Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Mol Brain ; 14(1): 134, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34488822

ABSTRACT

Growth differentiation factor 11 (GDF11) is a transforming factor-ß superfamily member that functions as a negative regulator of neurogenesis during embryonic development. However, when recombinant GDF11 (rGDF11) is administered systemically in aged mice, it promotes neurogenesis, the opposite of its role during development. The goal of the present study was to reconcile this apparent discrepancy by performing the first detailed investigation into the expression of endogenous GDF11 in the adult brain and its effects on neurogenesis. Using quantitative histological analysis, we observed that Gdf11 is most highly expressed in adult neurogenic niches and non-neurogenic regions within the hippocampus, choroid plexus, thalamus, habenula, and cerebellum. To investigate the role of endogenous GDF11 during adult hippocampal neurogenesis, we generated a tamoxifen inducible mouse that allowed us to reduce GDF11 levels. Depletion of Gdf11 during adulthood increased proliferation of neural progenitors and decreased the number of newborn neurons in the hippocampus, suggesting that endogenous GDF11 remains a negative regulator of hippocampal neurogenesis in adult mice. These findings further support the idea that circulating systemic GDF11 and endogenously expressed GDF11 in the adult brain have different target cells or mechanisms of action. Our data describe a role for GDF11-dependent signaling in adult neurogenesis that has implications for how GDF11 may be used to treat CNS disease.


Subject(s)
Bone Morphogenetic Proteins/physiology , Growth Differentiation Factors/physiology , Hippocampus/cytology , Nerve Tissue Proteins/physiology , Neurogenesis/physiology , Aging/metabolism , Animals , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Cell Division , Crosses, Genetic , Female , Growth Differentiation Factors/biosynthesis , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/genetics , Hippocampus/growth & development , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Neurogenesis/genetics , Organ Specificity , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Stem Cell Niche
2.
Cell Biochem Funct ; 39(4): 496-510, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33527439

ABSTRACT

Bone morphogenetic protein-11 (BMP11), also known as growth differentiation factor-11 (GDF11), is implicated in skeletal development and joint morphogenesis in mammals. However, its functions in adipogenesis and energy homeostasis are mostly unknown. The present study investigates crucial roles of BMP11 in cultured 3T3-L1 white and HIB1B brown adipocytes, using Bmp11 gene depletion and pharmacological inhibition of BMP11. The silencing of Bmp11 markedly decreases the expression levels of brown-fat signature proteins and beige-specific genes in white adipocytes and significantly down-regulates the expression levels of brown fat-specific genes in brown adipocytes. The deficiency of Bmp11 reduces the expressions of lipolytic protein markers in white and brown adipocytes. Moreover, BMP11 induces browning of 3T3-L1 adipocytes via coordination of multiple signalling pathways, including mTORC1-COX2 and p38MAPK-PGC-1α as non-canonical pathways, as well as Smad1/5/8 as a canonical pathway. We believe this study is the first to provide evidence of the potential roles of BMP11 for improvement of lipid catabolism in both cultured white and brown adipocytes, as well as the effect on browning of white adipocytes. Taken together, these results demonstrate the therapeutic potential for the treatment of obesity.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Thermogenesis , Animals , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Cells, Cultured , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/genetics , Mice , Mitochondria/metabolism
3.
J Cell Physiol ; 234(12): 23360-23368, 2019 12.
Article in English | MEDLINE | ID: mdl-31183862

ABSTRACT

Growth and differentiation factor 11 (GDF11) is a transforming growth factor ß family member that has been identified as the central player of anterior-posterior (A-P) axial skeletal patterning. Mice homozygous for Gdf11 deletion exhibit severe anterior homeotic transformations of the vertebrae and craniofacial defects. During early embryogenesis, Gdf11 is expressed predominantly in the primitive streak and tail bud regions, where new mesodermal cells arise. On the basis of this expression pattern of Gdf11 and the phenotype of Gdf11 mutant mice, it has been suggested that GDF11 acts to specify positional identity along the A-P axis either by local changes in levels of signaling as development proceeds or by acting as a morphogen. To further investigate the mechanism of action of GDF11 in the vertebral specification, we used a Cdx2-Cre transgene to generate mosaic mice in which Gdf11 expression is removed in posterior regions including the tail bud, but not in anterior regions. The skeletal analysis revealed that these mosaic mice display patterning defects limited to posterior regions where Gdf11 expression is deficient, whereas displaying normal skeletal phenotype in anterior regions where Gdf11 is normally expressed. Specifically, the mosaic mice exhibited seven true ribs, a pattern observed in wild-type (wt) mice (vs. 10 true ribs in Gdf11-/- mice), in the anterior axis and nine lumbar vertebrae, a pattern observed in Gdf11 null mice (vs. six lumbar vertebrae in wt mice), in the posterior axis. Our findings suggest that GDF11, rather than globally acting as a morphogen secreted from the tail bud, locally regulates axial vertebral patterning.


