injured native tissue introduces that of tissue engineering. Limited number of cells are expanded either in vitro or in vivo, and resided temporally on a scaffold of biomaterial, which also acts as a vehicle to transfer the cells to the recipient site. Three core elements constitute this technique: the cell, a biodegradable scaffold, and an environment suitable for cells to present their proposed activity. Modern researches have kept updating those elements for a better performance of such cultivation of living tissue.
Collapse
Affiliation(s)
- HONGSEN CHIANG
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - YI-YOU HUANG
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - CHING-CHUAN JIANG
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Park J, Bauer S, Pittrof A, Killian MS, Schmuki P, von der Mark K. Synergistic control of mesenchymal stem cell differentiation by nanoscale surface geometry and immobilized growth factors on TiO2 nanotubes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2012; 8:98-107. [PMID: 22095845 DOI: 10.1002/smll.201100790] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 08/08/2011] [Indexed: 05/18/2023]
Abstract
The aim of this study is to elucidate whether combined environmental signals provided by nanoscale topography and by growth factors control cell behavior of mesenchymal stem cells (MSCs) in a synergistic or simply additive manner. Chondrogenic and osteogenic differentiation of MSCs is studied on vertically aligned TiO(2) nanotubes of size 15 and 100 nm with and without immobilized bone morphogenetic protein-2 (BMP-2). Although BMP-2 coating stimulates both chondrogenic and osteogenic differentiation of MSCs, the response strongly depends on the surface nanoscale geometry of the BMP-2-coated nanotubes. Chondrogenic differentiation is strongly supported on 100 nm BMP-2-coated nanotubes, but not on 15 nm nanotubes, which induce spreading and de-differentiation of chondrocytes. A similar response is observed with primary chondrocytes, which maintain their chondrogenic phenotype on BMP-2-coated 100 nm nanotubes, but de-differentiate on 15 nm nanotubes. In contrast, osteogenic differentiation is greatly enhanced on 15 nm but not on 100 nm BMP-2-coated nanotubes as shown previously. Furthermore, covalent immobilization of BMP-2 rescues MSCs from apoptosis occurring on uncoated 100 nm TiO(2) nanotube surfaces. Thus, combined signals provided by BMP-2 immobilized to a defined lateral nanoscale spacing geometry seem to contain environmental cues that are able to modulate a lineage-specific decision of MSC differentiation and cell survival in a synergistic manner.
Collapse
Affiliation(s)
- Jung Park
- Department of Experimental Medicine I, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Glueckstrasse 5, 91054 Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Hajare M, Delphine C, Youssef H, Andree D, Jean-Claude V, nadia BJ. Osteogenic differentiation of ES cell-derived EBs mediated by embedded BMP-2 and TGF-beta-1 in a polyelectrolyte multilayer film. ACTA ACUST UNITED AC 2011. [DOI: 10.1557/proc-0950-d10-04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ABSTRACTIn recent years, considerable effort has been devoted to the design and controlled fabrication of structured materials with functional properties. The layer by layer buildup of polyelectrolyte multilayer films (PEM films) from oppositely charged polyelectrolytes1 offers new opportunities for the preparation of functionalized biomaterial coatings. This technique allows the preparation of supramolecular nano-architectures exhibiting specific properties in terms of control of cell activation and may also play a role in the development of local drug delivery systems. Peptides, proteins or DNA, chemically bound to polyelectrolytes, adsorbed or embedded in PEM films, have been shown to retain their biological activities. Recently, tissue engineering has merged with stem cell technology with interest to develop new sources of transplantable material for injury or disease treatment. Eminently interesting, are bone and joint injuries disorders because of the low self-regenerating capacity of the matrix secreting cells. We present here for the first time that embedded BMP-2 and TGFβ1 in a multilayered polyelectrolyte film can drive embryonic stem cells to the cartilage or bone differentiation depending on supplementary co-factors. We selected a model system made from layer by layer poly-ℓ-glutamic acid (PℓGA) and poly-ℓ-lysine succinylated (PℓLs) films into which BMP-2 and TGFβ1 have been embedded. Our results demonstrate clearly that we are able to induce osteogenesis in embryonic stem cells mediated by growth factors embedded in a polyelectrolyte multilayer film.
Collapse
|
19
|
Motaung SCKM, Di Cesare PE, Hari Reddi A. Differential response of cartilage oligomeric matrix protein (COMP) to morphogens of bone morphogenetic protein/transforming growth factor-β family in the surface, middle and deep zones of articular cartilage. J Tissue Eng Regen Med 2011; 5:e87-96. [DOI: 10.1002/term.358] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 07/12/2010] [Indexed: 12/21/2022]
|
20
|
Biological implications of growth factors in bone remodeling following fracture, surgical resection and bonegrafting. Part 1: Transforming growth factors, bone morphogenetic proteins and related factors. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.ajoms.2010.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
21
|
Kim BS, Kang KS, Kang SK. Soluble factors from ASCs effectively direct control of chondrogenic fate. Cell Prolif 2010; 43:249-61. [PMID: 20546243 DOI: 10.1111/j.1365-2184.2010.00680.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Adipose tissue-derived stem cells (ASCs) have great potential for regenerative medicine. For molecular understanding of specific functional molecules present in ASCs, we analysed 756 proteins including specific chondrogenic functional factors, using high-throughput nano reverse-phase liquid chromatography-electrospray ionization-tandem mass spectrometry. MATERIALS, METHODS AND RESULTS Of these proteins, 33 were identified as chondrogenic factors or proteins including type 2 collagen, biglycan, insulin-like growth factor-binding protein and transforming growth factor-beta 1 (TGF-beta1). ASCs are a possible cell source for cartilage regeneration as they are able to secrete a number of functional cytokines including chondrogenesis-inducing molecules such as TGF-beta1 and bone morphogenetic protein 4 (BMP4). The chondrogenic phenotype of cultured ASCs was effectively induced by ASC-culture media (CM) containing BMP4 and TGF-beta1, and maintained after pre-treatment for 14 days in vitro and subcutaneous implantation in vivo. Chondrogenic differentiation efficiency of cultured ASCs and cultured mouse skin-derived progenitor cells (SPCs) depended absolutely on ASC CM-fold concentration. Cell density was also a very important factor for chondrogenic behaviour development during differentiation of ASCs and SPCs. CONCLUSION ASC CM-derived TGF-beta1-induced chondrogenic differentiation of ASCs resulted in significant reduction in chondrogenic activity after inhibition of the p38 pathway, revealing involvement of this MAPK pathway in TGF-beta1 signalling. On the other hand, TGF-beta1 signalling also led to SMAD activation that could directly increase chondrogenic activity of ASCs.
