Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Bone ; 162: 116476, 2022 09.
Article in English | MEDLINE | ID: mdl-35760307

ABSTRACT

Lateral Meningocele or Lehman Syndrome (LMS) is associated with NOTCH3 mutations causing deletions of the PEST domain and a gain-of-NOTCH3 function. We demonstrated that Notch3em1Ecan mice harboring Notch3 mutations analogous to those found in LMS are osteopenic because of enhanced bone resorption. To determine the contribution of specific cell lineages to the phenotype, we created a conditional-by-inversion (Notch3COIN) model termed Notch3em2Ecan in which Cre recombination generates a Notch3INV allele expressing a NOTCH3 mutant lacking the PEST domain. Germ line Notch3COIN inversion caused osteopenia and phenocopied the Notch3em1Ecan mutant, validating the model. To induce the mutation in osteocytes, smooth muscle and endothelial cells, Notch3COIN mice were bred with mice expressing Cre from the Dmp1, Sm22a and Cdh5 promoters, respectively, creating experimental mice harboring Notch3INV alleles in Cre-expressing cells and control littermates harboring Notch3COIN alleles. Notch3COIN inversion in osteocytes led to femoral and vertebral cancellous bone osteopenia, whereas Notch3COIN inversion in mural Sm22a or endothelial Cdh5-expressing cells did not result in a skeletal phenotype. In conclusion, introduction of the LMS mutation in osteocytes but not in vascular cells causes osteopenia and phenocopies Notch3em1Ecan global mutant mice.


Subject(s)
Bone Diseases, Metabolic , Meningocele , Abnormalities, Multiple , Animals , Bone Diseases, Metabolic/metabolism , Endothelial Cells/metabolism , Male , Meningocele/complications , Meningocele/genetics , Meningocele/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Osteocytes/metabolism , Receptors, Notch/metabolism
2.
Article in English | MEDLINE | ID: mdl-35602658

ABSTRACT

Towards the transition to blended and remote education, evaluating the levels of students' digital competence and designing educational programs to advance them is of paramount importance. Existing validated digital competence scales usually ignore either important digital skills needed or new socio-technological innovations. This study proposes and validates a comprehensive digital competence scale for students in higher education. The suggested instrument includes skills of online learning and collaboration, social media, smart and mobile devices, safety, and data protection. The scale was evaluated on a sample of 156 undergraduate and postgraduate students just before and at the beginning of the COVID-19 crisis. The final scale is composed of 28 items and six digital competence components. The evaluation study revealed valid results in terms of model fit criteria, factor loadings, internal validity, and reliability. Individual factors like the students' field of study, computer experience and age revealed significant associations to the scale components, while gender revealed no significant differences. The suggested scale can be useful to the design of new actions and policies towards remote education and the digital skills' development of adult learners.

3.
J Bioeth Inq ; 17(4): 743-748, 2020 12.
Article in English | MEDLINE | ID: mdl-33169265

ABSTRACT

Consumer involvement in clinical research is an essential component of a comprehensive response during emergent health challenges. During the COVID-19 pandemic, the moderation of research policies and regulation to facilitate research may raise ethical issues. Meaningful, diverse consumer involvement can help to identify practical approaches to prioritize, design, and conduct rapidly developed clinical research amid current events. Consumer involvement might also elucidate the acceptability of flexible ethics review approaches that aim to protect participants whilst being sensitive to the challenging context in which research is taking place. This article describes the main ethical challenges arising from pandemic research and how involving consumers and the community could enable resolution of such issues.


Subject(s)
COVID-19 , Community Participation , Ethics, Research , Humans , Pandemics , SARS-CoV-2
4.
J Dent Res ; 97(7): 835-843, 2018 07.
Article in English | MEDLINE | ID: mdl-29489425