Subject(s)
Body Patterning , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Osteogenesis , Spine/metabolism , Animals , Body Patterning/genetics , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Gene Expression Regulation, Developmental , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Mosaicism , Osteogenesis/genetics , Signal Transduction , Spine/embryology
5.
Am J Physiol Heart Circ Physiol ; 317(1): H201-H212, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31125255

ABSTRACT

Administration of active growth differentiation factor 11 (GDF11) to aged mice can reduce cardiac hypertrophy, and low serum levels of GDF11 measured together with the related protein, myostatin (also known as GDF8), predict future morbidity and mortality in coronary heart patients. Using mice with a loxP-flanked ("floxed") allele of Gdf11 and Myh6-driven expression of Cre recombinase to delete Gdf11 in cardiomyocytes, we tested the hypothesis that cardiac-specific Gdf11 deficiency might lead to cardiac hypertrophy in young adulthood. We observed that targeted deletion of Gdf11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation when compared with control mice carrying only the Myh6-cre or Gdf11-floxed alleles, suggesting a possible etiology for dilated cardiomyopathy. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected model. First, whole heart Gdf11 expression did not decrease in Myh6-cre; Gdf11-floxed mice, possibly because of upregulation of Gdf11 in noncardiomyocytes in the heart. Second, we observed Cre-associated toxicity, with lower body weights and increased global fibrosis, in Cre-only control male mice compared with flox-only controls, making it challenging to infer which changes in Myh6-cre;Gdf11-floxed mice were the result of Cre toxicity versus deletion of Gdf11. Third, we observed differential expression of cre mRNA in Cre-only controls compared with the cardiomyocyte-specific knockout mice, also making comparison between these two groups difficult. Thus, targeted Gdf11 deletion in cardiomyocytes may lead to left ventricular dilation without hypertrophy, but alternative animal models are necessary to understand the mechanism for these findings. NEW & NOTEWORTHY We observed that targeted deletion of growth differentiation factor 11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation compared with control mice carrying only the Myh6-cre or growth differentiation factor 11-floxed alleles. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected mouse model.


Subject(s)
Bone Morphogenetic Proteins/genetics , Cardiomyopathy, Dilated/genetics , Gene Deletion , Growth Differentiation Factors/genetics , Integrases/genetics , Myocytes, Cardiac/metabolism , Age Factors , Animals , Bone Morphogenetic Proteins/deficiency , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Disease Progression , Female , Gene Knockdown Techniques , Genetic Predisposition to Disease , Growth Differentiation Factors/deficiency , Integrases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Phenotype , Ventricular Function, Left , Ventricular Remodeling
6.
Pathol Res Pract ; 215(1): 121-126, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30401582

ABSTRACT

INTRODUCTION: The expression of bone morphogenetic protein-10 (BMP-10) is downregulated in some cancer types, but its function and mechanism in ovarian cancer remains unclear. MATERIALS AND METHODS: BMP-10 expression was detected in ovarian cancer tissues and cell lines by using immunochemistry and western blotting. Prognostic value of BMP-10 was evaluated by Kaplan-Meier curve and Cox regression model. Knockdown or overexpression of BMP-10 was conducted by using specific siRNA or pcDNA-BMP-10 in ovarian cancer cell lines. The biological features induced by BMP-10 were observed by MTT assay, wound-healing and transwell assays. RESULTS: BMP-10 expression in ovarian cancer tissues was significantly lower than that in ovarian tissues. Low BMP-10 expression in ovarian cancer tissues was related to advance FIGO stage, higher histologic grade, lymph node metastasis, and peritoneal fluid. Kaplan-Meier analysis revealed that low BMP-10 expression was significantly associated with poor prognosis of patients with ovarian cancer. BMP-10 overexpression or knockdown significantly inhibited or promoted proliferation, migration, and invasion of ovarian cancer cells, respectively. Moreover, administration of neutralizing antibody or human recombinant BMP-10 would reverse these effects on ovarian cancer cells. CONCLUSION: Low BMP-10 expression was associated with poor prognosis and progression of ovarian cancer.