Collapse
Affiliation(s)
- B-S Kim
- Department of Veterinary Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | | | | |
Collapse
|
22
|
Minehara H, Urabe K, Naruse K, Mehlhorn AT, Uchida K, Südkamp NP, Itoman M. A new technique for seeding chondrocytes onto solvent-preserved human meniscus using the chemokinetic effect of recombinant human bone morphogenetic protein-2. Cell Tissue Bank 2010; 12:199-207. [PMID: 20556521 PMCID: PMC3135822 DOI: 10.1007/s10561-010-9185-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 05/28/2010] [Indexed: 11/27/2022]
Abstract
Many investigators are currently studying the use of decellularized tissue allografts from human cadavers as scaffolds onto which patients' cells could be seeded, or as carriers for genetically engineered cells to aid cell transplantation. However, it is difficult to seed cells onto very dense regular connective tissue which has few interstitial spaces. Here, we discuss the development of a chemotactic cell seeding technique using solvent-preserved human meniscus. A chemokinetic response to recombinant human bone morphogenetic protein-2 (rhBMP-2) was observed in a monolayer culture of primary chondrocytes derived from femoral epiphyseal cartilage of 2-day-old rats. The rhBMP-2 significantly increased their migration upto 10 ng/ml in a dose-dependent manner. When tested with solvent-preserved human meniscus as a scaffold, which has few interstitial spaces, rhBMP-2 was able to induce chondrocytes to migrate into the meniscus. After a 3-week incubation, newly-formed cartilaginous extracellular matrix was synthesized by migrated chondrocytes throughout the meniscus, down to a depth of 3 mm. These findings demonstrate that rhBMP-2 may be a natural chemokinetic factor in vivo, which induces migration of proliferative chondrocytes into the narrow interfibrous spaces. Our results suggest a potential application of rhBMP-2 for the designed distribution of chondrocytes into a scaffold to be used for tissue engineering.
Collapse
Affiliation(s)
- Hiroaki Minehara
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara, Japan
| | - Ken Urabe
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara, Japan
| | - Kouji Naruse
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara, Japan
| | - Alexander T. Mehlhorn
- Department of Orthopaedic and Trauma Surgery, Albert-Ludwig University, Freiburg, Germany
| | - Kentaroo Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara, Japan
| | - Norbert P. Südkamp
- Department of Orthopaedic and Trauma Surgery, Albert-Ludwig University, Freiburg, Germany
| | - Moritoshi Itoman
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara, Japan
| |
Collapse
|
23
|
van der Kraan PM, Blaney Davidson EN, van den Berg WB. Bone morphogenetic proteins and articular cartilage: To serve and protect or a wolf in sheep clothing's? Osteoarthritis Cartilage 2010; 18:735-41. [PMID: 20211748 DOI: 10.1016/j.joca.2010.03.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/02/2010] [Accepted: 03/01/2010] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Alterations in chondrocyte differentiation and matrix remodeling play a central role in osteoarthritis (OA). Chondrocyte differentiation and remodeling are amongst others regulated by the so-called Bone Morphogenetic Proteins (BMPs). Although BMPs are considered protective for articular cartilage these factors can also be involved in chondrocyte hypertrophy and matrix degradation. This review is focused on these opposed roles of BMPs in OA development and progression. METHODS Peer reviewed publications published prior to August 2009 were searched in the Pubmed database. Articles that were relevant for the role of endogenous BMPs in OA were selected. Since good quality reviews on the application of BMP supplementation in cartilage tissue engineering have been described this subject has not been covered in this review. RESULTS BMPs can stimulate both chondrocyte matrix synthesis and chondrocyte terminal differentiation. The latter results in elevated matrix metalloproteinase-13 (MMP-13) production. Stimulation of matrix synthesis will be protective for cartilage while elevated MMP-13 activity will drive matrix degradation. What action of BMPs is dominant in OA is not yet elucidated and their role might be different in patient subgroups. CONCLUSION BMPs can be protective for articular cartilage but can, due to their effect on chondrocyte differentiation, have harmful effects on articular cartilage and contribute to OA progression.
Collapse
Affiliation(s)
- P M van der Kraan
- Experimental Rheumatology & Advanced Therapeutics, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | |
Collapse
|
24
|
Facca S, Cortez C, Mendoza-Palomares C, Messadeq N, Dierich A, Johnston APR, Mainard D, Voegel JC, Caruso F, Benkirane-Jessel N. Active multilayered capsules for in vivo bone formation. Proc Natl Acad Sci U S A 2010; 107:3406-11. [PMID: 20160118 PMCID: PMC2840428 DOI: 10.1073/pnas.0908531107] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interest in the development of new sources of transplantable materials for the treatment of injury or disease has led to the convergence of tissue engineering with stem cell technology. Bone and joint disorders are expected to benefit from this new technology because of the low self-regenerating capacity of bone matrix secreting cells. Herein, the differentiation of stem cells to bone cells using active multilayered capsules is presented. The capsules are composed of poly-L-glutamic acid and poly-L-lysine with active growth factors embedded into the multilayered film. The bone induction from these active capsules incubated with embryonic stem cells was demonstrated in vitro. Herein, we report the unique demonstration of a multilayered capsule-based delivery system for inducing bone formation in vivo. This strategy is an alternative approach for in vivo bone formation. Strategies using simple chemistry to control complex biological processes would be particularly powerful, as they make production of therapeutic materials simpler and more easily controlled.
Collapse
Affiliation(s)
- S. Facca
- a: Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 977, Faculté de Médecine, 11 rue Humann, 67085 Strasbourg Cedex, France, b: Faculté de Chirurgie Dentaire Université de Strasbourg (UdS), 1 place de l’hôpital, 67084, Strasbourg, France
| | - C. Cortez
- Center for Nanoscience and Nanotechnology, Department of Chemical and Bimolecular Engineering, University of Melbourne, Victoria 3010, Australia
| | - C. Mendoza-Palomares
- a: Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 977, Faculté de Médecine, 11 rue Humann, 67085 Strasbourg Cedex, France, b: Faculté de Chirurgie Dentaire Université de Strasbourg (UdS), 1 place de l’hôpital, 67084, Strasbourg, France
| | - N. Messadeq
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut Clinique de la Souris (ICS), Centre National de la Recherche Scientifique (CNRS)/Institut National de la Santé et de la Recherche Médicale INSERM/UdS, Collège de France, BP 10142, Strasbourg, France
| | - A. Dierich
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut Clinique de la Souris (ICS), Centre National de la Recherche Scientifique (CNRS)/Institut National de la Santé et de la Recherche Médicale INSERM/UdS, Collège de France, BP 10142, Strasbourg, France
| | - A. P. R. Johnston
- Center for Nanoscience and Nanotechnology, Department of Chemical and Bimolecular Engineering, University of Melbourne, Victoria 3010, Australia
| | - D. Mainard
- Unité Mixte de Recherches 7561, Center National de la Recherche Scientifique-Université de Nancy, Faculté de Médecine, Vandoeuvre les Nancy, France; and
- Center Hospitalier Universtaire de Nancy, Hôpital Central (service d’orthopédie) 29 Avenue du Maréchal de Lattre de Tassigny, 54000 Nancy, France
| | - J.-C. Voegel
- a: Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 977, Faculté de Médecine, 11 rue Humann, 67085 Strasbourg Cedex, France, b: Faculté de Chirurgie Dentaire Université de Strasbourg (UdS), 1 place de l’hôpital, 67084, Strasbourg, France
| | - F. Caruso
- Center for Nanoscience and Nanotechnology, Department of Chemical and Bimolecular Engineering, University of Melbourne, Victoria 3010, Australia
| | - N. Benkirane-Jessel
- a: Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 977, Faculté de Médecine, 11 rue Humann, 67085 Strasbourg Cedex, France, b: Faculté de Chirurgie Dentaire Université de Strasbourg (UdS), 1 place de l’hôpital, 67084, Strasbourg, France
- Center for Nanoscience and Nanotechnology, Department of Chemical and Bimolecular Engineering, University of Melbourne, Victoria 3010, Australia