ABSTRACT

Formation of highly organized dental hard tissues is a complex process involving sequential and ordered deposition of an extracellular scaffold, followed by its mineralization. Odontoblast and ameloblast differentiation involves reciprocal and sequential epithelial-mesenchymal interactions. Similar to early tooth development, various Bmps are expressed during this process, although their functions have not been explored in detail. Here, we investigated the role of odontoblast-derived Bmp2 for tooth mineralization using Bmp2 conditional knockout mice. In developing molars, Bmp2LacZ reporter mice revealed restricted expression of Bmp2 in early polarized and functional odontoblasts while it was not expressed in mature odontoblasts. Loss of Bmp2 in neural crest cells, which includes all dental mesenchyme, caused a delay in dentin and enamel deposition. Immunohistochemistry for nestin and dentin sialoprotein (Dsp) revealed polarization defects in odontoblasts, indicative of a role for Bmp2 in odontoblast organization. Surprisingly, pSmad1/5/8, an indicator of Bmp signaling, was predominantly reduced in ameloblasts, with reduced expression of amelogenin ( Amlx), ameloblastin ( Ambn), and matrix metalloproteinase ( Mmp20). Quantitative real-time polymerase chain reaction (RT-qPCR) analysis and immunohistochemistry showed that loss of Bmp2 resulted in increased expression of the Wnt antagonists dickkopf 1 ( Dkk1) in the epithelium and sclerostin ( Sost) in mesenchyme and epithelium. Odontoblasts showed reduced Wnt signaling, which is important for odontoblast differentiation, and a strong reduction in dentin sialophosphoprotein ( Dspp) but not collagen 1 a1 ( Col1a1) expression. Mature Bmp2-deficient teeth, which were obtained by transplanting tooth germs from Bmp2-deficient embryos under a kidney capsule, showed a dentinogenesis imperfecta type II-like appearance. Micro-computed tomography and scanning electron microscopy revealed reduced dentin and enamel thickness, indistinguishable primary and secondary dentin, and deposition of ectopic osteodentin. This establishes that Bmp2 provides an early temporal, nonredundant signal for directed and organized tooth mineralization.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Odontoblasts/metabolism , Tooth Calcification/physiology , Amelogenin/metabolism , Animals , Dental Enamel Proteins/metabolism , Dentinogenesis Imperfecta/metabolism , Dentinogenesis Imperfecta/physiopathology , Extracellular Matrix Proteins/metabolism , Immunohistochemistry , Matrix Metalloproteinase 20/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Molar/metabolism , Nestin/metabolism , Phosphoproteins/metabolism , Real-Time Polymerase Chain Reaction , Sialoglycoproteins/metabolism , Signal Transduction , Smad Proteins/metabolism , X-Ray Microtomography
5.
Gene Ther ; 19(5): 483-93, 2012 May.
Article in English | MEDLINE | ID: mdl-21918547

ABSTRACT

Intraventricular ependymal infection by adenoviruses expressing brain-derived neurotrophic factor (BDNF) and noggin is sufficient to induce the heterotopic recruitment of new medium spiny neurons to the adult neostriatum, from endogenous subependymal neural progenitor cells. This approach was found to slow disease progression and extend survival in an R6/2 mouse model of Huntington's disease (HD). However, the practical therapeutic value of this strategy is limited by the transient expression and immunogenicity of adenoviral vectors. In addition, it has been unclear whether sustained overexpression of BDNF and noggin would yield similarly sustained neuronal production and striatal recruitment, or whether progenitor depletion or tachyphylaxis might supervene to limit the therapeutic potential of this approach. To address these issues, we used adeno-associated virus serotype 4 (AAV4), an ependymotrophic vector that is neither immunogenic nor neurotoxic, to achieve sustained BDNF and noggin expression. Using AAV4, we found that BDNF and noggin achieved levels sufficient to initiate and maintain, for at least 4 months, ongoing neuronal addition to the neostriatum and olfactory bulb. Over this period, we noted no diminution of treatment-associated neuronal recruitment from resident progenitors. AAV4:BDNF and noggin-induced neuronal addition may thus provide a means to provide longlasting and persistent striatal neuronal replacement in conditions of striatal neuronal loss, such as HD.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Carrier Proteins/genetics , Neostriatum/metabolism , Neurogenesis , Animals , Animals, Genetically Modified , Corpus Striatum/cytology , Dependovirus/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Transfer Techniques , Genetic Vectors , Neostriatum/cytology , Olfactory Bulb/metabolism , Rats
7.
Kidney Int ; 73(2): 181-91, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17943079