Subject(s)
Bone Morphogenetic Proteins/deficiency , Gene Expression Regulation, Neoplastic/genetics , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/mortality , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Humans , Lymphatic Metastasis/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , RNA, Long Noncoding/genetics
7.
Development ; 144(15): 2824-2836, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28694256

ABSTRACT

During development and homeostasis, precise control of Wnt/ß-catenin signaling is in part achieved by secreted and membrane proteins that negatively control activity of the Wnt co-receptors Lrp5 and Lrp6. Lrp4 is related to Lrp5/6 and is implicated in modulation of Wnt/ß-catenin signaling, presumably through its ability to bind to the Wise (Sostdc1)/sclerostin (Sost) family of Wnt antagonists. To gain insights into the molecular mechanisms of Lrp4 function in modulating Wnt signaling, we performed an array of genetic analyses in murine tooth development, where Lrp4 and Wise play important roles. We provide genetic evidence that Lrp4 mediates the Wnt inhibitory function of Wise and also modulates Wnt/ß-catenin signaling independently of Wise. Chimeric receptor analyses raise the possibility that the Lrp4 extracellular domain interacts with Wnt ligands, as well as the Wnt antagonists. Diverse modes of Lrp4 function are supported by severe tooth phenotypes of mice carrying a human mutation known to abolish Lrp4 binding to Sost. Our data suggest a model whereby Lrp4 modulates Wnt/ß-catenin signaling via interaction with Wnt ligands and antagonists in a context-dependent manner.


Subject(s)
Receptors, LDL/metabolism , Tooth/embryology , Tooth/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice , Mice, Mutant Strains , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tooth/pathology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics
8.
Bone ; 96: 51-62, 2017 03.
Article in English | MEDLINE | ID: mdl-27742500

ABSTRACT

Sclerosteosis and van Buchem disease are two rare bone sclerosing dysplasias caused by genetic defects in the synthesis of sclerostin. In this article we review the demographic, clinical, biochemical, radiological, and histological characteristics of patients with sclerosteosis and van Buchem disease that led to a better understanding of the role of sclerostin in bone metabolism in humans and we discuss the relevance of these findings for the development of new therapeutics for the treatment of patients with osteoporosis.


Subject(s)
Bone Morphogenetic Proteins/deficiency , Adaptor Proteins, Signal Transducing , Biomarkers/metabolism , Bone Density , Bone and Bones/pathology , Bone and Bones/physiopathology , Genetic Markers , Humans , Hyperostosis/diagnostic imaging , Hyperostosis/pathology , Hyperostosis/physiopathology , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/pathology , Osteochondrodysplasias/physiopathology , Syndactyly/diagnostic imaging , Syndactyly/pathology , Syndactyly/physiopathology
9.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(11): 1481-1485, 2016 Nov.
Article in Chinese | MEDLINE | ID: mdl-27774939

ABSTRACT

Objective To construct a recombinant adenovirus expressing siRNA targeting human sclerostin (SOST) gene, and test the function of MG-63 cells while co-cultured with MDA-MB-231 cells infected by Ad-siSOST. MethodsAccording to the RNA sequence of SOST gene, two pairs of primers which contained 3 siRNA sequences were designed, and a pB2B plasmid was taken as template to amplify 2 DNA sequences. Both of the 2 DNA sequences were ligated to pAdTrace-OK by Gibson DNA Assembly way. After homologous recombination between recombinant shuttle plasmid and adenovirus vector plasmid, the adenovirus was packaged in HEK-293 cells. PCR and ELISA were used to test the expression of SOST in MDA-MB-231 cells which were infected with Ad-siSOST. In the co-culture system of MG-63 cells and MDA-MB-231 cells infected Ad-siSOST, osteoprotegerin (OPG), osteocalcin (OCN), integrin binding sialoprotein (IBSP) and receptor activator of nuclear factor-κB ligand (RANKL) were tested by quantitative real-time PCR. Results Recombinant shuttle plasmid which contained 3 interfering fragments was constructed successfully, and Ad-siSOST was obtained after homologous recombination and packaging. SOST expression in MDA-MB-231 cells was downregulated significantly after infeceted with Ad-siSOST. The mRNA levels of OPG, OCN, IBSP in MG-63 cells increased significantly, while the level of RANKL mRNA decreased significantly, and all 4 gene expressions were reversed after the infection of Ad-siSOST. Conclusion Knockdown of SOST in MG-63 cells increases osteogenesis and ratio of OPG/RANKL in vitro.