- Center Hospitalier Universtaire de Nancy, Hôpital Central (service d’orthopédie) 29 Avenue du Maréchal de Lattre de Tassigny, 54000 Nancy, France
| |
Collapse
|
25
|
Mohan N, Nair PD, Tabata Y. Growth factor-mediated effects on chondrogenic differentiation of mesenchymal stem cells in 3D semi-IPN poly(vinyl alcohol)-poly(caprolactone) scaffolds. J Biomed Mater Res A 2010; 94:146-59. [DOI: 10.1002/jbm.a.32680] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
26
|
Lorda-Diez CI, Montero JA, Martinez-Cue C, Garcia-Porrero JA, Hurle JM. Transforming growth factors beta coordinate cartilage and tendon differentiation in the developing limb mesenchyme. J Biol Chem 2009; 284:29988-96. [PMID: 19717568 DOI: 10.1074/jbc.m109.014811] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor beta (TGFbeta) signaling has an increasing interest in regenerative medicine as a potential tool to repair cartilages, however the chondrogenic effect of this pathway in developing systems is controversial. Here we have analyzed the function of TGFbeta signaling in the differentiation of the developing limb mesoderm in vivo and in high density micromass cultures. In these systems highest signaling activity corresponded with cells at stages preceding overt chondrocyte differentiation. Interestingly treatments with TGFbetas shifted the differentiation outcome of the cultures from chondrogenesis to fibrogenesis. This phenotypic reprogramming involved down-regulation of Sox9 and Aggrecan and up-regulation of Scleraxis, and Tenomodulin through the Smad pathway. We further show that TGFbeta signaling up-regulates Sox9 in the in vivo experimental model system in which TGFbeta treatments induce ectopic chondrogenesis. Looking for clues explaining the dual role of TGFbeta signaling, we found that TGFbetas appear to be direct inducers of the chondrogenic gene Sox9, but the existence of transcriptional repressors of TGFbeta signaling modulates this role. We identified TGF-interacting factor Tgif1 and SKI-like oncogene SnoN as potential candidates for this inhibitory function. Tgif1 gene regulation by TGFbeta signaling correlated with the differential chondrogenic and fibrogenic effects of this pathway, and its expression pattern in the limb marks the developing tendons. In functional experiments we found that Tgif1 reproduces the profibrogenic effect of TGFbeta treatments.
Collapse
Affiliation(s)
- Carlos I Lorda-Diez
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria, Santander 39011, Spain
| | | | | | | | | |
Collapse
|
27
|
Chiang H, Jiang CC. Repair of articular cartilage defects: review and perspectives. J Formos Med Assoc 2009; 108:87-101. [PMID: 19251544 DOI: 10.1016/s0929-6646(09)60039-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Articular cartilage defects heal poorly and lead to catastrophic degenerative arthritis. Clinical experience has indicated that no existing medication substantially promotes the healing process and the cartilage defect requires surgical replacement, preferably with an autograft. However, there is a shortage of articular cartilage that can be donated for autografting. A review of previous unsuccessful experiences reveals the reason for the current strategy to graft cartilage defects with regenerated cartilage. Autologous cartilage regeneration is a cell-based therapy in which autogenous chondrocytes or other chondrogenic cells are cultured to constitute cartilaginous tissue according to the principles of tissue engineering. Current studies are concentrating on improving such techniques from the three elements of tissue engineering, namely the cells, biomaterial scaffolds, and culture conditions. Some models of articular cartilage regeneration have yielded good repair of cartilage defects, in animal models and clinical settings, but the overall results suggest that there is room for improvement of this technique before its routine clinical application. Autologous cartilage regeneration remains the mainstay for repairing articular cartilage defects but more studies are required to optimize the efficacy of regeneration. A more abundant supply of more stable cells, i.e. capable of maintaining the phenotype of chondrogenesis, has to be identified. Porous scaffolds of biocompatible, biodegradable materials that maintain and support the presentation of the chondrogenic cells need to be fabricated. If the cells are not implanted early to allow their in vivo constitution of cartilage, a suitable in vitro cultivation method has to be devised for a consistent yield of regenerative cartilage.
Collapse
Affiliation(s)
- Hongsen Chiang
- Department of Orthopedic Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | | |
Collapse
|
28
|
|
29
|
Sohier J, Moroni L, van Blitterswijk C, de Groot K, Bezemer JM. Critical factors in the design of growth factor releasing scaffolds for cartilage tissue engineering. Expert Opin Drug Deliv 2008; 5:543-66. [PMID: 18491981 DOI: 10.1517/17425247.5.5.543] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Trauma or degenerative diseases of the joints are common clinical problems resulting in high morbidity. Although various orthopedic treatments have been developed and evaluated, the low repair capacities of articular cartilage renders functional results unsatisfactory in the long term. Over the last decade, a different approach (tissue engineering) has emerged that aims not only to repair impaired cartilage, but also to fully regenerate it, by combining cells, biomaterials mimicking extracellular matrix (scaffolds) and regulatory signals. The latter is of high importance as growth factors have the potency to induce, support or enhance the growth and differentiation of various cell types towards the chondrogenic lineage. Therefore, the controlled release of different growth factors from scaffolds appears to have great potential to orchestrate tissue repair effectively. OBJECTIVE This review aims to highlight considerations and limitations of the design, materials and processing methods available to create scaffolds, in relation to the suitability to incorporate and release growth factors in a safe and defined manner. Furthermore, the current state of the art of signalling molecules release from scaffolds and the impact on cartilage regeneration in vitro and in vivo is reported and critically discussed. METHODS The strict aspects of biomaterials, scaffolds and growth factor release from scaffolds for cartilage tissue engineering applications are considered. CONCLUSION Engineering defined scaffolds that deliver growth factors in a controlled way is a task seldom attained. If growth factor delivery appears to be beneficial overall, the optimal delivery conditions for cartilage reconstruction should be more thoroughly investigated.
Collapse
Affiliation(s)
- J Sohier
- Laboratory for osteo-articular and dental tissue engineering (LIOAD), Faculté de chirurgie dentaire de Nantes, Inserm U791, 1 Place Alexis Ricordeau, 44042 Nantes Cedex 1, France.
| | | | | | | | | |
Collapse
|
30
|
Knothe Tate ML, Falls TD, McBride SH, Atit R, Knothe UR. Mechanical modulation of osteochondroprogenitor cell fate. Int J Biochem Cell Biol 2008; 40:2720-38. [PMID: 18620888 DOI: 10.1016/j.biocel.2008.05.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 05/01/2008] [Accepted: 05/04/2008] [Indexed: 12/27/2022]
Abstract
Mesenchymal cells are natural tissue builders. They exhibit an extraordinary capacity to metamorphize into differentiated cells, using extrinsic spatial and temporal inputs and intrinsic algorithms, as well as to build and adapt their own habitat. In addition to providing a habitat for osteoprogenitor cells, tissues of the skeletal system provide mechanical support and protection for the multiple organs of vertebrate organisms. This review examines the role of mechanics on determination of cell fate during pre-, peri- and postnatal development of the skeleton as well as during tissue genesis and repair in postnatal life. The role of cell mechanics is examined and brought into context of intrinsic cues during mesenchymal condensation. Remarkable new insights regarding structure function relationships in mesenchymal stem cells, and their influence on determination of cell fate are integrated in the context of de novo tissue generation and postnatal repair. Key differences in the formation of osteogenic and chondrogenic condensations are discussed in relation to direct intramembranous and indirect endochondral ossification. New approaches are discussed to elucidate and exploit extrinsic cues to generate tissues in the laboratory and in the clinic.