ABSTRACT

Once developed, end-stage renal disease cannot be reversed by any current therapy. Bone morphogenetic protein-7 (BMP-7), however, is a possible treatment for reversing end-stage renal disease. Previously, we showed that the BMP antagonist uterine sensitization-associated gene-1 (USAG-1, also known as ectodin and sclerostin domain-containing 1) negatively regulates the renoprotective action of BMP-7. Here, we show that the ratio between USAG-1 and BMP-7 expression increased dramatically in the later stage of kidney development, with USAG-1 expression overlapping BMP-7 only in differentiated distal tubules. Examination of USAG-1 expression in developing kidney indicated that a mosaic of proximal and distal tubule marker-positive cells reside side by side in the immature nephron. This suggests that each cell controls its own fate for becoming a proximal or distal tubule cell. In kidney injury models, the ratio of USAG-1 to BMP-7 expression decreased with kidney damage but increased after subsequent kidney regeneration. Our study suggests that USAG-1 expression in a kidney biopsy could be useful in predicting outcome.


Subject(s)
Bone Morphogenetic Proteins/analysis , Kidney Tubules/chemistry , Kidney Tubules/embryology , Transforming Growth Factor beta/analysis , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Cell Differentiation , Cisplatin/toxicity , Female , Kidney Tubules/drug effects , Mice , Mice, Inbred C57BL , Nephrons/chemistry , Prognosis , Regeneration , Transforming Growth Factor beta/genetics
8.
Connect Tissue Res ; 45(1): 50-9, 2004.
Article in English | MEDLINE | ID: mdl-15203940

ABSTRACT

Bone morphogenetic proteins (BMPs) are known to promote periodontal tissue regeneration, while noggin inhibits the biological activities of BMP-2, -4, and -7. To investigate the effect of BMPs and noggin gene transfer on cementogenesis, we used cloned murine cementoblasts (OCCM). Cells were transduced using adenoviruses encoding BMP-7 (Ad-BMP-7), noggin devoid of the heparin binding site (Ad-NOGDeltaB2), or a control adenovirus encoding green fluorescent protein (Ad-GFP). Cells were seeded into 3D polymer scaffolds and implanted into SCID mice to determine the in vivo mineral-inducing ability of the cells. Cells transduced with Ad-NOGDeltaB2 at 3 and 6 weeks postimplantation exhibited reduced mineral formation compared with all other groups. Although gene expression of osteocalcin and bone sialoprotein increased after Ad-BMP-7 transduction in vitro, following BMP-7 gene transfer in vivo, transcripts for OCN and BSP were not significantly different from controls, and mineral density was not significantly increased compared with Ad-GFP and NT groups. These results indicate that in mature cementoblast populations, gene transfer of noggin inhibits biomineralization induced by cementoblasts, whereas exogenous BMP has minimal effects on mineralization.


Subject(s)
Cementogenesis/drug effects , Dental Cementum/physiology , Minerals/antagonists & inhibitors , Proteins/pharmacology , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins , Cell Differentiation/drug effects , Cell Line, Transformed , Clone Cells , Dental Cementum/cytology , Gene Transfer Techniques , Mice , Mice, SCID , Minerals/metabolism , Proteins/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
9.
Endocrinology ; 144(5): 1972-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12697704

ABSTRACT

Skeletal cells synthesize bone morphogenetic proteins (BMPs) and BMP antagonists. Noggin is a glycoprotein that binds BMPs selectively and antagonizes BMP actions. Noggin expression in osteoblasts is induced by BMPs and noggin opposes the effects of BMPs on osteoblastic differentiation and function in vitro. However, its effects in vivo are not known. We investigated the direct in vivo effects of noggin on bone remodeling in transgenic mice overexpressing noggin under the control of the osteocalcin promoter. Noggin transgenics suffered long bone fractures in the first month of life. Total, vertebral, and femoral bone mineral densities were reduced by 23-29%. Static and dynamic histomorphometry of the femur revealed that noggin transgenic mice had decreased trabecular bone volume, number of trabeculae, and bone formation rate. Osteoblast surface and number of osteoblasts/trabecular area were not significantly decreased, indicating impaired osteoblastic function. Osteoclast surface and number were normal/decreased, there was no increase in bone resorption, and the tissue had the appearance of woven bone. Vertebral microcomputed tomography scanning confirmed decreased trabecular bone volume and trabecular number. In conclusion, transgenic mice overexpressing noggin in the bone microenvironment have decreased trabecular bone volume and impaired osteoblastic function, leading to osteopenia and fractures.