Subject(s)
Bone Morphogenetic Proteins/genetics , Breast Neoplasms/metabolism , Genetic Markers/genetics , Osteoblasts/metabolism , Adaptor Proteins, Signal Transducing , Bone Morphogenetic Proteins/deficiency , Breast Neoplasms/genetics , Cell Line, Tumor , Coculture Techniques , HEK293 Cells , Humans , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Osteoblasts/cytology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Real-Time Polymerase Chain Reaction
10.
Bone ; 92: 180-188, 2016 11.
Article in English | MEDLINE | ID: mdl-27601226

ABSTRACT

Sclerostin (Sost) is a negative regulator of bone formation that acts upon the Wnt signaling pathway. Sost is mechanically regulated at both mRNA and protein level such that loading represses and unloading enhances Sost expression, in osteocytes and in circulation. The non-coding evolutionarily conserved enhancer ECR5 has been previously reported as a transcriptional regulatory element required for modulating Sost expression in osteocytes. Here we explored the mechanisms by which ECR5, or several other putative transcriptional enhancers regulate Sost expression, in response to mechanical stimulation. We found that in vivo ulna loading is equally osteoanabolic in wildtype and Sost-/- mice, although Sost is required for proper distribution of load-induced bone formation to regions of high strain. Using Luciferase reporters carrying the ECR5 non-coding enhancer and heterologous or homologous hSOST promoters, we found that ECR5 is mechanosensitive in vitro and that ECR5-driven Luciferase activity decreases in osteoblasts exposed to oscillatory fluid flow. Yet, ECR5-/- mice showed similar magnitude of load-induced bone formation and similar periosteal distribution of bone formation to high-strain regions compared to wildtype mice. Further, we found that in contrast to Sost-/- mice, which are resistant to disuse-induced bone loss, ECR5-/- mice lose bone upon unloading to a degree similar to wildtype control mice. ECR5 deletion did not abrogate positive effects of unloading on Sost, suggesting that additional transcriptional regulators and regulatory elements contribute to load-induced regulation of Sost.


Subject(s)
Adaptation, Physiological/physiology , Enhancer Elements, Genetic/physiology , Glycoproteins/deficiency , Osteocytes/physiology , Osteogenesis/physiology , Adaptor Proteins, Signal Transducing , Animals , Biomechanical Phenomena/physiology , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Female , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mice, Transgenic , RNA, Untranslated/genetics
11.
BMC Dev Biol ; 16: 14, 2016 May 13.
Article in English | MEDLINE | ID: mdl-27178071

ABSTRACT

BACKGROUND: Wnt5a and Mrfzb1 genes are involved in the regulation of tooth size, and their expression levels are similar to that of Bmp7 during morphogenesis, including during the cap and early bell stages of tooth formation. We previously reported that Usag-1-deficient mice form supernumerary maxillary incisors. Thus, we hypothesized that BMP7 and USAG-1 signaling molecules may play important roles in tooth morphogenesis. In this study, we established double genetically modified mice to examine the in vivo inter-relationships between Bmp7 and Usag-1. RESULTS: We measured the volume and cross-sectional areas of the mandibular incisors using micro-computed tomography (micro-CT) in adult Bmp7- and Usag-1-LacZ knock-in mice and their F2 generation upon interbreeding. The mandibular incisors of adult Bmp7+/- mice were significantly larger than those of wild-type (WT) mice. The mandibular incisors of adult Usag-1-/- mice were the largest of all genotypes examined. In the F2 generation, the effects of these genes were additive; Bmp7+/- was most strongly associated with the increase in tooth size using generalized linear models, and the total area of mandibular supernumerary incisors of Usag-1-/-Bmp7+/- mice was significantly larger than that of Usag-1-/-Bmp7 +/+ mice. At embryonic day 15 (E15), BrdU assays demonstrated that the labeling index of Bmp7+/- embryos was significantly higher than that of WT embryos in the cervical loop. Additionally, the labeling index of Usag-1-/- embryos was significantly the highest of all genotypes examined in dental papilla. CONCLUSIONS: Bmp7 heterozygous mice exhibited significantly increased tooth sizes, suggesting that tooth size was controlled by specific gene expression. Our findings may be useful in applications of regenerative medicine and dentistry.