Collapse
Affiliation(s)
- Melissa L Knothe Tate
- Department of Biomedical Engineering, Wickenden 307, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | | | | | | | | |
Collapse
|
31
|
Histochemical and molecular overview of the thymus as site for T-cells development. ACTA ACUST UNITED AC 2008; 43:73-120. [PMID: 18555891 DOI: 10.1016/j.proghi.2008.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 03/11/2008] [Indexed: 12/19/2022]
Abstract
The thymus represents the primary site for T cell lymphopoiesis, providing a coordinated set for critical factors to induce and support lineage commitment, differentiation and survival of thymus-seeding cells. One irrefutable fact is that the presence of non-lymphoid cells through the thymic parenchyma serves to provide coordinated migration and differentiation of T lymphocytes. Moreover, the link between foetal development and normal anatomy has been stressed in this review. Regarding thymic embryology, its epithelium is derived from the embryonic endodermal layer, with possible contributions from the ectoderm. A series of differentiating steps is essential, each of which must be completed in order to provide the optimum environment for thymic development and function. The second part of this article is focused on thymic T-cell development and differentiation, which is a stepwise process, mediated by a variety of stromal cells in different regions of the organ. It depends strongly on the thymic microenvironment, a cellular network formed by epithelial cells, macrophages, dendritic cells and fibroblasts, that provide the combination of cellular interactions, cytokines and chemokines to induce thymocyte precursors for the generation of functional T cells. The mediators of this process are not well defined but it has been demonstrated that some interactions are under neuroendocrine control. Moreover, some studies pointed out that reciprocal signals from developing T cells also are essential for establishment and maintenance of the thymic microenvironment. Finally, we have also highlighted the heterogeneity of the lymphoid, non-lymphoid components and the multi-phasic steps of thymic differentiation. In conclusion, this review contributes to an understanding of the complex mechanisms in which the foetal and postnatal thymus is involved. This could be a prerequisite for developing new therapies specifically aimed to overcome immunological defects, linked or not-linked to aging.
Collapse
|
32
|
Zilberman Y, Kallai I, Gafni Y, Pelled G, Kossodo S, Yared W, Gazit D. Fluorescence molecular tomography enables in vivo visualization and quantification of nonunion fracture repair induced by genetically engineered mesenchymal stem cells. J Orthop Res 2008; 26:522-30. [PMID: 17985393 DOI: 10.1002/jor.20518] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Fluorescence molecular tomography (FMT) is a novel tomographic near-infrared (NIR) imaging modality that enables 3D quantitative determination of fluorochrome distribution in tissues of live small animals at any depth. This study demonstrates a noninvasive, quantitative method of monitoring engineered bone remodeling via FMT. Murine mesenchymal stem cells overexpressing the osteogenic gene BMP2 (mMSCs-BMP2) were implanted into the thigh muscle and into a radial nonunion bone defect model in C3H/HeN mice. Real-time imaging of bone formation was performed following systemic administration of the fluorescent bisphosphonate imaging agent OsteoSense, an hydroxyapatite-directed bone-imaging probe. The mice underwent imaging on days 7, 14, and 21 postimplantation. New bone formation at the implantation sites was quantified using micro-computed tomography (micro-CT) imaging. A higher fluorescent signal occurred at the site of the mMSC-BMP2 implants than that found in controls. Micro-CT imaging revealed a mass of mature bone formed in the implantation sites on day 21, a finding also confirmed by histology. These findings highlight the effectiveness of FMT as a functional platform for molecular imaging in the field of bone regeneration and tissue engineering.
Collapse
Affiliation(s)
- Yoram Zilberman
- Skeletal Biotechnology Laboratory, Hebrew University, Hadassah Medical Campus, P.O. Box 12272, Ein Kerem, Jerusalem 91120, Israel
| | | | | | | | | | | | | |
Collapse
|
33
|
Anderson EJ, Knothe Tate ML. Design of Tissue Engineering Scaffolds as Delivery Devices for Mechanical and Mechanically Modulated Signals. ACTA ACUST UNITED AC 2007; 13:2525-38. [PMID: 17822359 DOI: 10.1089/ten.2006.0443] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
New approaches to tissue engineering aim to exploit endogenous strategies such as those occurring in prenatal development and recapitulated during postnatal healing. Defining tissue template specifications to mimic the environment of the condensed mesenchyme during development allows for exploitation of tissue scaffolds as delivery devices for extrinsic cues, including biochemical and mechanical signals, to drive the fate of mesenchymal stem cells seeded within. Although a variety of biochemical signals that modulate stem cell fate have been identified, the mechanical signals conducive to guiding pluripotent cells toward specific lineages are less well characterized. Furthermore, not only is spatial and temporal control of mechanical stimuli to cells challenging, but also tissue template geometries vary with time due to tissue ingrowth and/or scaffold degradation. Hence, a case study was carried out to analyze flow regimes in a testbed scaffold as a first step toward optimizing scaffold architecture. A pressure gradient was applied to produce local (nm-micron) flow fields conducive to migration, adhesion, proliferation, and differentiation of cells seeded within, as well as global flow parameters (micron-mm), including flow velocity and permeability, to enhance directed cell infiltration and augment mass transport. Iterative occlusion of flow channel dimensions was carried out to predict virtually the effect of temporal geometric variation (e.g., due to tissue development and growth) on delivery of local and global mechanical signals. Thereafter, insights from the case study were generalized to present an optimization scheme for future development of scaffolds to be implemented in vitro or in vivo. Although it is likely that manufacture and testing will be required to finalize design specifications, it is expected that the use of the rational design optimization will reduce the number of iterations required to determine final prototype geometries and flow conditions. As the range of mechanical signals conducive to guiding cell fate in situ is further elucidated, these refined design criteria can be integrated into the general optimization rubric, providing a technological platform to exploit nature's endogenous tissue engineering strategies for targeted tissue generation in the lab or the clinic.
Collapse
Affiliation(s)
- Eric J Anderson
- Department of Mechanical and Aerospace Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | | |
Collapse
|
34
|
Zuscik MJ, Ma L, Buckley T, Puzas JE, Drissi H, Schwarz EM, O’Keefe RJ. Lead induces chondrogenesis and alters transforming growth factor-beta and bone morphogenetic protein signaling in mesenchymal cell populations. ENVIRONMENTAL HEALTH PERSPECTIVES 2007; 115:1276-82. [PMID: 17805416 PMCID: PMC1964910 DOI: 10.1289/ehp.10028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Accepted: 07/02/2007] [Indexed: 05/17/2023]
Abstract
BACKGROUND It has been established that skeletal growth is stunted in lead-exposed children. Because chondrogenesis is a seminal step during skeletal development, elucidating the impact of Pb on this process is the first step toward understanding the mechanism of Pb toxicity in the skeleton. OBJECTIVES The aim of this study was to test the hypothesis that Pb alters chondrogenic commitment of mesenchymal cells and to assess the effects of Pb on various signaling pathways. METHODS We assessed the influence of Pb on chondrogenesis in murine limb bud mesenchymal cells (MSCs) using nodule formation assays and gene analyses. The effects of Pb on transforming growth factor-beta (TGF-beta) and bone morphogenetic protein (BMP) signaling was studied using luciferase-based reporters and Western analyses, and luciferase-based assays were used to study cyclic adenosine monophosphate response element binding protein (CREB), beta-catenin, AP-1, and nuclear factor-kappa B (NF-kappaB) signaling. We also used an ectopic bone formation assay to determine how Pb affects chondrogenesis in vivo. RESULTS Pb-exposed MSCs showed enhanced basal and TGF-beta/BMP induction of chondrogenesis, evidenced by enhanced nodule formation and up-regulation of Sox-9, type 2 collagen, and aggrecan, all key markers of chondrogenesis. We observed enhanced chondrogenesis during ectopic bone formation in mice preexposed to Pb via drinking water. In MSCs, Pb enhanced TGF-beta but inhibited BMP-2 signaling, as measured by luciferase reporter assays and Western analyses of Smad phosphorylation. Although Pb had no effect on basal CREB or Wnt/beta-catenin pathway activity, it induced NFkappaB signaling and inhibited AP-1 signaling. CONCLUSIONS The in vitro and in vivo induction of chondrogenesis by Pb likely involves modulation and integration of multiple signaling pathways including TGF-beta, BMP, AP-1, and NFkappaB.
