Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Stem Cells ; 41(5): 493-504, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36888549

ABSTRACT

Regulator of G protein signaling 5 (RGS5) is a GTPase activator for heterotrimeric G-protein α-subunits, shown to be a marker of pericytes. Bone marrow stromal cell population (BMSCs) is heterogeneous. Populations of mesenchymal progenitors, cells supportive of hematopoiesis, and stromal cells regulating bone remodeling have been recently identified. Periosteal and bone marrow mesenchymal stem cells (MSCs) are participating in fracture healing, but it is difficult to distinguish the source of cells within the callus. Considering that perivascular cells exert osteoprogenitor potential, we generated an RGS5 transgenic mouse model (Rgs5-CreER) which when crossed with Ai9 reporter animals (Rgs5/Tomato), is suitable for lineage tracing during growth and post-injury. Flow cytometry analysis and histology confirmed the presence of Rgs5/Tomato+ cells within CD31+ endothelial, CD45+ hematopoietic, and CD31-CD45- mesenchymal/perivascular cells. A tamoxifen chase showed expansion of Rgs5/Tomato+ cells expressing osterix within the trabeculae positioned between mineralized matrix and vasculature. Long-term chase showed proportion of Rgs5/Tomato+ cells contributes to mature osteoblasts expressing osteocalcin. Following femoral fracture, Rgs5/Tomato+ cells are observed around newly formed bone within the BM cavity and expressed osterix and osteocalcin, while contribution within periosteum was low and limited to fibroblastic callus with very few positive chondrocytes. In addition, BM injury model confirmed that RGS5-Cre labels population of BMSCs expands during injury and participates in osteogenesis. Under homeostatic conditions, lineage-traced RGS5 cells within the trabecular area demonstrate osteoprogenitor capacity that in an injury model contributes to new bone formation primarily within the BM niche.


Subject(s)
Bony Callus , RGS Proteins , Mice , Animals , Osteocalcin/metabolism , Bony Callus/metabolism , Bony Callus/pathology , Osteogenesis , Fracture Healing/physiology , Chondrocytes/metabolism , Mice, Transgenic , Osteoblasts/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism
2.
Sci Rep ; 10(1): 4963, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32188889

ABSTRACT

Enamel is secreted by ameloblasts derived from tooth epithelial stem cells (SCs). Humans cannot repair or regenerate enamel, due to early loss of tooth epithelial SCs. Contrarily in the mouse incisors, epithelial SCs are maintained throughout life and endlessly generate ameloblasts, and thus enamel. Here we isolated Sox2-GFP+ tooth epithelial SCs which generated highly cellular spheres following a novel in vitro strategy. This system enabled analysis of SC regulation by various signaling molecules, and supported the stimulatory and inhibitory roles of Shh and Bmp, respectively; providing better insight into the heterogeneity of the SCs. Further, we generated a novel mouse reporter, Enamelin-tdTomato for identification of ameloblasts in live tissues and cells, and used it to demonstrate presence of ameloblasts in the new 3D co-culture system of dental SCs. Collectively, our results provide means of generating 3D tooth epithelium from adult SCs which can be utilized toward future generation of enamel.


Subject(s)
Ameloblasts/cytology , Cell Differentiation , Epithelial Cells/cytology , Stem Cells/cytology , Tooth/cytology , Ameloblasts/metabolism , Animals , Cells, Cultured , Coculture Techniques , Epithelial Cells/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Stem Cells/metabolism , Tooth/metabolism
3.
Methods Mol Biol ; 1723: 385-396, 2018.
Article in English | MEDLINE | ID: mdl-29344873

ABSTRACT

Laser-capture microdissection (LCM) coupled to downstream RNA analysis poses unique difficulties for the evaluation of mineralized tissues. A rapid protocol was thus developed to enable sufficient integrity of bone and cartilage tissue for reliable sectioning, while minimizing RNA loss associated with prolonged decalcification and purification steps. Specifically, the protocol involves pump-assisted, cardiac perfusion-fixation with paraformaldehyde, and moderate digestion of LCM-acquired tissue with proteinase K followed by DNase treatment and separation of RNA using magnetic beads. Reverse transcription and cDNA synthesis are performed immediately after RNA purification, without need for further protein removal.