Subject(s)
Bone Morphogenetic Protein 7/deficiency , Bone Morphogenetic Proteins/deficiency , Morphogenesis , Tooth/embryology , Adaptor Proteins, Signal Transducing , Aging , Animals , Apoptosis , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/metabolism , Bromodeoxyuridine/metabolism , Cell Proliferation , Crosses, Genetic , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , In Situ Nick-End Labeling , Incisor/diagnostic imaging , Incisor/metabolism , Linear Models , Male , Mandible/diagnostic imaging , Mandible/metabolism , Mice, Inbred C57BL , Molar/metabolism , Organ Size , Phenotype , Staining and Labeling , Tooth/diagnostic imaging , Tooth/metabolism , X-Ray Microtomography , beta-Galactosidase/metabolism
12.
Circ Res ; 118(7): 1125-41; discussion 1142, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27034275

ABSTRACT

Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor ß superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.


Subject(s)
Bone Morphogenetic Proteins/physiology , Growth Differentiation Factors/physiology , Myostatin/physiology , Adult , Aging/physiology , Amino Acid Sequence , Animals , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/deficiency , Brain/growth & development , Brain/physiology , Dimerization , Female , Follistatin/metabolism , Follistatin-Related Proteins/metabolism , Growth Differentiation Factors/chemistry , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/therapeutic use , Heart/physiology , Heart Diseases/metabolism , Humans , Male , Mice , Models, Molecular , Molecular Sequence Data , Muscles/physiology , Myocardium/metabolism , Myostatin/chemistry , Myostatin/deficiency , Organ Specificity , Protein Conformation , Protein Structure, Tertiary , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Structure-Activity Relationship
13.
Circ Res ; 118(7): 1143-50; discussion 1150, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27034276

ABSTRACT

This "Controversies in Cardiovascular Research" article evaluates the evidence for and against the hypothesis that the circulating blood level of growth differentiation factor 11 (GDF11) decreases in old age and that restoring normal GDF11 levels in old animals rejuvenates their skeletal muscle and reverses pathological cardiac hypertrophy and cardiac dysfunction. Studies supporting the original GDF11 hypothesis in skeletal and cardiac muscle have not been validated by several independent groups. These new studies have either found no effects of restoring normal GDF11 levels on cardiac structure and function or have shown that increasing GDF11 or its closely related family member growth differentiation factor 8 actually impairs skeletal muscle repair in old animals. One possible explanation for what seems to be mutually exclusive findings is that the original reagent used to measure GDF11 levels also detected many other molecules so that age-dependent changes in GDF11 are still not well known. The more important issue is whether increasing blood [GDF11] repairs old skeletal muscle and reverses age-related cardiac pathologies. There are substantial new and existing data showing that GDF8/11 can exacerbate rather than rejuvenate skeletal muscle injury in old animals. There is also new evidence disputing the idea that there is pathological hypertrophy in old C57bl6 mice and that GDF11 therapy can reverse cardiac pathologies. Finally, high [GDF11] causes reductions in body and heart weight in both young and old animals, suggestive of a cachexia effect. Our conclusion is that elevating blood levels of GDF11 in the aged might cause more harm than good.


Subject(s)
Aging/pathology , Bone Morphogenetic Proteins/therapeutic use , Growth Differentiation Factors/therapeutic use , Muscular Diseases/drug therapy , Aging/blood , Animals , Bone Morphogenetic Proteins/blood , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/pharmacology , Bone Morphogenetic Proteins/toxicity , Cachexia/chemically induced , Cells, Cultured , Drug Evaluation, Preclinical , Growth Differentiation Factors/blood , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/pharmacology , Growth Differentiation Factors/toxicity , Heart/drug effects , Humans , Hypertrophy , Mice, Inbred C57BL , Models, Animal , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Muscles/pathology , Muscular Diseases/physiopathology , Myocardium/pathology , Myostatin/physiology , Myostatin/therapeutic use , Myostatin/toxicity , Parabiosis , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Regeneration/drug effects , Reproducibility of Results , Signal Transduction , Single-Blind Method , Smad2 Protein/physiology , Smad3 Protein/physiology
14.
Bone ; 88: 20-30, 2016 07.
Article in English | MEDLINE | ID: mdl-27102547

