Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Hum Mol Genet ; 21(11): 2503-13, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22367969

ABSTRACT

The fibroblast growth factor receptor 3 (FGFR3) plays a critical role in the regulation of endochondral ossification. Fgfr3 gain-of-function mutations cause achondroplasia, the most common form of dwarfism, and a spectrum of chondrodysplasias. Despite a significant number of studies on the role of FGFR3 in cartilage, to date, none has investigated the influence of Fgfr3-mediated effects of the growth plate on bone formation. We studied three mouse models, each expressing Fgfr3 mutation either ubiquitously (CMV-Fgfr3(Y367C/+)), in chondrocytes (Col II-Fgfr3(Y367C/+)) or in mature osteoblasts (Col I-Fgfr3(Y367C/+)). Interestingly, we demonstrated that dwarfism with a significant defect in bone formation during growth was only observed in mouse models expressing mutant Fgfr3 in the cartilage. We observed a dramatic reduction in cartilage matrix mineralization and a strong defect of primary spongiosa. Anomalies of primary spongiosa were associated with an increase in osteoclast recruitment and a defect of osteoblasts at the mineralization front. A significant decrease in bone volume, trabecular thickness and number was also observed in the trabecular bone. Interestingly, no anomalies in proliferation and differentiation of primary osteoblasts from CMV-Fgfr3(Y367C/+) mice were observed. Based on these data, we excluded a potential function of Fgfr3 directly on osteoblasts at 3 weeks of age and we obtained evidence that the disorganization of the growth plate is responsible for the anomalies of the trabecular bone during bone formation. Herein, we propose that impaired FGFR3 signaling pathways may affect trabecular bone formation via a paracrine mechanism during growth. These results redefine our understanding of endochondral ossification in FGFR3-related chondrodysplasias.


Subject(s)
Mutation , Paracrine Communication/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Animals , Cell Proliferation , Chondrocytes/metabolism , Growth Plate/metabolism , Mice , Mice, Transgenic , Osteoblasts/metabolism , Osteogenesis/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism
2.
FASEB J ; 27(9): 3608-18, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23742809

ABSTRACT

In osteoclasts, Src controls podosome organization and bone degradation, which leads to an osteopetrotic phenotype in src(-/-) mice. Since this phenotype was even more severe in src(-/-)hck(-/-) mice, we examined the individual contribution of Hck in bone homeostasis. Compared to wt mice, hck(-/-) mice exhibited an osteopetrotic phenotype characterized by an increased density of trabecular bone and decreased bone degradation, although osteoclastogenesis was not impaired. Podosome organization and matrix degradation were found to be defective in hck(-/-) osteoclast precursors (preosteoclast) but were normal in mature hck(-/-) osteoclasts, probably through compensation by Src, which was specifically overexpressed in mature osteoclasts. As a consequence of podosome defects, the 3-dimensional migration of hck(-/-) preosteoclasts was strongly affected in vitro. In vivo, this translated by altered bone homing of preosteoclasts in hck(-/-) mice: in metatarsals of 1-wk-old mice, when bone formation strongly depends on the recruitment of these cells, reduced numbers of osteoclasts and abnormal developing trabecular bone were observed. This phenotype was still detectable in adults. In summmary, Hck is one of the very few effectors of preosteoclast recruitment described to date and thereby plays a critical role in bone remodeling.


Subject(s)
Bone and Bones/cytology , Bone and Bones/metabolism , Cell Movement/physiology , Osteoclasts/cytology , Osteopetrosis/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Animals , Cell Movement/genetics , Cells, Cultured , Female , Homeostasis/genetics , Homeostasis/physiology , Male , Mice , Mice, Knockout , Osteoclasts/metabolism , Osteopetrosis/genetics , Proto-Oncogene Proteins c-hck/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
3.
EMBO J ; 28(6): 686-96, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19197242