Collapse
Affiliation(s)
| | | | - Taylor Buckley
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - J. Edward Puzas
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Hicham Drissi
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Regis J. O’Keefe
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
35
|
Ding HF, Liu R, Li BG, Lou JR, Dai KR, Tang TT. Biologic effect and immunoisolating behavior of BMP-2 gene-transfected bone marrow-derived mesenchymal stem cells in APA microcapsules. Biochem Biophys Res Commun 2007; 362:923-7. [PMID: 17767917 DOI: 10.1016/j.bbrc.2007.08.094] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 08/15/2007] [Indexed: 01/14/2023]
Abstract
We investigated the encapsulation of BMP-2 gene-modified mesenchymal stem cells (MSCs) in alginate-poly-L-lysine (APA) microcapsules for the persistent delivery of bone morphogenic protein-2 (BMP-2) to induce bone formation. An electrostatic droplet generator was employed to produce APA microcapsules containing encapsulated beta-gal or BMP-2 gene-transfected bone marrow-derived MSCs. We found that X-gal staining was still positive 28 days after encapsulation. Encapsulated BMP-2 gene-transfected cells were capable of constitutive delivery of BMP-2 proteins for at least 30 days. The encapsulated BMP-2 gene-transfected MSCs or the encapsulated non-gene transfer MSCs (control group) were cocultured with the undifferentiated MSCs. The gene products from the encapsulated BMP-2 cells could induce the undifferentiated MSCs to become osteoblasts that had higher alkaline phosphatase (ALP) activity than those in the control group (p<0.05). The APA microcapsules could inhibit the permeation of fluorescein isothiocyanate-conjuncted immunoglobulin G. Mixed lymphocyte reaction also indicates that the APA microcapsules could prevent the encapsulated BMP-2 gene-transfected MSCs from initiating the cellular immune response. These results demonstrated that the nonautologous BMP-2 gene-transfected stem cells are of potential utility for enhancement of bone repair and bone regeneration in vivo.
Collapse
Affiliation(s)
- H F Ding
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, PR China
| | | | | | | | | | | |
Collapse
|
36
|
Toom A, Arend A, Gunnarsson D, Ulfsparre R, Suutre S, Haviko T, Selstam G. Bone formation zones in heterotopic ossifications: histologic findings and increased expression of bone morphogenetic protein 2 and transforming growth factors beta2 and beta3. Calcif Tissue Int 2007; 80:259-67. [PMID: 17401695 DOI: 10.1007/s00223-007-9000-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2006] [Accepted: 02/01/2007] [Indexed: 11/27/2022]
Abstract
Heterotopic ossifications (HOs) formed after total endoprosthetic replacement of the hip joint were collected during revision surgery (n = 7). Tissues collected during regular hip arthroplasty (n = 12) were used as reference. Histomorphometric analysis was performed for assessment of bone formation activity in HOs and reference bone. HOs were dissected with histological guidance into three zones: formed bone, zone of active bone formation, and zone with fibrous connective and fibrocartilagineous tissue. Relative expression of the mRNA for bone morphogenetic protein 2 (BMP-2), transforming growth factor beta2 (TGF-beta2), and TGF-beta3 was determined by reverse-transcription polymerase chain reaction relative to beta-actin. Expression of all three growth factors was higher than in orthotopic bone. Similarly, the osteoid surface density was increased in HOs. The levels of all growth factors were higher in the zone of active bone formation or remodeling than in the zone of formed bone. In matured HOs, the osteoid surface density as well as mRNA levels were lower, although still significantly raised, indicating that bone formation slows down after 2 years. Immunohistochemical analysis demonstrated the presence of TGF-beta1, TGF-beta2, TGF-beta3, and BMP-2 proteins in the zone of bone formation. We conclude that bone formation after heterotopic bone induction is initially intense, slows down within 2 years, and thereupon continues as active remodeling mainly on the border of HO. Our data indicate that BMP-2, TGF-beta2, and TGF-beta3 are involved in bone formation in HO.
Collapse
Affiliation(s)
- A Toom
- Clinic of Traumatology and Orthopedics, University of Tartu, Puusepa 8, Tartu, 51014, Estonia.
| | | | | | | | | | | | | |
Collapse
|
37
|
Nakanishi K, Chan YS, Ito K. Notch signaling is required for the chondrogenic specification of mouse mesencephalic neural crest cells. Mech Dev 2007; 124:190-203. [PMID: 17241776 DOI: 10.1016/j.mod.2006.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 12/13/2006] [Accepted: 12/14/2006] [Indexed: 11/25/2022]
Abstract
We examined the roles of Notch signaling in the chondrogenesis of mouse mesencephalic neural crest cells. The present study demonstrated that the activation of Notch signaling or the treatment with fibroblast growth factors (FGFs) promotes the differentiation of proliferative and prehypertrophic chondrocytes expressing collagen type II. Notch activation or FGF2 exposure during the first 24h in culture was critical for the differentiation of proliferative and prehypertrophic chondrocytes. The expression of SOX9, a transcription activator of collagen type II, was also upregulated by Notch activation or FGF2 treatment. The promotion of proliferative and prehypertrophic chondrocyte differentiation by FGF2 was significantly suppressed by the inhibition of Notch signaling using Notch-1 siRNA. These results suggest that FGFs activate Notch signaling and that this activation promotes the chondrogenic specification of mouse mesencephalic neural crest cells. Furthermore, we investigated the expression patterns of Notch-1, SOX9, and p75, which is a marker of undifferentiated neural crest cells, in the mandibular arch where mesencephalic neural crest cells colonize and undergo chondrogenesis. These in vivo observations, coupled with the results of the present in vitro study, suggest that Notch signaling as well as FGFs is a component of epithelial-mesenchymal interactions that promote the chondrogenic specification of mouse mesencephalic neural crest cells.