ABSTRACT

Loss of Sostdc1, a growth factor paralogous to Sost, causes the formation of ectopic incisors, fused molars, abnormal hair follicles, and resistance to kidney disease. Sostdc1 is expressed in the periosteum, a source of osteoblasts, fibroblasts and mesenchymal progenitor cells, which are critically important for fracture repair. Here, we investigated the role of Sostdc1 in bone metabolism and fracture repair. Mice lacking Sostdc1 (Sostdc1(-/-)) had a low bone mass phenotype associated with loss of trabecular bone in both lumbar vertebrae and in the appendicular skeleton. In contrast, Sostdc1(-/-) cortical bone measurements revealed larger bones with higher BMD, suggesting that Sostdc1 exerts differential effects on cortical and trabecular bone. Mid-diaphyseal femoral fractures induced in Sostdc1(-/-) mice showed that the periosteal population normally positive for Sostdc1 rapidly expands during periosteal thickening and these cells migrate into the fracture callus at 3days post fracture. Quantitative analysis of mesenchymal stem cell (MSC) and osteoblast populations determined that MSCs express Sostdc1, and that Sostdc1(-/-) 5day calluses harbor >2-fold more MSCs than fractured wildtype controls. Histologically a fraction of Sostdc1-positive cells also expressed nestin and α-smooth muscle actin, suggesting that Sostdc1 marks a population of osteochondral progenitor cells that actively participate in callus formation and bone repair. Elevated numbers of MSCs in D5 calluses resulted in a larger, more vascularized cartilage callus at day 7, and a more rapid turnover of cartilage with significantly more remodeled bone and a thicker cortical shell at 21days post fracture. These data support accelerated or enhanced bone formation/remodeling of the callus in Sostdc1(-/-) mice, suggesting that Sostdc1 may promote and maintain mesenchymal stem cell quiescence in the periosteum.


Subject(s)
Bone Morphogenetic Proteins/deficiency , Fracture Healing , Mesenchymal Stem Cells/cytology , Periosteum/cytology , Actins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Biomechanical Phenomena , Bone Morphogenetic Proteins/metabolism , Bony Callus/pathology , Calcification, Physiologic , Cancellous Bone/diagnostic imaging , Cancellous Bone/pathology , Cell Differentiation , Cell Proliferation , Cortical Bone/diagnostic imaging , Cortical Bone/pathology , Femur/pathology , Gene Deletion , Mice, Inbred C57BL , Nestin/metabolism , Organ Size , Osteoblasts/metabolism , Osteogenesis , Phenotype , Sp7 Transcription Factor/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway , X-Ray Microtomography
16.
PLoS One ; 9(5): e96938, 2014.
Article in English | MEDLINE | ID: mdl-24816837

ABSTRACT

Bone morphogenetic proteins (BMPs) are highly conserved signaling molecules that are part of the transforming growth factor (TGF)-beta superfamily, and function in the patterning and morphogenesis of many organs including development of the dentition. The functions of the BMPs are controlled by certain classes of molecules that are recognized as BMP antagonists that inhibit BMP binding to their cognate receptors. In this study we tested the hypothesis that USAG-1 (uterine sensitization-associated gene-1) suppresses deciduous incisors by inhibition of BMP-7 function. We learned that USAG-1 and BMP-7 were expressed within odontogenic epithelium as well as mesenchyme during the late bud and early cap stages of tooth development. USAG-1 is a BMP antagonist, and also modulates Wnt signaling. USAG-1 abrogation rescued apoptotic elimination of odontogenic mesenchymal cells. BMP signaling in the rudimentary maxillary incisor, assessed by expressions of Msx1 and Dlx2 and the phosphorylation of Smad protein, was significantly enhanced. Using explant culture and subsequent subrenal capsule transplantation of E15 USAG-1 mutant maxillary incisor tooth primordia supplemented with BMP-7 demonstrated in USAG-1+/- as well as USAG-1-/- rescue and supernumerary tooth development. Based upon these results, we conclude that USAG-1 functions as an antagonist of BMP-7 in this model system. These results further suggest that the phenotypes of USAG-1 and BMP-7 mutant mice reported provide opportunities for regenerative medicine and dentistry.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/metabolism , Organogenesis , Tooth, Supernumerary/embryology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Protein 7/antagonists & inhibitors , Bone Morphogenetic Proteins/deficiency , Epithelial Cells/metabolism , Incisor/embryology , Mesoderm/metabolism , Mice , Protein Binding , Protein Transport , Signal Transduction , Tooth, Supernumerary/metabolism , Tooth, Supernumerary/pathology
17.
J Bone Miner Res ; 29(10): 2144-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24753092