Subject(s)
Bone and Bones/metabolism , Cartilage/metabolism , Disease Models, Animal , Induced Pluripotent Stem Cells/metabolism , Laser Capture Microdissection/methods , Mesenchymal Stem Cells/metabolism , RNA/analysis , Skull/metabolism , Animals , Bone and Bones/pathology , Cartilage/pathology , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/pathology , Mesenchymal Stem Cells/pathology , Mice , Perfusion , RNA/genetics , RNA/isolation & purification , Skull/pathology
4.
Dev Biol ; 426(1): 56-68, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28438606

ABSTRACT

Limb synovial joints are composed of distinct tissues, but it is unclear which progenitors produce those tissues and how articular cartilage acquires its functional postnatal organization characterized by chondrocyte columns, zone-specific cell volumes and anisotropic matrix. Using novel Gdf5CreERT2 (Gdf5-CE), Prg4-CE and Dkk3-CE mice mated to R26-Confetti or single-color reporters, we found that knee joint progenitors produced small non-migratory progenies and distinct local tissues over prenatal and postnatal time. Stereological imaging and quantification indicated that the columns present in juvenile-adult tibial articular cartilage consisted of non-daughter, partially overlapping lineage cells, likely reflecting cell rearrangement and stacking. Zone-specific increases in cell volume were major drivers of tissue thickening, while cell proliferation or death played minor roles. Second harmonic generation with 2-photon microscopy showed that the collagen matrix went from being isotropic and scattered at young stages to being anisotropic and aligned along the cell stacks in adults. Progenitor tracing at prenatal or juvenile stages showed that joint injury provoked a massive and rapid increase in synovial Prg4+ and CD44+/P75+ cells some of which filling the injury site, while neighboring chondrocytes appeared unresponsive. Our data indicate that local cell populations produce distinct joint tissues and that articular cartilage growth and zonal organization are mainly brought about by cell volume expansion and topographical cell rearrangement. Synovial Prg4+ lineage progenitors are exquisitely responsive to acute injury and may represent pioneers in joint tissue repair.


Subject(s)
Cartilage, Articular , Cell Size , Chondrogenesis/physiology , Knee Injuries/metabolism , Knee Joint/growth & development , Mesenchymal Stem Cells/metabolism , Animals , Cartilage, Articular/cytology , Cartilage, Articular/embryology , Cartilage, Articular/growth & development , Cartilage, Articular/injuries , Cell Differentiation/physiology , Cell Lineage , Cell Proliferation , Chondrocytes/cytology , Collagen/metabolism , Growth Differentiation Factor 5/metabolism , Knee Joint/cytology , Mice , Mice, Transgenic , Synovial Membrane/cytology
5.
Connect Tissue Res ; 57(6): 507-515, 2016 11.
Article in English | MEDLINE | ID: mdl-27184388

ABSTRACT

Purpose of this study: To elucidate the origin of cell populations that contribute to rotator cuff healing, we developed a mouse surgical model where a full-thickness, central detachment is created in the supraspinatus. MATERIALS AND METHODS: Three different inducible Cre transgenic mice with Ai9-tdTomato reporter expression (PRG4-9, αSMA-9, and AGC-9) were used to label different cell populations in the shoulder. The defect was created surgically in the supraspinatus. The mice were injected with tamoxifen at surgery to label the cells and sacrificed at 1, 2, and 5 weeks postoperatively. Frozen sections were fluorescently imaged then stained with Toluidine Blue and re-imaged. RESULTS: Three notable changes were apparent postoperatively. (1) A long thin layer of tissue formed on the bursal side overlying the supraspinatus tendon. (2) The tendon proximal to the defect initially became hypercellular and disorganized. (3) The distal stump at the insertion underwent minimal remodeling. In the uninjured shoulder, tdTomato expression was seen in the tendon midsubstance and paratenon cell on the bursal side in PRG4-9, in paratenon, blood vessels, and periosteum of acromion in SMA-9, and in articular cartilage, unmineralized fibrocartilage of supraspinatus enthesis, and acromioclavicular joint in AGC-9 mice. In the injured PRG4-9 and SMA-9 mice, the healing tissues contained an abundant number of tdTomato+ cells, while minimal contribution of tdTomato+ cells was seen in AGC-9 mice. CONCLUSIONS: The study supports the importance of the bursal side of the tendon to rotator cuff healing and PRG4 and αSMA may be markers for these progenitor cells.