Collapse
Affiliation(s)
- Kouichi Nakanishi
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | | | | |
Collapse
|
38
|
Song JJ, Aswad R, Kanaan RA, Rico MC, Owen TA, Barbe MF, Safadi FF, Popoff SN. Connective tissue growth factor (CTGF) acts as a downstream mediator of TGF-beta1 to induce mesenchymal cell condensation. J Cell Physiol 2007; 210:398-410. [PMID: 17111364 DOI: 10.1002/jcp.20850] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal cell (MC) condensation or the aggregation of MCs precedes chondrocyte differentiation and is required for subsequent cartilage formation during endochondral ossification. In this study, we used micromass cultures of C3H10T1/2 cells as an in vitro model system for studying MC condensation and the events important for this process. Transforming growth factor beta1 (TGF-beta1) served as the initiator of MC condensation in our model system and we were interested in determining whether CTGF functions as a downstream mediator of TGF-beta1. CTGF is a matricellular protein that has been found to be expressed in MC condensations and in the perichondrium. Micromass cultures of C3H10T1/2 cells condensed under TGF-beta1 stimulation concomitant with dramatic up-regulation of CTGF mRNA and protein levels. CTGF silencing by either CTGF siRNA or CTGF antisense oligonucleotide approaches showed that TGF-beta1-induced condensation was CTGF dependent. Furthermore, silencing of CTGF expression resulted in significant reductions in cell proliferation and migration, events that are crucial during MC condensation. In addition, up-regulation of Fibronectin (FN) and suppression of Sox9 expression by TGF-beta1 was also found to be mediated by CTGF. Immunofluorescence of developing mouse vertebrae showed that CTGF, TGF-beta1 and FN were co-expressed in condensations of MCs, while Sox9 expression was low at this stage. During subsequent chondrogenesis, Sox9 expression was high in chondrocytes while CTGF expression was limited to the perichondrium. Thus, CTGF is an essential downstream mediator of TGF-beta1-induced MC condensation through its effects on cell proliferation and migration. CTGF is also involved in up-regulating FN and suppressing Sox9 expression during TGF-beta1 induced MC condensation.
Collapse
Affiliation(s)
- Jason J Song
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Xu D, Gechtman Z, Hughes A, Collins A, Dodds R, Cui X, Jolliffe L, Higgins L, Murphy A, Farrell F. Potential involvement of BMP receptor type IB activation in a synergistic effect of chondrogenic promotion between rhTGFbeta3 and rhGDF5 or rhBMP7 in human mesenchymal stem cells. Growth Factors 2006; 24:268-78. [PMID: 17381068 DOI: 10.1080/08977190601075865] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Chondrogenic promotion by rhGDF5 with or without rhTGFbeta3 was studied in pellet culture of human mesenchymal stem cells (HMSCs). A synergy between rhGDF5 and rhTGFbeta3 was observed in promoting chondrogenesis. rhBMP2, rhBMP6, rhBMP7 and rhTGFbeta1 were further tested and showed the same effect. To explore the mechanism, the expression of TGFbetatype I and II receptors, ALK5, ALK2, ALK3, ALK6, TGFbetaRII, BMPRII, ActRII was studied. ALK6 showed increase by the rhTGFbeta1 or rhTGFbeta3 treatment. ALK6 protein expression also showed increase by rhTGFbeta3. rhTGFbeta1/rhTGFbeta3 induced ALK6 up-regulation was inhibited by SD-208, a TGFbeta type I receptor inhibitor. Chondrogenesis by rhTGFbetal/rhTGFbeta3 or the combination between rhTGFbetal/rhTGFbeta3 and rhGDF5 also was diminished by SD-208. SMAD1/5/8 phosphorylation in nascent human mesenchymal stem cells (HMSCs) was stimulated weakly by rhGDF5 but strongly by rhBMP7. The rhGDF5 stimulated SMAD1/5/8 phosphorylation was enhanced by rhTGFbetal/rhTGFbeta3 but inhibited by SD-208. The rhBMP7 stimulated SMAD1/5/8 phosphorylation did not show influence by rhTGFbeta3 and SD-208. Our results indicated the potential involvement of ALK6 activation by rhTGFbetas in the synergy between rhTGFbetas and rhBMPs.
Collapse
Affiliation(s)
- Danlin Xu
- Growth Factor Team, Drug Discovery, Johnson & Johnson Pharmaceutical Research and Development LLC, 1000 Route 202 South, Raritan, NJ 08869, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Schmidl M, Adam N, Surmann-Schmitt C, Hattori T, Stock M, Dietz U, de Crombrugghe B, Po¨schl E, von der Mark K. Twisted Gastrulation Modulates Bone Morphogenetic Protein-induced Collagen II and X Expression in Chondrocytes in Vitro and in Vivo. J Biol Chem 2006. [DOI: 10.1016/s0021-9258(19)84094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
41
|
Schmidl M, Adam N, Surmann-Schmitt C, Hattori T, Stock M, Dietz U, de Crombrugghe B, Pöschl E, von der Mark K. Twisted Gastrulation Modulates Bone Morphogenetic Protein-induced Collagen II and X Expression in Chondrocytesin Vitroandin Vivo. J Biol Chem 2006; 281:31790-800. [PMID: 16905550 DOI: 10.1074/jbc.m603419200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Twisted gastrulation (TSG) is an extracellular modulator of bone morphogenetic protein (BMP) activity and regulates dorsoventral axis formation in early Drosophila and Xenopus development. Studies on tsg-deficient mice also indicated a role of this protein in skeletal growth, but the mechanism of TSG activity in this process has not yet been investigated. Here we show for the first time by in situ hybridization and immunohistochemistry that TSG is strongly expressed in bovine and mouse growth plate cartilage as well as in fetal ribs, vertebral cartilage, and cartilage anlagen of the skull. Furthermore we provide evidence that TSG is directly involved in BMP-regulated chondrocyte differentiation and maturation. In vitro, TSG impaired the dose-dependent BMP-2 stimulation of collagen II and X expression in cultures of MC615 chondrocytes and primary mouse chondrocytes. In the presence of chordin, a BMP antagonist, the inhibitory effect of TSG was further enhanced. TSG also inhibited BMP-2-stimulated phosphorylation of Smad factors in chondrocytes, confirming the role of TSG as a modulator of BMP signaling. For analysis of TSG functions in cartilage development in vivo, the gene was overexpressed in transgenic mice under the control of the cartilage-specific Col2a1 promoter. As a result, Col10a1 expression was significantly reduced in the growth plates of transgenic embryos and newborns in comparison with wild type littermates as shown by in situ hybridization and by real time PCR analysis. The data suggest that TSG is an important modulator of BMP-regulated cartilage development and chondrocyte differentiation.
Collapse
Affiliation(s)
- Martina Schmidl
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Aslan H, Zilberman Y, Arbeli V, Sheyn D, Matan Y, Liebergall M, Li JZ, Helm GA, Gazit D, Gazit Z. Nucleofection-BasedEx VivoNonviral Gene Delivery to Human Stem Cells as a Platform for Tissue Regeneration. ACTA ACUST UNITED AC 2006; 12:877-89. [PMID: 16674300 DOI: 10.1089/ten.2006.12.877] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
There are several gene therapy approaches to tissue regeneration. Although usually efficient, virusbased approaches may elicit an immune response against the viral proteins. An alternative approach, nonviral transfer, is safer, and can be controlled and reproduced. We hypothesized that in vivo bone formation could be achieved using human mesenchymal stem cells (hMSCs) nonvirally transfected with the human bone morphogenetic protein-2 (hBMP-2) or -9 (hBMP-9) gene. Human MSCs were transfected using nucleofection, a unique electropermeabilization-based technique. Postnucleofection, cell viability was 53.6 +/- 2.5% and gene delivery efficiency was 51% to 88% (mean 68.2 +/- 4.1%), as demonstrated by flow cytometry in enhanced green fluorescent protein (EGFP)-nucleofected hMSCs. Transgene expression lasted longer than 14 days and was very low 21 days postnucleofection. Both hBMP-2- and hBMP-9-nucleofected hMSCs in culture demonstrated a significant increase in calcium deposition compared with EGFP-nucleofected hMSCs. Human BMP-2- and hBMP-9-nucleofected hMSCs transplanted in ectopic sites in NOD/SCID mice induced bone formation 4 weeks postinjection. We conclude that in vivo bone formation can be achieved by using nonvirally nucleofected hMSCs. This could lead to a breakthrough in the field of regenerative medicine, in which safer, nonviral therapeutic strategies present a very attractive alternative.