ABSTRACT

Bone undergoes continuous remodelling throughout adult life, and the equilibrium between bone formation by osteoblasts and bone resorption by osteoclasts defines the final bone mass. Here we show that Snail1 regulates this balance by controlling osteoblast differentiation. Snail1 is necessary for the early steps of osteoblast development, and it must be downregulated for their final differentiation. At the molecular level, Snail1 controls bone mass by repressing the transcription of both the osteoblast differentiation factor Runx2 and the vitamin D receptor (VDR) genes in osteoblasts. Sustained activation of Snail1 in transgenic mice provokes deficient osteoblast differentiation, which, together with the loss of vitamin D signalling in the bone, also impairs osteoclastogenesis. Indeed, the mineralisation of the bone matrix is severely affected, leading to hypocalcemia-independent osteomalacia. Our data show that the impact of Snail1 activity on the osteoblast population regulates the course of bone cells differentiation and ensures normal bone remodelling.


Subject(s)
Bone and Bones/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation , Osteoblasts/cytology , Receptors, Calcitriol/genetics , Transcription Factors/metabolism , Animals , Bone Remodeling , Bone and Bones/pathology , Calcification, Physiologic , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Fluoresceins/metabolism , Mice , Mice, Transgenic , Models, Biological , Organ Size , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteomalacia/genetics , Osteomalacia/physiopathology , Receptors, Calcitriol/metabolism , Snail Family Transcription Factors , Transcription, Genetic
4.
Blood ; 112(13): 5074-83, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18768394

ABSTRACT

Finding that activated T cells control osteoclast (OCL) differentiation has revealed the importance of the interactions between immune and bone cells. Dendritic cells (DCs) are responsible for T-cell activation and share common precursors with OCLs. Here we show that DCs participate in bone resorption more directly than simply through T-cell activation. We show that, among the splenic DC subsets, the conventional DCs have the higher osteoclastogenic potential in vitro. We demonstrate that conventional DCs differentiate into functional OCLs in vivo when injected into osteopetrotic oc/oc mice defective in OCL resorptive function. Moreover, this differentiation involves the presence of activated CD4(+) T cells controlling a high RANK-L expression by bone marrow stromal cells. Our results open new insights in the differentiation of OCLs and DCs and offer new basis for analyzing the relations between bone and immune systems.


Subject(s)
Bone Marrow/physiology , Cell Differentiation , Dendritic Cells/cytology , Osteoclasts/cytology , Stem Cell Niche/cytology , Animals , Bone Resorption , Dendritic Cells/immunology , Lymphocyte Activation , Mice , RANK Ligand/biosynthesis , Stromal Cells/metabolism , T-Lymphocytes
5.
J Cell Biol ; 164(4): 509-14, 2004 Feb 16.
Article in English | MEDLINE | ID: mdl-14970190

ABSTRACT

Amylin is a member of the calcitonin family of hormones cosecreted with insulin by pancreatic beta cells. Cell culture assays suggest that amylin could affect bone formation and bone resorption, this latter function after its binding to the calcitonin receptor (CALCR). Here we show that Amylin inactivation leads to a low bone mass due to an increase in bone resorption, whereas bone formation is unaffected. In vitro, amylin inhibits fusion of mononucleated osteoclast precursors into multinucleated osteoclasts in an ERK1/2-dependent manner. Although Amylin +/- mice like Amylin-deficient mice display a low bone mass phenotype and increased bone resorption, Calcr +/- mice display a high bone mass due to an increase in bone formation. Moreover, compound heterozygote mice for Calcr and Amylin inactivation displayed bone abnormalities observed in both Calcr +/- and Amylin +/- mice, thereby ruling out that amylin uses CALCR to inhibit osteoclastogenesis in vivo. Thus, amylin is a physiological regulator of bone resorption that acts through an unidentified receptor.