Subject(s)
Rotator Cuff Injuries/pathology , Rotator Cuff/pathology , Wound Healing , Animals , Deltoid Muscle/pathology , Disease Models, Animal , Genes, Reporter , Integrases/metabolism , Mice, Transgenic , Shoulder Dislocation/pathology , Shoulder Injuries , Shoulder Joint/pathology
6.
PLoS One ; 8(8): e71318, 2013.
Article in English | MEDLINE | ID: mdl-23951132

ABSTRACT

We have carried out fate mapping studies using Osterix-EGFPCre and Osterix-CreERt animal models and found Cre reporter expression in many different cell types that make up the bone marrow stroma. Constitutive fate mapping resulted in the labeling of different cellular components located throughout the bone marrow, whereas temporal fate mapping at E14.5 resulted in the labeling of cells within a region of the bone marrow. The identity of cell types marked by constitutive and temporal fate mapping included osteoblasts, adipocytes, vascular smooth muscle, perineural, and stromal cells. Prolonged tracing of embryonic precursors labeled at E14.5dpc revealed the continued existence of their progeny up to 10 months of age, suggesting that fate mapped, labeled embryonic precursors gave rise to long lived bone marrow progenitor cells. To provide further evidence for the marking of bone marrow progenitors, bone marrow cultures derived from Osterix-EGFPCre/Ai9 mice showed that stromal cells retained Cre reporter expression and yielded a FACS sorted population that was able to differentiate into osteoblasts, adipocytes, and chondrocytes in vitro and into osteoblasts, adipocytes, and perivascular stromal cells after transplantation. Collectively, our studies reveal the developmental process by which Osterix-Cre labeled embryonic progenitors give rise to adult bone marrow progenitors which establish and maintain the bone marrow stroma.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow/ultrastructure , Integrases/analysis , Stem Cells/cytology , Stromal Cells/cytology , Transcription Factors/analysis , Animals , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/cytology , Mice , Sp7 Transcription Factor , Staining and Labeling/methods
7.
Stem Cells ; 30(2): 187-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22083974

ABSTRACT

Adult mesenchymal progenitor cells have enormous potential for use in regenerative medicine. However, the true identity of the progenitors in vivo and their progeny has not been precisely defined. We hypothesize that cells expressing a smooth muscle α-actin promoter (αSMA)-directed Cre transgene represent mesenchymal progenitors of adult bone tissue. By combining complementary colors in combination with transgenes activating at mature stages of the lineage, we characterized the phenotype and confirmed the ability of isolated αSMA(+) cells to progress from a progenitor to fully mature state. In vivo lineage tracing experiments using a new bone formation model confirmed the osteogenic phenotype of αSMA(+) cells. In vitro analysis of the in vivo-labeled SMA9(+) cells supported their differentiation potential into mesenchymal lineages. Using a fracture-healing model, αSMA9(+) cells served as a pool of fibrocartilage and skeletal progenitors. Confirmation of the transition of αSMA9(+) progenitor cells to mature osteoblasts during fracture healing was assessed by activation of bone-specific Col2.3emd transgene. Our findings provide a novel in vivo identification of defined population of mesenchymal progenitor cells with active role in bone remodeling and regeneration.


Subject(s)
Cell Lineage , Mesenchymal Stem Cells/metabolism , Actins/genetics , Actins/metabolism , Animals , Antigens, Differentiation/metabolism , Bone Marrow Cells/metabolism , Bone Regeneration , Bone Remodeling , Cell Differentiation , Female , Fracture Healing , Gene Expression Regulation , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , Phenotype , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Tibia/pathology
8.
PLoS One ; 5(7): e11853, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20686615

ABSTRACT

BACKGROUND: Directed differentiation of human induced pluripotent stem cells (hiPSC) into functional, region-specific neural cells is a key step to realizing their therapeutic promise to treat various neural disorders, which awaits detailed elucidation. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed neural differentiation from various hiPSC lines generated by others and ourselves. Although heterogeneity in efficiency of neuroepithelial (NE) cell differentiation was observed among different hiPSC lines, the NE differentiation process resembles that from human embryonic stem cells (hESC) in morphology, timing, transcriptional profile, and requirement for FGF signaling. NE cells differentiated from hiPSC, like those from hESC, can also form rostral phenotypes by default, and form the midbrain or spinal progenitors upon caudalization by morphogens. The rostrocaudal neural progenitors can further mature to develop forebrain glutamatergic projection neurons, midbrain dopaminergic neurons, and spinal motor neurons, respectively. Typical ion channels and action potentials were recorded in the hiPSC-derived neurons. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that hiPSC, regardless of how they were derived, can differentiate into a spectrum of rostrocaudal neurons with functionality, which supports the considerable value of hiPSC for study and treatment of patient-specific neural disorders.