ABSTRACT

High bone mass in animals and humans with sclerostin deficiency is associated with increased bone strength, which is not the case for all disorders with high bone mineral density, some of which are even associated with fragility fractures owing to unfavorable bone composition. In the current study we investigated whether alterations in bone composition may contribute to the bone strength characteristics associated with lack of sclerostin. We examined cortical bone of Sost-knockout (KO) mice (n = 9, 16 weeks old) and sclerosteosis patients (young [4 to 14 years], n = 4 and adults [24 and 43 years], n = 2) by quantitative backscattered electron imaging and Raman microspectroscopy and compared it to bone from wild-type mice and healthy subjects, respectively. In Sost-KO mice endocortical bone exhibited altered bone composition, whereas subperiosteal bone was unchanged. When comparing endocortical bone tissue of identical tissue age as defined by sequential dual fluorochrome labeling the average bone matrix mineralization was reduced -1.9% (p < 0.0001, younger tissue age) and -1.5% (p < 0.05, older tissue age), and the relative proteoglycan content was significantly increased. Similarly, bone matrix mineralization density distribution was also shifted toward lower matrix mineralization in surgical samples of compact bone of sclerosteosis patients. This was associated with an increase in mineralization heterogeneity in the young population. In addition, and consistently, the relative proteoglycan content was increased. In conclusion, we observed decreased matrix mineralization and increased relative proteoglycan content in bone subcompartments of Sost-KO mice-a finding that translated into sclerosteosis patients. We hypothesize that the altered bone composition contributes to the increased bone strength of patients with sclerostin deficiency.


Subject(s)
Bone Density , Bone Morphogenetic Proteins/deficiency , Glycoproteins/deficiency , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Analysis of Variance , Animals , Bone Morphogenetic Proteins/metabolism , Calcification, Physiologic , Child , Child, Preschool , Electrons , Genetic Markers , Glycoproteins/metabolism , Humans , Hyperostosis/pathology , Hyperostosis/physiopathology , Intercellular Signaling Peptides and Proteins , Mice, Knockout , Microscopy, Fluorescence , Spectrum Analysis, Raman , Syndactyly/pathology , Syndactyly/physiopathology , Young Adult
18.
J Clin Endocrinol Metab ; 99(2): E252-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24302746

ABSTRACT

CONTEXT: Sclerostin and Dickkopf 1 (DKK1) are antagonists of the canonical Wnt signaling pathway, both binding to the same low-density lipoprotein receptor-related protein 5/6 on osteoblasts, thereby inhibiting bone formation. It is not known whether there is an interaction between sclerostin and DKK1. OBJECTIVE: We examined whether a lack of sclerostin is compensated by increased DKK1 levels. DESIGN, SETTING, AND PATIENTS: We measured DKK1 levels in serum samples of patients and carriers of sclerosteosis (19 patients, 24 carriers) and van Buchem disease (VBD) (13 patients, 22 carriers) and 25 healthy controls. Sclerosteosis and VBD are caused by deficient sclerostin synthesis and are characterized by increased bone formation and hyperostotic phenotypes. MAIN OUTCOME MEASURES: DKK1 levels were compared between patients and carriers, and between patients and healthy controls. We also examined associations between levels of DKK1 and the bone turnover markers procollagen type 1 amino-terminal propeptide and carboxy-terminal cross-linking telopeptide. RESULTS: We found that DKK1 levels were significantly higher in patients with both sclerosteosis (4.28 ng/mL [95% confidence interval (CI), 3.46-5.11 ng/mL]) and VBD (5.28 ng/mL [95% CI, 3.84-6.71 ng/mL]), compared to levels in carriers of the two diseases (sclerosteosis, 2.03 ng/mL [95% CI, 1.78-2.29 ng/mL], P < .001; VBD, 3.47 ng/mL [95% CI, 2.97-3.97 ng/mL], P = 0.017) and to levels in healthy controls (2.77 ng/mL [95% CI, 2.45-3.08 ng/mL]; P = 0.004 and P < .001, respectively). Serum DKK1 levels were positively associated with levels of procollagen type 1 amino-terminal propeptide and carboxy-terminal cross-linking telopeptide in both disorders. CONCLUSIONS: These results suggest that increased DKK1 levels observed in patients with sclerosteosis and VBD represent an adaptive response to the increased bone formation characterizing these diseases, although these increased levels do not compensate for the lack of sclerostin on bone formation.