Subject(s)
Amyloid/metabolism , Bone Resorption , Osteogenesis/physiology , Receptors, Calcitonin/metabolism , Amyloid/genetics , Animals , Bone Density , Bone and Bones/abnormalities , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Differentiation/physiology , Islet Amyloid Polypeptide , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/cytology , Osteoclasts/physiology , Phenotype
6.
J Bone Miner Res ; 29(5): 1158-69, 2014.
Article in English | MEDLINE | ID: mdl-24127173

ABSTRACT

Receptor activator of NF-κB ligand (RANKL) plays a key role in osteoclast-induced bone resorption across a range of degenerative bone diseases, and its specific inhibition has been recently approved as a treatment for women with postmenopausal osteoporosis at high or increased risk of fracture in the United States and globally. In the present study, we generated transgenic mice (TghuRANKL) carrying the human RANKL (huRANKL) genomic region and achieved a physiologically relevant pattern of RANKL overexpression in order to establish novel genetic models for assessing skeletal and extraskeletal pathologies associated with excessive RANKL and for testing clinical therapeutic candidates that inhibit human RANKL. TghuRANKL mice of both sexes developed early-onset bone loss, and the levels of huRANKL expression were correlated with bone resorption and disease severity. Low copy Tg5516 mice expressing huRANKL at low levels displayed a mild osteoporotic phenotype as shown by trabecular bone loss and reduced biomechanical properties. Notably, overexpression of huRANKL, in the medium copy Tg5519 line, resulted in severe early-onset osteoporosis characterized by lack of trabecular bone, destruction of the growth plate, increased osteoclastogenesis, bone marrow adiposity, increased bone remodeling, and severe cortical bone porosity accompanied by decreased bone strength. An even more severe skeletal phenotype developed in the high copy Tg5520 founder with extensive soft tissue calcification. Model validation was further established by evidence that denosumab, an antibody that inhibits human but not murine RANKL, fully corrected the hyper-resorptive and osteoporotic phenotypes of Tg5519 mice. Furthermore, overexpression of huRANKL rescued osteopetrotic phenotypes of RANKL-defective mice. These novel huRANKL transgenic models of osteoporosis represent an important advance for understanding the pathogenesis and treatment of high-turnover bone diseases and other disease states caused by excessive RANKL.


Subject(s)
Gene Expression Regulation , Osteoporosis/genetics , Osteoporosis/metabolism , RANK Ligand/biosynthesis , Animals , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/pathology , Disease Models, Animal , Female , Growth Plate/metabolism , Growth Plate/pathology , Humans , Mice , Mice, Transgenic , Models, Genetic , Osteoclasts/pathology , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/pathology , Osteoporosis/pathology , RANK Ligand/genetics
7.
Autophagy ; 10(11): 1965-77, 2014.
Article in English | MEDLINE | ID: mdl-25484092

ABSTRACT

Bone remodeling is a tightly controlled mechanism in which osteoblasts (OB), the cells responsible for bone formation, osteoclasts (OC), the cells specialized for bone resorption, and osteocytes, the multifunctional mechanosensing cells embedded in the bone matrix, are the main actors. Increased oxidative stress in OB, the cells producing and mineralizing bone matrix, has been associated with osteoporosis development but the role of autophagy in OB has not yet been addressed. This is the goal of the present study. We first show that the autophagic process is induced in OB during mineralization. Then, using knockdown of autophagy-essential genes and OB-specific autophagy-deficient mice, we demonstrate that autophagy deficiency reduces mineralization capacity. Moreover, our data suggest that autophagic vacuoles could be used as vehicles in OB to secrete apatite crystals. In addition, autophagy-deficient OB exhibit increased oxidative stress and secretion of the receptor activator of NFKB1 (TNFSF11/RANKL), favoring generation of OC, the cells specialized in bone resorption. In vivo, we observed a 50% reduction in trabecular bone mass in OB-specific autophagy-deficient mice. Taken together, our results show for the first time that autophagy in OB is involved both in the mineralization process and in bone homeostasis. These findings are of importance for mineralized tissues which extend from corals to vertebrates and uncover new therapeutic targets for calcified tissue-related metabolic pathologies.