Subject(s)
Cell Differentiation/physiology , Glutamic Acid/metabolism , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , Prosencephalon/cytology , Cell Differentiation/genetics , Electrophysiology , Flow Cytometry , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction
9.
Bone ; 47(5): 916-25, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20673822

ABSTRACT

While human bone marrow derived mesenchymal stem cells (BMSCs) are of great interest for their potential therapeutic value, their murine equivalent remains an important basic research model that can provide critical insights into the biology of this progenitor cell population. Here we present a novel transgenic strategy that allowed for the selective identification and isolation of murine BMSCs at the early stages of stromal cell culture. This strategy involved crossing Twist2 -Cre mice with Cre reporter mice such as Z/EG or Ai9, which express EGFP or Tomato fluorescent protein, respectively, upon Cre mediated excision of a stop sequence. Using this approach, we identified an adherent fluorescent protein+cell population (T2C+) that is present during the earliest stages of colony formation and by day 5 of culture represents ~20% of the total cell population. Cell surface profiling by flow cytometry showed that T2C+cells are highly positive for SCA1 and CD29 and negative for CD45, CD117, TIE2, and TER119. Isolation of T2C+cells by FACS selected for a cell population with skeletal potential that can be directed to differentiate into osteoblasts, adipocytes, or chondrocytes. We also demonstrated in a calvarial bone defect model that T2C+cells retain a strong efficacy for osteogenic repair and can support a hematopoietic environment. Collectively, these studies provide evidence that the Twist2-Cre x Cre reporter breeding strategy can be used to positively identify and isolate multipotent murine BMSCs.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Animals , Bone Diseases/therapy , Bone Marrow Cells/metabolism , Cell Differentiation , Flow Cytometry , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Parietal Bone/pathology , Polymerase Chain Reaction
10.
Bone ; 47(2): 360-70, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20510392

ABSTRACT

Here we determine the Fibroblast Growth Factor-2 (FGF2) dependency of the time course of changes in bone mass in female mice. This study extends our earlier reports that knockout of the FGF2 gene (Fgf2) caused low turnover bone loss in Fgf2(-/-) male mice by examining bone loss with age in Fgf2(-/-) female mice, and by assessing whether reduced bone formation is associated with differentiation of bone marrow stromal cells (BMSCs) towards the adipocyte lineage. Bone mineral density (BMD) was similar in 3-month-old female Fgf2(+/+) and Fgf2(-/-) mice but was significantly reduced as early as 5 months of age in Fgf2(-/-) mice. In vivo studies showed that there was a greater accumulation of marrow fat in long bones of 14 and 20 month old Fgf2(-/-) mice compared with Fgf2(+/+) littermates. To study the effect of disruption of FGF2 on osteoblastogenesis and adipogenesis, BMSCs from both genotypes were cultured in osteogenic or adipogenic media. Reduced alkaline phosphatase positive (ALP), mineralized colonies and a marked increase in adipocytes were observed in Fgf2(-/-) BMSC cultures. These cultures also showed an increase in the mRNA of the adipogenic transcription factor PPARgamma2 as well as the downstream target genes aP2 and adiponectin. Treatment with exogenous FGF2 blocked adipocyte formation and increased ALP colony formation and ALP activity in BMSC cultures of both genotypes. These results support an important role for endogenous FGF2 in osteoblast (OB) lineage determination. Alteration in FGF2 signaling may contribute to impaired OB bone formation capacity and to increased bone marrow fat accumulation both of which are characteristics of aged bone.


Subject(s)
Adipogenesis/genetics , Bone Marrow Cells/cytology , Fibroblast Growth Factor 2/genetics , Gene Deletion , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Absorptiometry, Photon , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Aging/drug effects , Animals , Bone Density/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Count , Cell Proliferation/drug effects , Cells, Cultured , Colony-Forming Units Assay , Female , Fibroblast Growth Factor 2/deficiency , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/drug effects , Mice , Mice, Knockout , Osteogenesis/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism
11.
Coll Antropol ; 33(3): 919-24, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19860126