Collapse
Affiliation(s)
- Hadi Aslan
- Skeletal Biotechnology Laboratory, Hebrew University, Hadassah Medical Center, Ein Kerem, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
In the early 1970s, the isolation and identification of bone morphogenetic proteins (BMPs) was a major breakthrough for the understanding of the distinct biological events occurring during bone formation. Not surprising, since their discovery, BMPs have been perhaps one of the most intensively studied group of factors in various physiological processes. The prompt development of recombinant BMPs and various delivery methods made BMPs currently available for clinical use. Research and clinical studies on BMPs are ongoing, aiming to refine further our understanding of their activities in vivo and to optimise and expand their clinical use in humans.
Collapse
Affiliation(s)
- Rozalia Dimitriou
- Academic Department of Trauma and Orthopaedic Surgery, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | | |
Collapse
|
44
|
|
45
|
Heng BC, Cao T, Lee EH. Directing stem cell differentiation into the chondrogenic lineage in vitro. Stem Cells 2005; 22:1152-67. [PMID: 15579636 DOI: 10.1634/stemcells.2004-0062] [Citation(s) in RCA: 171] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A major area in regenerative medicine is the application of stem cells in cartilage tissue engineering and reconstructive surgery. This requires well-defined and efficient protocols for directing the differentiation of stem cells into the chondrogenic 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 upon 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 chondrogenesis and cartilaginous tissue biology. The development of pharmacokinetic and cytotoxicity/genotoxicity screening tests for cartilage-related biomaterials and drugs could also utilize protocols developed for the chondrogenic differentiation of stem cells. Hence, this review critically examines the various strategies that could be used to direct the differentiation of stem cells into the chondrogenic lineage in vitro.
Collapse
Affiliation(s)
- Boon Chin Heng
- Faculty of Dentistry, National University of Singapore, Singapore
| | | | | |
Collapse
|
46
|
Li J, Yoon ST, Hutton WC. Effect of bone morphogenetic protein-2 (BMP-2) on matrix production, other BMPs, and BMP receptors in rat intervertebral disc cells. ACTA ACUST UNITED AC 2005; 17:423-8. [PMID: 15385883 DOI: 10.1097/01.bsd.0000112084.85112.5d] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE An in vitro experiment study using rat disc cells was carried out to determine the effect of bone morphogenetic protein-2 (BMP-2) on extracellular matrix production, other BMPs, and BMP receptors (BMPRs). METHODS Cells from the anulus fibrosus and transition zone were harvested and cultured. When the cells reached 80% confluence, BMP-2 was added to reach a final concentration of 200 ng/mL. Three days later, the culture media were collected for the assay of sulfated glycosaminoglycans (sGAG) and collagen types I and II. The cells were harvested for RNA extraction to determine the genes expressed. All experiments were performed at least three times to ensure repeatability. RESULTS BMP-2 significantly increased aggrecan and collagen type II mRNA expression 8.30 and 4.61 times, respectively, and decreased versican mRNA expression 0.54 times as compared with control. Collagen type I production and mRNA level were not changed. BMP-2 significantly increased transforming growth factor-beta1 (TGFbeta1) and BMP-7 mRNA expression 2.32 and 2.45 times, respectively, compared with control. There was no significant change in BMP-6 mRNA expression. BMPR type IB and II mRNA expressions were increased and BMPR type 1A mRNA expression was decreased, but none of these differences was significant. CONCLUSIONS The results of this study show that in rat intervertebral disc cells, BMP-2 increases aggrecan and collagen type II mRNA expression and decreases versican gene expression. BMP-2 also up-regulates mRNA expression for BMP-7 and TGFbeta but has no significant effect on the BMPRs.
Collapse
MESH Headings
- Aggrecans
- Animals
- Bone Morphogenetic Protein 2
- Bone Morphogenetic Protein 7
- Bone Morphogenetic Protein Receptors
- Bone Morphogenetic Protein Receptors, Type I
- Bone Morphogenetic Protein Receptors, Type II
- Bone Morphogenetic Proteins/drug effects
- Bone Morphogenetic Proteins/genetics
- Bone Morphogenetic Proteins/metabolism
- Bone Morphogenetic Proteins/pharmacology
- Cartilage/cytology
- Cartilage/drug effects
- Cartilage/metabolism
- Cells, Cultured
- Collagen Type I/genetics
- Collagen Type II/genetics
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Extracellular Matrix Proteins/genetics
- Gene Expression/drug effects
- Gene Expression/physiology
- Glycosaminoglycans/genetics
- Intervertebral Disc/cytology
- Intervertebral Disc/drug effects
- Intervertebral Disc/metabolism
- Lectins, C-Type
- Protein Serine-Threonine Kinases/genetics
- Proteoglycans/genetics
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Growth Factor/drug effects
- Receptors, Growth Factor/genetics
- Receptors, Growth Factor/metabolism
- Regeneration/drug effects
- Regeneration/physiology
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
- Transforming Growth Factor beta1
- Up-Regulation/drug effects
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Jun Li
- Emory University, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | | | | |
Collapse
|
47
|
zur Nieden NI, Kempka G, Rancourt DE, Ahr HJ. Induction of chondro-, osteo- and adipogenesis in embryonic stem cells by bone morphogenetic protein-2: effect of cofactors on differentiating lineages. BMC DEVELOPMENTAL BIOLOGY 2005; 5:1. [PMID: 15673475 PMCID: PMC548146 DOI: 10.1186/1471-213x-5-1] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Accepted: 01/26/2005] [Indexed: 11/10/2022]
Abstract
Background Recently, tissue engineering has merged with stem cell technology with interest to develop new sources of transplantable material for injury or disease treatment. Eminently interesting, are bone and joint injuries/disorders because of the low self-regenerating capacity of the matrix secreting cells, particularly chondrocytes. ES cells have the unlimited capacity to self-renew and maintain their pluripotency in culture. Upon induction of various signals they will then differentiate into distinctive cell types such as neurons, cardiomyocytes and osteoblasts. Results We present here that BMP-2 can drive ES cells to the cartilage, osteoblast or adipogenic fate depending on supplementary co-factors. TGFβ1, insulin and ascorbic acid were identified as signals that together with BMP-2 induce a chondrocytic phenotype that is characterized by increased expression of cartilage marker genes in a timely co-ordinated fashion. Expression of collagen type IIB and aggrecan, indicative of a fully mature state, continuously ascend until reaching a peak at day 32 of culture to approximately 80-fold over control values. Sox9 and scleraxis, cartilage specific transcription factors, are highly expressed at very early stages and show decreased expression over the time course of EB differentiation. Some smaller proteoglycans, such as decorin and biglycan, are expressed at earlier stages. Overall, proteoglycan biosynthesis is up-regulated 7-fold in response to the supplements added. BMP-2 induced chondrocytes undergo hypertrophy and begin to alter their expression profile towards osteoblasts. Supplying mineralization factors such as β-glycerophosphate and vitamin D3 with the culture medium can facilitate this process. Moreover, gene expression studies show that adipocytes can also differentiate from BMP-2 treated ES cells. Conclusions Ultimately, we have found that ES cells can be successfully triggered to differentiate into chondrocyte-like cells, which can further alter their fate to become hypertrophic, and adipocytes. Compared with previous reports using a brief BMP-2 supplementation early in differentiation, prolonged exposure increased chondrogenic output, while supplementation with insulin and ascorbic acid prevented dedifferentiation. These results provide a foundation for the use of ES cells as a potential therapy in joint injury and disease.
Collapse
Affiliation(s)
- Nicole I zur Nieden
- Molecular & Genetic Toxicology, Bayer HealthCare AG, Wuppertal, Germany
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
- Faculty of Medicine, Dept. of Biochemistry & Molecular Biology, University of Calgary, HMRB 331, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Grazyna Kempka
- Molecular & Genetic Toxicology, Bayer HealthCare AG, Wuppertal, Germany
| | - Derrick E Rancourt
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
| | - Hans-Jürgen Ahr
- Molecular & Genetic Toxicology, Bayer HealthCare AG, Wuppertal, Germany
| |
Collapse
|
48
|
Hing KA. Bone repair in the twenty-first century: biology, chemistry or engineering? PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2004; 362:2821-2850. [PMID: 15539372 DOI: 10.1098/rsta.2004.1466] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Increases in reconstructive orthopaedic surgery, such as total hip replacement and spinal fusion, resulting from advances in surgical practice and the ageing population, have lead to a demand for bone graft that far exceeds supply. Consequently, a number of synthetic bone-graft substitutes (BGSs) have been developed with mixed success and surgical acceptance. Skeletal tissue regeneration requires the interaction of three basic elements: cells, growth factors (GFs) and a permissive scaffold. This can be achieved by pre-loading a synthetic scaffold with GFs or pre-expanded cells; however, a 'simpler' approach is to design intrinsic 'osteoinductivity' into your BGS, i.e. the capability to recruit and stimulate the patient's own GFs and stem cells. Through investigation of the mechanisms controlling bone repair in BGSs, linking interactions between the local chemical and physical environment, scientists are currently developing osteoinductive materials that can stimulate bone regeneration through control of the scaffold chemistry and structure. Moreover, this body of research is providing the foundations for future generations of BGSs and bone-repair therapies and may ultimately contribute towards improving the quality of life through maintenance of the skeleton and reversal of disease states, as opposed to the mending of broken bones that we currently practice. Will we be able to grow our own bones in a bioreactor for use as autologous graft materials in the future? Could surgery be limited to accidental trauma cases, with greater restoration of function through biochemical or gene therapies? The technology and research probes necessary to this task are currently being developed with the advent of nanotechnology, genomics and proteomics: are we about to embark on a chemical revolution in medicine? This paper aims to discuss some of the current thinking on the mechanisms behind bioactivity and biocompatibility in bone and how a fuller understanding of the interactions between cells and the materials used today could bring about completely new approaches for the treatment of bone fracture and disease tomorrow.
Collapse
Affiliation(s)
- Karin A Hing
- Interdisciplinary Research Centre in Biomedical Materials, Queen Mary, University of London, London E1 4NS, UK.
| |
Collapse
|
49
|
Gafni Y, Pelled G, Zilberman Y, Turgeman G, Apparailly F, Yotvat H, Galun E, Gazit Z, Jorgensen C, Gazit D. Gene therapy platform for bone regeneration using an exogenously regulated, AAV-2-based gene expression system. Mol Ther 2004; 9:587-95. [PMID: 15093189 DOI: 10.1016/j.ymthe.2003.12.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2002] [Accepted: 12/17/2003] [Indexed: 11/15/2022] Open
Abstract
Viral delivery of the therapeutic gene bone morphogenetic protein-2 (BMP-2) is a promising approach for bone regeneration. The human parvovirus adeno-associated virus (AAV) type 2 is considered one of the most encouraging viral vector systems because of its high transduction rates and biosafety ratings. Bone morphogenetic protein-2 is a highly potent osteoinductive protein, which induces bone formation in vivo and osteogenic differentiation in vitro. The exogenous regulation of BMP-2 expression in bone-regenerating sites is required to control BMP-2 protein secretion, thus promoting safe and controlled bone formation and regeneration. We have therefore constructed a dual-construct vector for the recombinant AAV (rAAV)-based recombinant human BMP-2 (rhBMP-2) gene delivery system, which is regulated by the tetracycline-sensitive promoter (TetON). Each vector was encapsidated separately, yielding two recombinant viruses. We evaluated the efficiency of rAAV-hBMP-2 to induce bone formation in ectopic and orthotopic sites. Doxycycline (Dox), an analogue of tetracycline, was orally administered to mice via their drinking water to induce rhBMP-2 expression. Bone formation was measured using quantitative imaging-microcomputerized tomography and cooled charge-coupled device imaging-to detect osteogenic activity at the cellular level, detecting osteocalcin expression. The rAAV-hBMP-2-treated mice that were given Dox demonstrated bone formation in both in vivo models compared to none in mice prevented from receiving Dox. Thus, the Tet-regulated rAAV-hBMP-2 vector is an effective means of induction and regulation of bone regeneration and repair.
Collapse
Affiliation(s)
- Yossi Gafni
- Skeletal Biotechnology Laboratory, Hebrew University-Hadassah Medical Center, Ein Kerem, P.O. Box 12272, Jerusalem 91120, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jay KE, Rouleau A, Underhill TM, Bhatia M. Identification of a novel population of human cord blood cells with hematopoietic and chondrocytic potential. Cell Res 2004; 14:268-82. [PMID: 15353124 DOI: 10.1038/sj.cr.7290228] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
With the exception of mature erythrocytes, cells within the human hematopoietic system are characterized by the cell surface expression of the pan-leukocyte receptor CD45. Here, we identify a novel subset among mononuclear cord blood cells depleted of lineage commitment markers (Lin-) that are devoid of CD45 expression. Surprisingly, functional examination of Lin-CD45- cells also lacking cell surface CD34 revealed they were capable of multipotential hematopoietic progenitor capacity. Co-culture with mouse embryonic limb bud cells demonstrated that Lin-CD45-CD34- cells were capable of contributing to cartilage nodules and differentiating into human chondrocytes. BMP-4, a mesodermal factor known to promote chondrogenesis, significantly augmented Lin-CD45-CD34- differentiation into chondrocytes. Moreover, unlike CD34+ human hematopoietic stem cells, Lin-CD45-CD34- cells were unable to proliferate or survive in liquid cultures, whereas single Lin-CD45-CD34- cells were able to chimerize the inner cell mass (ICM) of murine blastocysts and proliferate in this embryonic environment. Our study identifies a novel population of Lin-CD45-CD34- cells capable of commitment into both hematopoietic and chondrocytic lineages, suggesting that human cord blood may provide a more ubiquitous source of tissue with broader developmental potential than previously appreciated.
Collapse
Affiliation(s)
- Karen E Jay
- Stem Cell Biology and Regenerative Medicine, The John P. Robarts Research Institute, 100 Perth Drive, London, Ontario, N6A 5K8, Canada
| | | | | | | |
Collapse
|