Subject(s)
Bone Diseases, Metabolic/etiology , Bone and Bones/metabolism , Osteogenesis/physiology , Proteins/metabolism , Animals , Body Weight , Bone Density , Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/pathology , Bone Remodeling/physiology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Carrier Proteins , Mice , Mice, Transgenic/genetics , Proteins/genetics , Reference Values , Tomography, X-Ray Computed
10.
Bone ; 32(2): 111-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12633782

ABSTRACT

Noggin is a glycoprotein that binds bone morphogenetic proteins (BMPs) selectively and, when added to osteoblasts, it opposes the effects of BMPs. However, the consequences of its continued expression in stromal cells are not known. We investigated the effects of noggin overexpression under the control of a constitutive promoter, on murine ST-2 stromal cells, and its impact on stromal cells from transgenic mice overexpressing noggin under the control of the osteocalcin promoter. ST-2 cells were transduced with a retroviral vector (pLPCX) or a vector driving noggin (pLPCX noggin). Untreated (pLPCX) ST-2 cells developed the appearance of mineralized nodules and expressed osteocalcin. pLPCX noggin delayed the appearance of mineralized nodules and prevented the expression of osteocalcin. Noggin also prevented the cortisol-dependent induction of peroxisome proliferator-activated receptor gamma2 and adipsin transcripts, indicating a generalized inhibition of cell differentiation. Primary stromal cells from noggin transgenic mice displayed impaired differentiation when compared to cells from wild-type animals and did not express osteocalcin mRNA. In conclusion, noggin arrests the differentiation of stromal cells, preventing cellular maturation.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Proteins/physiology , Animals , Apoptosis/physiology , Carrier Proteins , Cell Differentiation/physiology , Cells, Cultured , Humans , Mice , Protein Biosynthesis , Proteins/genetics , Stromal Cells/cytology , Stromal Cells/metabolism
11.
Article in English | MEDLINE | ID: mdl-18244879

ABSTRACT

We present the STack ARchitecture (STAR) automaton. It is a fixed structure, multiaction, reward-penalty learning automaton, characterized by a star-shaped state transition diagram. Each branch of the star contains D states associated with a particular action. The branches are connected to a central "neutral" state. The most general version of STAR involves probabilistic state transitions in response to reward and/or penalty, but deterministic transitions can also be used. The learning behavior of STAR results from the stack-like operation of the branches; the learning parameter is D. By mathematical analysis, it is shown that STAR with deterministic reward/probabilistic penalty and a sufficiently large D can be rendered /spl epsi/-optimal in every stationary environment. By numerical simulation it is shown that in nonstationary, switching environments, STAR usually outperforms classical variable structure automata such as L/sub R-P/, L/sub R-I/, and L/sub R-/spl epsi/P/.

12.
Clin Sci (Lond) ; 101(4): 367-76, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11566074

ABSTRACT

This study examines the electrophysiological and metabolic changes that occur in rabbit hearts during hypothermic storage in vitro. Hearts were microperfused at 4 degrees C for 6 or 24 h with either normal Krebs-Henseleit buffer (KHB) or KHB containing 2,3-butanedione monoxime (BDM). After hypothermic storage, hearts were rewarmed to 37 degrees C with KHB. Cardiac function was then assessed in Langendorff perfusion mode. Electrophysiological changes were also assessed from the ventricular paced-evoked responses. After storage, mitochondria were isolated from the hearts and their respiratory control ratio, rate of ATP synthesis and outer membrane intactness were assessed. Compared with values from fresh non-stored hearts, hearts stored hypothermically for 24 h showed significant decreases in both left ventricular developed pressure and coronary flow when reperfused in Langendorff mode. On the other hand, the decrease in left ventricular developed pressure in hearts that were stored for only 6 h (with or without BDM) was not significant. Compared with values obtained from fresh non-stored hearts, hypothermic storage significantly decreased the R-wave amplitude, and both the R-E and ST-E intervals of paced-evoked responses. This was true for hearts microperfused for 6 h (with or without BDM) and for hearts microperfused with buffer containing BDM for 24 h. The ST-R intervals in hearts microperfused hypothermically for 6 h were prolonged, but this change was not statistically significant compared with those obtained from unstored hearts. In hearts microperfused with KHB containing BDM for 24 h, the ST-R interval was significantly prolonged. Hypothermic microperfusion for 24 h significantly decreased both the mitochondrial coupling ratio and the rate of ATP synthesis. In hearts microperfused with BDM for 6 h, mitochondrial coupling ratios and the rate of ATP synthesis were not significantly different from those in fresh hearts. In conclusion, the present study has shown that long-term hypothermic storage significantly impaired both paced-evoked responses and mitochondrial function. Inclusion of BDM in the perfusion buffer during storage significantly ameliorated some of these changes.


Subject(s)
Diacetyl/analogs & derivatives , Heart/physiology , Organ Preservation/methods , Refrigeration , Adenosine Triphosphate/biosynthesis , Animals , Coronary Circulation , Diacetyl/pharmacology , Electrophysiology , Glucose/pharmacology , Heart/drug effects , Mitochondria, Heart/physiology , Organ Preservation Solutions/pharmacology , Rabbits , Rewarming , Time Factors , Tromethamine/pharmacology , Ventricular Function, Left , Ventricular Pressure
13.
Proc Natl Acad Sci U S A ; 98(20): 11353-8, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562478

ABSTRACT

Secreted noggin protein regulates bone morphogenetic protein activity during development. In mice, a complete loss of noggin protein leads to multiple malformations including joint fusion, whereas mice heterozygous for Nog loss-of-function mutations are normal. In humans, heterozygous NOG missense mutations have been found in patients with two autosomal dominant disorders of joint development, multiple synostosis syndrome (SYNS1) and a milder disorder proximal symphalangism (SYM1). This study investigated the effect of one SYNS1 and two SYM1 disease-causing missense mutations on the structure and function of noggin. The SYNS1 mutation abolished, and the SYM1 mutations reduced, the secretion of functional noggin dimers in transiently transfected COS-7 cells. Coexpression of mutant noggin with wild-type noggin, to resemble the heterozygous state, did not interfere with wild-type noggin secretion. These data indicate that the human disease-causing mutations are hypomorphic alleles that reduce secretion of functional dimeric noggin. Therefore, we conclude that noggin has both species-specific and joint-specific dosage-dependent roles during joint formation. Surprisingly, in contrast to the COS-7 cell studies, the SYNS1 mutant was able to form dimers in Xenopus laevis oocytes. This finding indicates that there also exist species-specific differences in the ability to process mutant noggin polypeptides.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Mutation, Missense , Proteins/genetics , Proteins/metabolism , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , COS Cells , Carrier Proteins , Chlorocebus aethiops , Dimerization , Disulfides , Female , Humans , Oocytes/physiology , Protein Biosynthesis , Recombinant Proteins/metabolism , Synostosis/genetics , Transfection , Xenopus laevis
14.
Medicine (Baltimore) ; 80(2): 123-33, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11307588

ABSTRACT

Immunodeficiency with thymoma (Good syndrome, GS) is a rare, adult-onset condition that is characterized by thymoma, hypogammaglobulinemia, and low numbers of peripheral B cells. CD4+ T lymphopenia and an inverted CD4:CD8+ T-cell ratio may be present. Here we report 5 patients with GS and infectious complications who were seen at 3 institutions between 1983 and 1999. Three patients had recurrent sinopulmonary infections, 3 had severe cytomegalovirus (CMV) disease, and 1 had Pneumocystis carinii pneumonia. Review of the literature identified 46 other reports of infections in GS patients. The infections reported in all 51 patients included recurrent sinopulmonary infection (19 cases with documented respiratory pathogens), generally with encapsulated bacteria, most often Haemophilus influenzae (11 cases); CMV disease (5 cases); bacteremia (7 cases); oral or esophageal candidiasis (6 cases); persistent mucocutaneous candidiasis (5 cases); chronic diarrhea (5 cases with documented stool pathogens); urinary tract infections (4 cases); P. carinii pneumonia (3 cases); tuberculosis (2 cases); Kaposi sarcoma (1 case); disseminated varicella (1 case); candidemia (1 case); wound infection with Clostridium perfringens (1 case); Mycoplasma arthritis (1 case); and other infections. Patients with GS present with a spectrum of sinopulmonary infections and pathogens similar to common variable immunodeficiency (CVID). Compared with patients with CVID, opportunistic infections, including severe CMV disease, P. carinii pneumonia, and mucocutaneous candidiasis, appear to be more common in patients with GS, and patients with GS may have a worse prognosis. GS should be ruled out in patients with thymoma or CVID who develop severe, especially opportunistic, infections. Treatment with intravenous immune globulin is recommended for all patients with GS.


Subject(s)
Agammaglobulinemia/complications , Opportunistic Infections/etiology , Thymoma/complications , Thymus Neoplasms/complications , Agammaglobulinemia/diagnosis , Agammaglobulinemia/etiology , Agammaglobulinemia/therapy , Aged , Female , Humans , Immunoglobulins, Intravenous/therapeutic use , Male , Middle Aged , Opportunistic Infections/diagnosis , Opportunistic Infections/therapy , Recurrence , Thymoma/diagnosis , Thymoma/therapy , Thymus Neoplasms/diagnosis , Thymus Neoplasms/therapy , Treatment Outcome
15.
J Bone Miner Res ; 16(3): 497-500, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11277267

ABSTRACT

Bone morphogenetic proteins (BMPs) are expressed and secreted during fracture repair. Although they are likely to be required for this process, little is known about their physiological role in bone regeneration. Noggin is a protein that specifically binds and inactivates several BMPs. It plays fundamental roles during early embryonal development and limb morphogenesis by this BMP-inactivating activity. This study shows that Noggin can modify bone formation in vivo in the adult animal and, thus, indirectly, that BMP signaling is indispensable in this process. A noggin mutein (hNgdeltaB2-Fc) engineered so as to display increased bioavailability was used. Bilateral titanium bone chambers were inserted in 70 rats, and side comparisons for bone formation in the chambers were done. The hNgdeltaB2-Fc had no effect on total amount of tissue formed in the chamber but decreased the amount of bone compared with both buffer controls and a control made up of an Fc-tagged IL-6Ralpha protein, which had no effects of its own. Also, wild-type noggin inhibited bone formation. Thus, endogenous BMP signaling is necessary for normal bone regeneration.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Osteogenesis/physiology , Proteins/metabolism , Animals , Bone Regeneration , Carrier Proteins , Proteins/genetics , Rats , Signal Transduction
16.
Endocrinology ; 141(12): 4558-63, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108268

ABSTRACT

Bone morphogenetic proteins (BMP) induce the differentiation of cells of the osteoblastic lineage and enhance the function of the osteoblast. Growth factor activity is regulated by binding proteins, and we previously showed that BMPs induce noggin, a glycoprotein that binds and blocks BMP action. Recently, additional BMP antagonists, such as gremlin, have been described, but there is no information about their expression or function in osteoblasts. We tested for the expression of gremlin and studied its induction by BMPs in cultures of osteoblast-enriched cells from 22-day-old fetal rat calvariae (Ob cells). BMP-2 caused a time- and dose-dependent increase in gremlin messenger RNA and polypeptide levels, as determined by Northern and Western blot analyses. The effects of BMP-2 on gremlin transcripts were independent of new protein synthesis. BMP-2 increased the rate of gremlin transcription as determined by nuclear run-on assays. Fibroblast growth factor-2 and platelet-derived growth factor BB also induced gremlin, but other hormones and growth factors had no effect. Gremlin prevented the stimulatory effects of BMP-2 on DNA, collagen, noncollagen protein synthesis, and alkaline phosphatase activity in Ob cells. In conclusion, BMPs induce gremlin transcription in Ob cells, a mechanism that probably limits BMP action in osteoblasts.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Gene Expression/drug effects , Intercellular Signaling Peptides and Proteins , Osteoblasts/metabolism , Proteins/genetics , Transforming Growth Factor beta , Animals , Becaplermin , Blotting, Northern , Blotting, Western , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 6 , Bone and Bones/embryology , COS Cells , Cells, Cultured , Collagen/biosynthesis , DNA/biosynthesis , Fibroblast Growth Factor 2/pharmacology , Humans , Platelet-Derived Growth Factor/pharmacology , Protein Biosynthesis , Proteins/pharmacology , Proto-Oncogene Proteins c-sis , RNA, Messenger/analysis , Rats
17.
J Bone Miner Res ; 15(4): 663-73, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10780858