ABSTRACT

We analyzed proliferation and differentiation of calvarial osteoblasts derived from Msx2 deficient in comparison with wild type mice. Calvarial osteoblast cultures from five to eight days old Msx2 deficient, heterozygous and wild type mice were studied for difference in proliferation and differentiation. Proliferation rate was assessed by counting cell number, BrdU and Calcein AM labeling. Differentiation was assessed by Von Kossa and alkaline phosphatase staining, northern blot hybridization with bone differentiation markers, infection of cell cultures with retrovirus expressing GFP under the control of type I collagen promoter fragment. At day six, cell number in cell culture derived from Msx2 deficient mice was 20% lower then in culture from wild type mice. There were 16.8% BrdU labeled cells in cell culture from Msx2 deficient mice, 20.9% in culture from heterozygous mice and 21.6% in culture from wild type mice. Cell cultures from Msx2 deficient mice showed lower intensity of fluorescence when marked with Calcein AM then cultures from wild type mice. Von Kossa staining showed increased mineralization and northern blot analysis showed increased levels of bone differentiation markers in cell cultures derived from Msx2 deficient mice. GFP came on earlier in Msx2 deficient cultures after infection with Col2.3 GFP retrovirus. We conclude that calvarial osteoblasts derived from Msx2 deficient mice have a lower rate of proliferation and demonstrate increased osteoblastic differentiation when compared to osteoblasts derived from wild type mice.


Subject(s)
Homeodomain Proteins/physiology , Osteoblasts/cytology , Skull/growth & development , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL
12.
Bone ; 45(4): 682-92, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19539797

ABSTRACT

Osteocytes represent the most abundant cellular component of mammalian bones with important functions in bone mass maintenance and remodeling. To elucidate the differential gene expression between osteoblasts and osteocytes we completed a comprehensive analysis of their gene profiles. Selective identification of these two mature populations was achieved by utilization of visual markers of bone lineage cells. We have utilized dual GFP reporter mice in which osteocytes are expressing GFP (topaz) directed by the DMP1 promoter, while osteoblasts are identified by expression of GFP (cyan) driven by 2.3 kb of the Col1a1 promoter. Histological analysis of 7-day-old neonatal calvaria confirmed the expression pattern of DMP1GFP in osteocytes and Col2.3 in osteoblasts and osteocytes. To isolate distinct populations of cells we utilized fluorescent activated cell sorting (FACS). Cell suspensions were subjected to RNA extraction, in vitro transcription and labeling of cDNA and gene expression was analyzed using the Illumina WG-6v1 BeadChip. Following normalization of raw data from four biological replicates, 3444 genes were called present in all three sorted cell populations: GFP negative, Col2.3cyan(+) (osteoblasts), and DMP1topaz(+) (preosteocytes and osteocytes). We present the genes that showed in excess of a 2-fold change for gene expression between DMP1topaz(+) and Col2.3cyan(+) cells. The selected genes were classified and grouped according to their associated gene ontology terms. Genes clustered to osteogenesis and skeletal development such as Bmp4, Bmp8a, Dmp1, Enpp1, Phex and Ank were highly expressed in DMP1topaz(+)cells. Most of the genes encoding extracellular matrix components and secreted proteins had lower expression in DMP1topaz(+) cells, while most of the genes encoding plasma membrane proteins were increased. Interestingly a large number of genes associated with muscle development and function and with neuronal phenotype were increased in DMP1topaz(+) cells, indicating some new aspects of osteocyte biology. Although a large number of genes differentially expressed in DMP1topaz(+) and Col2.3cyan(+) cells in our study have already been assigned to bone development and physiology, for most of them we still lack any substantial data. Therefore, isolation of osteocyte and osteoblast cell populations and their subsequent microarray analysis allowed us to identify a number or genes and pathways with potential roles in regulation of bone mass.


Subject(s)
Gene Expression Profiling , Osteocytes/metabolism , Animals , Animals, Newborn , Cell Membrane/metabolism , Cell Separation , Extracellular Matrix/genetics , Flow Cytometry , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Muscles/cytology , Muscles/embryology , Muscles/metabolism , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Skull/cytology , Skull/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Dev Biol ; 316(2): 336-49, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18339367

ABSTRACT

To address the functions of FGFR2 and FGFR3 signaling during mandibular skeletogenesis, we over-expressed in the developing chick mandible, replication-competent retroviruses carrying truncated FGFR2c or FGFR3c that function as dominant negative receptors (RCAS-dnFGFR2 and RCAS-dnFGFR3). Injection of RCAS-dnFGFR3 between HH15 and 20 led to reduced proliferation, increased apoptosis, and decreased differentiation of chondroblasts in Meckel's cartilage. These changes resulted in the formation of a hypoplastic mandibular process and truncated Meckel's cartilage. This treatment also affected the proliferation and survival of osteoprogenitor cells in osteogenic condensations, leading to the absence of five mandibular bones on the injected side. Injection of RCAS-dnFGFR2 between HH15 and 20 or RCAS-dnFGFR3 at HH26 did not affect the morphogenesis of Meckel's cartilage but resulted in truncations of the mandibular bones. RCAS-dnFGFR3 affected the proliferation and survival of the cells within the periosteum and osteoblasts. Together these results demonstrate that FGFR3 signaling is required for the elongation of Meckel's cartilage and FGFR2 and FGFR3 have roles during intramembranous ossification of mandibular bones.


Subject(s)
Cartilage/embryology , Mandible/embryology , Morphogenesis/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Animals , Cells, Cultured , Chick Embryo , Chickens , Female , Osteogenesis , Ovum/physiology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/physiology , Receptor, Fibroblast Growth Factor, Type 3/genetics , Sequence Deletion , Signal Transduction
14.
Dev Biol ; 316(2): 458-70, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18280462

ABSTRACT

Our laboratory and others have shown that overexpression of Dlx5 stimulates osteoblast differentiation. Dlx5(-/-)/Dlx6(-/-) mice have more severe craniofacial and limb defects than Dlx5(-/-), some of which are potentially due to defects in osteoblast maturation. We wished to investigate the degree to which other Dlx genes compensate for the lack of Dlx5, thus allowing normal development of the majority of skeletal elements in Dlx5(-/-) mice. Dlx gene expression in cells from different stages of the osteoblast lineage isolated by FACS sorting showed that Dlx2, Dlx5 and Dlx6 are expressed most strongly in less mature osteoblasts, whereas Dlx3 is very highly expressed in differentiated osteoblasts and osteocytes. In situ hybridization and Northern blot analysis demonstrated the presence of endogenous Dlx3 mRNA within osteoblasts and osteocytes. Dlx3 strongly upregulates osteoblastic markers with a potency comparable to Dlx5. Cloned chick or mouse Dlx6 showed stimulatory effects on osteoblast differentiation. Our results suggest that Dlx2 and Dlx6 have the potential to stimulate osteoblastic differentiation and may compensate for the absence of Dlx5 to produce relatively normal osteoblastic differentiation in Dlx5 knockout mice, while Dlx3 may play a distinct role in late stage osteoblast differentiation and osteocyte function.


Subject(s)
Homeodomain Proteins/genetics , Osteoblasts/physiology , Osteocytes/physiology , Transcription Factors/genetics , Animals , Animals, Newborn , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Cloning, Molecular , Gene Expression Regulation , Mice , Mice, Inbred Strains , Mice, Transgenic , Osteoblasts/cytology , Osteocytes/cytology , Polymerase Chain Reaction , RNA, Messenger/genetics , Stromal Cells/cytology , Stromal Cells/physiology
15.
Coll Antropol ; 30(2): 401-4, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16848158

ABSTRACT

The aim of this paper is identification of regulatory sequences downstream of -1683 base pairs (bp) in the rat Col1a1 promoter important for expression in osteoblasts. Previous findings suggest that a rat Col1a1 gene fragment extending from -1719 to + 115 bp linked to the chloramphenicol acetyl transferase (CAT) reporter gene (ColCAT1719) is highly and selectively expressed in osteoblasts. Three internal deletions within the ColCAT1719 construct were generated and stably transfected into ROS 17/2.8 cells. CAT activity was measured in cell extracts. An internal deletion of ColCAT1719 from -1637 to -504 bp caused an almost complete loss of CAT activity, whereas deletions of -1284 to -905 bp and -1284 to -451 bp had little effect on CAT activity. We hypothesized that removal of a Runx2/Cbfa1 consensus site at -1376 bp may have caused the loss of activity produced by the -1637 to -504 bp deletion. To test this hypothesis, we produced a more restricted internal deletion of ColCAT1719 from -1418 to -1284 bp, which removes this site. This deletion did not affect promoter activity. Our results suggest that the Runx2 site at -1376 bp by itself does not influence Col1719 promoter activity. Future studies will focus on the region between -1637 to 1418 bp, which contains several potentially interesting transcription factor binding sites.


Subject(s)
Base Sequence , Collagen Type I/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Promoter Regions, Genetic/genetics , Sequence Deletion , Animals , Cell Line , Chloramphenicol O-Acetyltransferase/metabolism , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation/genetics , Osteoblasts/metabolism , Rats , Transfection
16.
Int J Dev Biol ; 48(7): 645-53, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15470637

ABSTRACT

The Cre/loxP recombination system can be used to circumvent many of the limitations of generalized gene ablation in mice. Here we present the development and characterization of transgenic mice in which Cre recombinase has been targeted to cells of the osteoblast lineage with 2.3 kb (Col 2.3-Cre) and 3.6 kb (Col 3.6-Cre) fragments of the rat Col1a1 promoter. Cre mRNA was detected in calvaria and long bone of adult Col 2.3-Cre and Col 3.6-Cre mice, as well as in tendon and skin of Col 3.6-Cre mice. To obtain a historical marking of the temporal and spatial pattern of Cre-mediated gene rearrangement, Col-Cre mice were bred with ROSA26 (R26R) mice in which Cre-mediated excision of a floxed cassette results in LacZ expression. In Col 2.3-Cre;R26R and Col 3.6-Cre;R26R progeny, calvarial and long bone osteoblasts showed intense beta-gal staining at embryonic day 18 and postnatal day 5. The spatial pattern of beta-gal staining was more restricted in bone and in bone marrow stromal cultures established from Col 2.3-Cre;R26R mice. Similar differences in the spatial patterns of expression were seen in transgenic bone carrying Col1a1-GFP visual reporters. Our data suggest that Col 2.3-Cre and Col 3.6-Cre transgenic mice may be useful for conditional gene targeting in vivo or for obtaining osteoblast populations for in vitro culture in which a gene of interest has been inactivated.


Subject(s)
Genetic Techniques , Muscle, Skeletal/metabolism , Osteoblasts/metabolism , Animals , Blotting, Northern , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Cloning, Molecular , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Genotype , Green Fluorescent Proteins/metabolism , Integrases/metabolism , Mice , Mice, Transgenic , Models, Genetic , Plasmids/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA/metabolism , Rats , Time Factors , Tissue Distribution , Transgenes , beta-Galactosidase/metabolism
17.
Nucleic Acids Res ; 32(4): 1512-7, 2004.
Article in English | MEDLINE | ID: mdl-14999093

ABSTRACT

We have previously demonstrated that a modified U1 snRNA inhibits expression of a number of targeted transgenes. Here we exploit the ability of the modified U1 snRNA to inhibit endogenous gene expression and define the mechanism responsible for this inhibitory action. MC3T3-E1 cells stably transfected with U1 anti-Cbfa1 show a change of morphology from polygonal to fibroblast-like cells. This visual observation was supported by an 80% reduction of Cbfa1 expression and suppression of downstream genes associated with osteoblast differentiation. In rat ROS 17/2.8 cells, osteocalcin and Col1a1 gene expression was reduced up to 90% by the U1 anti-osteocalcin or U1 anti-Col1a1 construct, respectively. The length of mature osteocalcin mRNA poly(A) tail was significantly shortened in the targeted mRNA by transcript-specific poly(A) test (PAT)-PCR. These data demonstrate that the modified U1 snRNA is able to reduce endogenous gene expression by limiting the polyadenylation of the targeted pre-mRNA transcript.


Subject(s)
Gene Expression Regulation , Polyadenylation , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , Animals , Cell Line , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit , Genetic Engineering , Mice , Mutagenesis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Osteocalcin/biosynthesis , Osteocalcin/genetics , RNA, Messenger/chemistry , Rats , Transcription Factors/biosynthesis , Transcription Factors/genetics
18.
Croat Med J ; 44(4): 407-11, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12950142

ABSTRACT

AIM: To study the effect of Dlx5 introduced by replication-competent avian splice-acceptor (RCAS) in mouse calvarial and bone marrow stromal cells, and to demonstrate that RCAS vector can be a useful system for studying gene expression in mammalian cells derived from Beta-AKE mouse. METHOD: Beta-AKE mouse used in experiments is a transgenic mouse line expressing the receptor for the Bryant polymerase subgroup A of RCAS vector (RCAS-BP(A) vector). Primary calvarial osteoblast cultures were obtained from 7-day-old Beta-AKE mice. Bone marrow stromal cells were derived from the long bones of 8-week-old Beta-AKE mice. Expression of genes cloned into RCAS vector in mouse cells was first established by detecting green fluorescent protein (GFP) in cells infected with RCAS-BP(A)-GFP sapphire by using fluorescence microscopy. Cells were then infected with RCAS-BP(A)-Dlx5 or RCAS-BP(A) alone as a control, for three days. After differentiation, cells were harvested for mRNA analysis at different time points (day 6 or 7, 11 or 12, 14 or 18, and 21 or 25). The cells were cultured in the presence of ascorbic acid and Beta-glycerophosphate, which promotes osteoblastic differentiation. RESULTS: Mouse calvarial and bone marrow stromal cells infected with RCAS-BP(A)-GFP sapphire were fluorescent compared with the controls. Both types of cells infected with RCAS-BP(A)Dlx5 consistently expressed increased levels of bone differentiation markers - type 1 collagen (Col1a1), osteocalcin, and bone sialoprotein mRNA. CONCLUSION: RCAS-BP(A) vector transduction of cells from Beta-AKE mice is a useful system for studying the role of gene expression in mouse osteoblastic cells. Dlx5 overexpression mediated by an RCAS-BP(A) vector stimulates mouse osteoblastic differentiation in Beta-AKE transgenic mice. Dlx5 induces osteoblast differentiation from bones formed either by endochondral or by membranous ossification.