Subject(s)
Autophagy , Bone and Bones/metabolism , Osteoblasts/cytology , Animals , Bone Remodeling , Bone Resorption , Cell Line, Tumor , Female , Green Fluorescent Proteins/metabolism , Homeostasis , Mice , Mice, Transgenic , Microscopy, Confocal , NF-kappa B p50 Subunit/metabolism , Osteoclasts/metabolism , Oxidative Stress , RANK Ligand/metabolism , Rats , X-Ray Microtomography
8.
Genome Biol ; 14(7): R82, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902802

ABSTRACT

BACKGROUND: The mouse inbred line C57BL/6J is widely used in mouse genetics and its genome has been incorporated into many genetic reference populations. More recently large initiatives such as the International Knockout Mouse Consortium (IKMC) are using the C57BL/6N mouse strain to generate null alleles for all mouse genes. Hence both strains are now widely used in mouse genetics studies. Here we perform a comprehensive genomic and phenotypic analysis of the two strains to identify differences that may influence their underlying genetic mechanisms. RESULTS: We undertake genome sequence comparisons of C57BL/6J and C57BL/6N to identify SNPs, indels and structural variants, with a focus on identifying all coding variants. We annotate 34 SNPs and 2 indels that distinguish C57BL/6J and C57BL/6N coding sequences, as well as 15 structural variants that overlap a gene. In parallel we assess the comparative phenotypes of the two inbred lines utilizing the EMPReSSslim phenotyping pipeline, a broad based assessment encompassing diverse biological systems. We perform additional secondary phenotyping assessments to explore other phenotype domains and to elaborate phenotype differences identified in the primary assessment. We uncover significant phenotypic differences between the two lines, replicated across multiple centers, in a number of physiological, biochemical and behavioral systems. CONCLUSIONS: Comparison of C57BL/6J and C57BL/6N demonstrates a range of phenotypic differences that have the potential to impact upon penetrance and expressivity of mutational effects in these strains. Moreover, the sequence variants we identify provide a set of candidate genes for the phenotypic differences observed between the two strains.


Subject(s)
Genome/genetics , Animals , Behavior, Animal , Disease Resistance/immunology , Eye/pathology , Female , Femur/diagnostic imaging , Hypersensitivity/immunology , INDEL Mutation/genetics , Killer Cells, Natural/immunology , Listeriosis/immunology , Listeriosis/microbiology , Male , Maze Learning , Mice, Inbred C57BL , Phenotype , Polymorphism, Single Nucleotide/genetics , Spleen/immunology , X-Ray Microtomography
9.
PLoS One ; 6(10): e26627, 2011.
Article in English | MEDLINE | ID: mdl-22046317

ABSTRACT

Osteoporosis is one of the most common bone pathologies, which are characterized by a decrease in bone mass. It is well established that bone mass, which results from a balanced bone formation and bone resorption, is regulated by many hormonal, environmental and genetic factors. Here we report that the immune semaphorin 4D (Sema4D) is a novel factor controlling bone resorption. Sema4D-deficient primary osteoclasts showed impaired spreading, adhesion, migration and resorption due to altered ß3 integrin sub-unit downstream signaling. In apparent accordance with these in vitro results, Sema4D deletion in sexually mature female mice led to a high bone mass phenotype due to defective bone resorption by osteoclasts. Mutant males, however, displayed normal bone mass and the female osteopetrotic phenotype was only detected at the onset of sexual maturity, indicating that, in vivo, this intrinsic osteoclast defect might be overcome in these mice. Using bone marrow cross transplantation, we confirmed that Sema4D controls bone resorption through an indirect mechanism. In addition, we show that Sema4D -/- mice were less fertile than their WT littermates. A decrease in Gnrh1 hypothalamic expression and a reduced number of ovarian follicles can explain this attenuated fertility. Interestingly, ovariectomy abrogated the bone resorption phenotype in Sema4D -/- mice, providing the evidence that the observed high bone mass phenotype is strictly dependent on ovarian function. Altogether, this study reveals that, in vivo, Sema4D is an indirect regulator of bone resorption, which acts via its effect on reproductive function.