ABSTRACT

Bone morphogenetic proteins (BMPs) have been heretofore implicated in the induction of osteoblast differentiation from uncommitted progenitors during embryonic skeletogenesis and fracture healing. We have tested the hypothesis that BMPs are also involved in the osteoblastogenesis that takes place in the bone marrow in postnatal life. To do this, we took advantage of the properties of noggin, a recently discovered protein that binds BMP-2 and -4 and blocks their action. Addition of human recombinant noggin to bone marrow cell cultures from normal adult mice inhibited both osteoblast and osteoclast formation; these effects were reversed by exogenous BMP-2. Consistent with these findings, BMP-2 and -4 and BMP-2/4 receptor transcripts and proteins were detected in these primary cultures, in a bone marrow-derived stromal/osteoblastic cell line, as well as in murine adult whole bone; noggin expression was also documented in all these preparations. Moreover, addition of antinoggin antibody caused an increase in osteoblast progenitor formation. These findings suggest that BMP-2 and -4 are expressed in the bone marrow in postnatal life and serve to maintain the continuous supply of osteoblasts and osteoclasts; and that, in fact, BMP-2/4-induced commitment to the osteoblastic lineage is a prerequisite for osteoclast development. Hence, BMPs, perhaps in balance with noggin and possibly other antagonists, may provide the tonic baseline control of the rate of bone remodeling on which other inputs (e.g., hormonal, biomechanical, etc.) operate.


Subject(s)
Bone Marrow Cells/cytology , Bone Morphogenetic Proteins/antagonists & inhibitors , Osteoblasts/cytology , Osteoclasts/cytology , Proteins/pharmacology , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Protein Receptors, Type II , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Carrier Proteins , Cell Differentiation/drug effects , Cells, Cultured , Humans , Mice , Neutralization Tests , Protein Serine-Threonine Kinases/genetics , Receptors, Growth Factor/genetics , Recombinant Proteins/pharmacology
18.
Development ; 126(23): 5515-22, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10556075

ABSTRACT

In this study, we have analyzed the expression and function of Gremlin in the developing avian limb. Gremlin is a member of the DAN family of BMP antagonists highly conserved through evolution able to bind and block BMP2, BMP4 and BMP7. At early stages of development, gremlin is expressed in the dorsal and ventral mesoderm in a pattern complementary to that of bmp2, bmp4 and bmp7. The maintenance of gremlin expression at these stages is under the control of the AER, ZPA, and BMPs. Exogenous administration of recombinant Gremlin indicates that this protein is involved in the control of limb outgrowth. This function appears to be mediated by the neutralization of BMP function to maintain an active AER, to restrict the extension of the areas of programmed cell death and to confine chondrogenesis to the central core mesenchyme of the bud. At the stages of digit formation, gremlin is expressed in the proximal boundary of the interdigital mesoderm of the chick autopod. The anti-apoptotic influence of exogenous Gremlin, which results in the formation of soft tissue syndactyly in the chick, together with the expression of gremlin in the duck interdigital webs, indicates that Gremlin regulates the regression of the interdigital tissue. At later stages of limb development, gremlin is expressed in association with the differentiating skeletal pieces, muscles and the feather buds. The different expression of Gremlin in relation with other BMP antagonists present in the limb bud, such as Noggin, Chordin and Follistatin indicates that the functions of BMPs are regulated specifically by the different BMP antagonists, acting in a complementary fashion rather than being redundant signals.


Subject(s)
Apoptosis/physiology , Bone Morphogenetic Proteins/metabolism , Chondrogenesis/physiology , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins , Proteins/metabolism , Trans-Activators , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Carrier Proteins , Chick Embryo , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ectoderm/metabolism , Fibroblast Growth Factor 4 , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Follistatin , Glycoproteins/genetics , Glycoproteins/metabolism , Hedgehog Proteins , Homeodomain Proteins , Limb Buds , Mesoderm , MyoD Protein/genetics , MyoD Protein/metabolism , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
19.
Nature ; 401(6750): 243-51, 1999 Sep 16.
Article in English | MEDLINE | ID: mdl-10499580

ABSTRACT

In the chick embryo, left-right asymmetric patterns of gene expression in the lateral plate mesoderm are initiated by signals located in and around Hensen's node. Here we show that Caronte (Car), a secreted protein encoded by a member of the Cerberus/Dan gene family, mediates the Sonic hedgehog (Shh)-dependent induction of left-specific genes in the lateral plate mesoderm. Car is induced by Shh and repressed by fibroblast growth factor-8 (FGF-8). Car activates the expression of Nodal by antagonizing a repressive activity of bone morphogenic proteins (BMPs). Our results define a complex network of antagonistic molecular interactions between Activin, FGF-8, Lefty-1, Nodal, BMPs and Car that cooperate to control left-right asymmetry in the chick embryo.


Subject(s)
Avian Proteins , Body Patterning/physiology , Proteins/physiology , Trans-Activators , Amino Acid Sequence , Animals , Body Patterning/genetics , Bone Morphogenetic Proteins/physiology , COS Cells , Chick Embryo , Cloning, Molecular , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental , Genes, Homeobox , Hedgehog Proteins , Homeodomain Proteins/physiology , Humans , Intercellular Signaling Peptides and Proteins , Left-Right Determination Factors , Molecular Sequence Data , Nodal Protein , Proteins/genetics , Sequence Homology, Amino Acid , Transcription Factors/physiology , Transforming Growth Factor beta/physiology , Xenopus Proteins
20.
Development ; 126(10): 2161-70, 1999 May.
Article in English | MEDLINE | ID: mdl-10207141

ABSTRACT

Major advances in the genetics of vertebrate limb development have been obtained in recent years. However, the nature of the signals which trigger differentiation of the mesoderm to form the limb skeleton remains elusive. Previously, we have obtained evidence for a role of TGFbeta2 in digit formation. Here, we show that activins A and B and/or AB are also signals involved in digit skeletogenesis. activin betaA gene expression correlates with the initiation of digit chondrogenesis while activin betaB is expressed coincidently with the formation of the last phalanx of each digit. Exogenous administration of activins A, B or AB into the interdigital regions induces the formation of extra digits. follistatin, a natural antagonist of activins, is expressed, under the control of activin, peripherally to the digit chondrogenic aggregates marking the prospective tendinous blastemas. Exogenous application of follistatin blocks physiological and activin-induced digit formation. Evidence for a close interaction between activins and other signalling molecules, such as BMPs and FGFs, operating at the distal tip of the limb at these stages is also provided. Chondrogenesis by activins is mediated by BMPs through the regulation of the BMP receptor bmpR-1b and in turn activin expression is upregulated by BMP signalling. In addition, AER hyperactivity secondary to Wnt3A misexpression or local administration of FGFs, inhibits activin expression. In correlation with the restricted expression of activins in the course of digit formation, neither activin nor follistatin treatment affects the development of the skeletal components of the stylopod or zeugopod indicating that the formation of the limb skeleton is regulated by segment-specific chondrogenic signals.


Subject(s)
Extremities/embryology , Inhibins/metabolism , Signal Transduction , Activin Receptors, Type II , Activins , Animals , Cell Death , Chick Embryo , Follistatin , Glycoproteins/metabolism , Inhibins/genetics , Morphogenesis , Receptors, Growth Factor/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...