Subject(s)
Bone Morphogenetic Proteins/deficiency , Bone Remodeling/physiology , Hyperostosis/blood , Intercellular Signaling Peptides and Proteins/blood , Osteochondrodysplasias/blood , Syndactyly/blood , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Genetic Markers , Humans , Male , Middle Aged
19.
Proc Natl Acad Sci U S A ; 110(39): E3713-22, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-24019467

ABSTRACT

Myostatin (MSTN) and growth and differentiation factor-11 (GDF-11) are highly related TGF-ß family members that have distinct biological functions. MSTN is expressed primarily in skeletal muscle and acts to limit muscle growth. GDF-11 is expressed more widely and plays multiple roles, including regulating axial skeletal patterning during development. Several MSTN and GDF-11 binding proteins have been identified, including GDF-associated serum protein-1 (GASP-1) and GASP-2, which are capable of inhibiting the activities of these ligands. Here, we show that GASP-1 and GASP-2 act by blocking the initial signaling event (namely, the binding of the ligand to the type II receptor). Moreover, we show that mice lacking Gasp1 and Gasp2 have phenotypes consistent with overactivity of MSTN and GDF-11. Specifically, we show that Gasp2(-/-) mice have posteriorly directed transformations of the axial skeleton, which contrast with the anteriorly directed transformations seen in Gdf11(-/-) mice. We also show that both Gasp1(-/-) and Gasp2(-/-) mice have reductions in muscle weights, a shift in fiber type from fast glycolytic type IIb fibers to fast oxidative type IIa fibers, and impaired muscle regeneration ability, which are the reverse of what are seen in Mstn(-/-) mice. All of these findings suggest that both GASP-1 and GASP-2 are important modulators of GDF-11 and MSTN activity in vivo.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Growth Differentiation Factors/metabolism , Myostatin/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Body Patterning/genetics , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/deficiency , Bone and Bones/embryology , Bone and Bones/metabolism , Cardiotoxins , Carrier Proteins/genetics , Follistatin/deficiency , Follistatin/metabolism , Gene Expression Profiling , Gene Expression Regulation , Growth Differentiation Factors/antagonists & inhibitors , Growth Differentiation Factors/deficiency , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Mutation/genetics , Myostatin/antagonists & inhibitors , Myostatin/genetics , Organ Size , Oxidation-Reduction , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, Transforming Growth Factor beta/metabolism , Regeneration/genetics , Signal Transduction/genetics
20.
Dev Cell ; 23(5): 981-94, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23153495

ABSTRACT

How dermal papilla (DP) niche cells regulate hair follicle progenitors to control hair growth remains unclear. Using Tbx18(Cre) to target embryonic DP precursors, we ablate the transcription factor Sox2 early and efficiently, resulting in diminished hair shaft outgrowth. We find that DP niche expression of Sox2 controls the migration speed of differentiating hair shaft progenitors. Transcriptional profiling of Sox2 null DPs reveals increased Bmp6 and decreased BMP inhibitor Sostdc1, a direct Sox2 transcriptional target. Subsequently, we identify upregulated BMP signaling in knockout hair shaft progenitors and demonstrate that Bmp6 inhibits cell migration, an effect that can be attenuated by Sostdc1. A shorter and Sox2-negative hair type lacks Sostdc1 in the DP and shows reduced migration and increased BMP activity of hair shaft progenitors. Collectively, our data identify Sox2 as a key regulator of hair growth that controls progenitor migration by fine-tuning BMP-mediated mesenchymal-epithelial crosstalk.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Hair Follicle/embryology , Hair Follicle/metabolism , Hair/growth & development , SOXB1 Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Proteins/deficiency , Cell Movement , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Hair Follicle/cytology , Hair Follicle/growth & development , Mice , Mice, Knockout , Mice, Transgenic , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics , Signal Transduction , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...