Subject(s)
Antigens, CD/physiology , Bone Resorption , Ovary/physiology , Semaphorins/physiology , Animals , Female , Male , Mice , Osteoclasts/cytology , Osteoclasts/physiology , Ovarian Function Tests , Semaphorins/deficiency , Sex Factors
10.
J Bone Miner Res ; 26(9): 2036-44, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21594896

ABSTRACT

Excess thyroid hormone (TH) in adults causes osteoporosis and increases fracture risk. However, the mechanisms by which TH affects bone turnover are not elucidated. In particular, the roles of thyroid hormone receptor (TR) isotypes in the mediation of TH effects on osteoblast-mediated bone formation and osteoclast-mediated bone resorption are not established. In this study we have induced experimental hypothyroidism or hyperthyroidism in adult wild-type, TRα- or TRß-deficient mice and analyzed the effects of TH status on the structure and remodeling parameters of trabecular bone. In wild-type mice, excess TH decreased bone volume and mineralization. High TH concentrations were associated with a high bone-resorption activity, assessed by increased osteoclast surfaces and elevated concentrations of serum bone-resorption markers. Serum markers of bone formation also were higher in TH-treated mice. TRα deficiency did not prevent TH action on bone volume, bone mineralization, bone formation, or bone resorption. In contrast, TRß deficiency blocked all the early effects of excess TH observed in wild-type mice. However, prolonged exposure to low or high TH concentrations of TRß-deficient mice induced mild modifications of bone structure and remodeling parameters. Together our data suggest that TRß receptors mediate the acute effects produced by transient changes of TH concentrations on bone remodeling, whereas TRα receptors mediate long-term effects of chronic alterations of TH metabolism. These data shed new light on the respective roles of TRs in the control of bone metabolism by TH.


Subject(s)
Bone Remodeling/drug effects , Bone and Bones/pathology , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormones/pharmacology , Aging/drug effects , Aging/metabolism , Aging/pathology , Animals , Bone Density/drug effects , Bone Resorption/blood , Bone Resorption/complications , Bone Resorption/pathology , Bone Resorption/physiopathology , Bone and Bones/drug effects , Hyperthyroxinemia/blood , Hyperthyroxinemia/complications , Hyperthyroxinemia/pathology , Hyperthyroxinemia/physiopathology , Mice , Mice, Knockout , Organ Size/drug effects , Osteogenesis/drug effects , Thyroid Hormone Receptors alpha/deficiency , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/deficiency , Triiodothyronine/blood
11.
PLoS One ; 2(7): e585, 2007 Jul 04.
Article in English | MEDLINE | ID: mdl-17611620

ABSTRACT

Bone resorption by osteoclasts is necessary to maintain bone homeostasis. Osteoclast differentiation from hematopoietic progenitors and their activation depend on M-CSF and RANKL, but also requires co-stimulatory signals acting through receptors associated with DAP12 and FcRgamma adaptors. Dap12 mutant mice (KDelta75) are osteopetrotic due to inactive osteoclasts but, surprisingly, these mice are more sensitive than WT mice to bone loss following an ovariectomy. Because estrogen withdrawal is known to disturb bone mass, at least in part, through lymphocyte interaction, we looked at the role of mature lymphocytes on osteoclastogenesis and bone mass in the absence of functional DAP12. Lymphocytes were found to stimulate an early osteoclast differentiation response from Dap12-deficient progenitors in vitro. In vivo, Rag1-/- mice lacking mature lymphocytes did not exhibit any bone phenotype, but lost their bone mass after ovariectomy like KDelta75 mice. KDelta75;Rag1-/- double mutant female mice exhibited a more severe osteopetrosis than Dap12-deficient animals but lost their bone mass after ovariectomy, like single mutants. These results suggest that both DAP12 and mature lymphocytes act synergistically to maintain bone mass under physiological conditions, while playing similar but not synergistic co-stimulatory roles in protecting bone loss after gonadal failure. Thus, our data support a role for lymphocytes during osteoclast differentiation and suggest that they may function as accessory cells when regular osteoclast function is compromised.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Lymphocytes/physiology , Osteoclasts/physiology , Ovariectomy , Adaptor Proteins, Signal Transducing/deficiency , Animals , Bone Resorption/genetics , Bone Resorption/physiopathology , Bone and Bones/anatomy & histology , Cell Differentiation , Female , Macrophage Colony-Stimulating Factor/physiology , Mice , Mice, Knockout , Mice, Mutant Strains , Organ Size , Osteoclasts/cytology , Osteogenesis , Osteopetrosis/genetics , Osteopetrosis/prevention & control , RANK Ligand/physiology , Uterus/anatomy & histology
12.
Cell ; 130(3): 456-69, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17693256