Subject(s)
Cell Differentiation/genetics , Homeodomain Proteins/genetics , Osteoblasts/physiology , Retroviridae/genetics , Animals , Blotting, Northern , Bone Marrow Cells/physiology , Cells, Cultured , Gene Expression Regulation, Developmental , Genetic Vectors , In Situ Hybridization , Mice , Mice, Transgenic , RNA, Messenger/analysis , Sensitivity and Specificity
19.
Croat Med J ; 44(4): 412-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12950143

ABSTRACT

AIM: As quantitative and spatial analyses of promoter reporter constructs are not easily performed in intact bone, we designed a reporter gene specific to bone, which could be analyzed both visually and quantitatively by using chloramphenicol acetyltransferase (CAT) and a cyan version of green fluorescent protein (GFPcyan), driven by a 2.3-kb fragment of the rat collagen promoter (Col2.3). METHODS: The construct Col2.3CATiresGFPcyan was used for generating transgenic mice. Quantitative measurement of promoter activity was performed by CAT analysis of different tissues derived from transgenic animals; localization was performed by visualized GFP in frozen bone sections. To assess transgene expression during in vitro differentiation, marrow stromal cell and neonatal calvarial osteoblast cultures were analyzed for CAT and GFP activity. RESULTS: In mice, CAT activity was detected in the calvaria, long bone, teeth, and tendon, whereas histology showed that GFP expression was limited to osteoblasts and osteocytes. In cell culture, increased activity of CAT correlated with increased differentiation, and GFP activity was restricted to mineralized nodules. CONCLUSION: The concept of a dual reporter allows a simultaneous visual and quantitative analysis of transgene activity in bone.


Subject(s)
Cell Differentiation/genetics , Chloramphenicol O-Acetyltransferase/genetics , Osteoblasts/physiology , Promoter Regions, Genetic , Animals , Cells, Cultured , Gene Expression Regulation, Developmental , Genetic Markers , Mice , Mice, Transgenic , Osteoblasts/cytology , Sensitivity and Specificity , Transgenes/genetics
20.
J Bone Miner Res ; 17(6): 1008-14, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12054155

ABSTRACT

Our laboratory and others have shown that a homeodomain protein binding site plays an important role in transcription of the Collal gene in osteoblasts. This suggests that homeodomain proteins have an important role in osteoblast differentiation. We have investigated the role of Dlx5 in osteoblastic differentiation. In situ hybridization studies indicated that Dlx5 is expressed in chick calvarial osteoblasts (cCOB) in vivo. Northern blot analysis indicated that Dlx5 expression in cultured cCOBs is induced concurrently with osteoblastic markers. To study the effect of overexpression of Dlx5 on osteoblast differentiation, we infected primary osteoblast cultures from 15-day-old embryonal chicken calvaria with replication competent retroviral vectors [RCASBP(A)] expressing Dlx5 or control replication competent avian splice acceptor brianhightiter polymerase subtype A [RCASBP(A)]. Expression of Collal, osteopontin, alkaline phosphatase, and osteocalcin messenger RNA (mRNA) occurred sooner and at higher levels in cultures infected with RCASBP(A)DLX5 than in RCASBP(A)-infected cultures. Mineralization of Dlx5-expressing cultures was evident by days 12-14, and RCAS-infected control osteoblasts did not begin to mineralize until day 17. Dlx5 also stimulated osteoblastic differentiation of calvarial cells that do not normally undergo osteoblastic differentiation in vitro. Our results suggest that Dlx5 plays an important role in inducing calvarial osteoblast differentiation.


Subject(s)
Cell Differentiation/genetics , Homeodomain Proteins/genetics , Osteoblasts/cytology , Skull/metabolism , Animals , Blotting, Northern , Calcification, Physiologic , Chick Embryo , Collagen Type I/genetics , Genetic Vectors , Osteopontin , RNA, Messenger/genetics , Retroviridae/genetics , Sialoglycoproteins/genetics , Skull/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...