ABSTRACT

The regulation of bone remodeling by an adipocyte-derived hormone implies that bone may exert a feedback control of energy homeostasis. To test this hypothesis we looked for genes expressed in osteoblasts, encoding signaling molecules and affecting energy metabolism. We show here that mice lacking the protein tyrosine phosphatase OST-PTP are hypoglycemic and are protected from obesity and glucose intolerance because of an increase in beta-cell proliferation, insulin secretion, and insulin sensitivity. In contrast, mice lacking the osteoblast-secreted molecule osteocalcin display decreased beta-cell proliferation, glucose intolerance, and insulin resistance. Removing one Osteocalcin allele from OST-PTP-deficient mice corrects their metabolic phenotype. Ex vivo, osteocalcin can stimulate CyclinD1 and Insulin expression in beta-cells and Adiponectin, an insulin-sensitizing adipokine, in adipocytes; in vivo osteocalcin can improve glucose tolerance. By revealing that the skeleton exerts an endocrine regulation of sugar homeostasis this study expands the biological importance of this organ and our understanding of energy metabolism.


Subject(s)
Bone and Bones/physiology , Energy Metabolism/physiology , Glucose/physiology , Insulin/physiology , Obesity/metabolism , Animals , Bone and Bones/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Genes, Lethal , Glucose Intolerance/enzymology , Glucose Intolerance/genetics , Glucose Intolerance/prevention & control , Hypoglycemia/enzymology , Hypoglycemia/genetics , Hypoglycemia/prevention & control , Insulin Resistance/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Animal , Obesity/genetics , Obesity/prevention & control , Protein Tyrosine Phosphatases/deficiency , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3
13.
Dev Dyn ; 224(2): 245-51, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12112477

ABSTRACT

Cell- and time-specific gene inactivation should enhance our knowledge of bone biology. Implementation of this technique requires construction of transgenic mouse lines expressing Cre recombinase in osteoblasts, the bone forming cell. We tested several promoter fragments for their ability to drive efficient Cre expression in osteoblasts. In the first mouse transgenic line, the Cre gene was placed under the control of the 2.3-kb proximal fragment of the alpha1(I)-collagen promoter, which is expressed at high levels in osteoblasts throughout their differentiation. Transgenic mice expressing this transgene in bone were bred with the ROSA26 reporter (R26R) strain in which the ROSA26 locus is targeted with a conditional LacZ reporter cassette. In R26R mice, Cre expression and subsequent Cre-mediated recombination lead to expression of the LacZ reporter gene, an event that can be monitored by LacZ staining. LacZ staining was detected in virtually all osteoblasts of alpha1(I)-Cre;R26R mice indicating that homologous recombination occurred in these cells. No other cell type stained blue. In the second line studied, the 1.3-kb fragment of osteocalcin gene 2 (OG2) promoter, which is active in differentiated osteoblasts, was used to drive Cre expression. OG2-Cre mice expressed Cre specifically in bone. However, cross of OG2-Cre mice with R26R mice did not lead to any detectable LacZ staining in osteoblasts. Lastly, we tested a more active artificial promoter derived from the OG2 promoter. The artificial OG2-Cre transgene was expressed by reverse transcriptase-polymerase chain reaction in cartilage and bone samples. After cross of the artificial OG2-Cre mice with R26R mice, we detected a LacZ staining in articular chondrocytes but not in osteoblasts. Our data suggest that the only promoter able to drive Cre expression at a level sufficient to induce recombination in osteoblasts is the alpha1(I)-collagen promoter.


Subject(s)
Collagen Type I/genetics , Gene Transfer Techniques , Integrases/genetics , Promoter Regions, Genetic , Viral Proteins/genetics , Animals , Cells, Cultured , Genes, Reporter , Integrases/metabolism , Lac Operon , Mice , Mice, Transgenic , Models, Genetic , Osteoblasts/metabolism , Plasmids/metabolism , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transfection , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL