Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 114(47): 12448-12453, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109273

ABSTRACT

The TGF-ß family ligands myostatin, GDF11, and activins are negative regulators of skeletal muscle mass, which have been reported to primarily signal via the ActRIIB receptor on skeletal muscle and thereby induce muscle wasting described as cachexia. Use of a soluble ActRIIB-Fc "trap," to block myostatin pathway signaling in normal or cachectic mice leads to hypertrophy or prevention of muscle loss, perhaps suggesting that the ActRIIB receptor is primarily responsible for muscle growth regulation. Genetic evidence demonstrates however that both ActRIIB- and ActRIIA-deficient mice display a hypertrophic phenotype. Here, we describe the mode of action of bimagrumab (BYM338), as a human dual-specific anti-ActRIIA/ActRIIB antibody, at the molecular and cellular levels. As shown by X-ray analysis, bimagrumab binds to both ActRIIA and ActRIIB ligand binding domains in a competitive manner at the critical myostatin/activin binding site, hence preventing signal transduction through either ActRII. Myostatin and the activins are capable of binding to both ActRIIA and ActRIIB, with different affinities. However, blockade of either single receptor through the use of specific anti-ActRIIA or anti-ActRIIB antibodies achieves only a partial signaling blockade upon myostatin or activin A stimulation, and this leads to only a small increase in muscle mass. Complete neutralization and maximal anabolic response are achieved only by simultaneous blockade of both receptors. These findings demonstrate the importance of ActRIIA in addition to ActRIIB in mediating myostatin and activin signaling and highlight the need for blocking both receptors to achieve a strong functional benefit.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Hypertrophy/chemically induced , Muscle, Skeletal/drug effects , Activin Receptors, Type II/metabolism , Activins/metabolism , Animals , Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bone Morphogenetic Proteins/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Growth Differentiation Factors/metabolism , HEK293 Cells , Humans , Hypertrophy/pathology , Male , Mice , Mice, SCID , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myostatin/metabolism , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Wasting Syndrome/drug therapy , Wasting Syndrome/pathology
2.
Front Cell Dev Biol ; 5: 47, 2017.
Article in English | MEDLINE | ID: mdl-28523267

ABSTRACT

Mammalian limb development is driven by the integrative input from several signaling pathways; a failure to receive or a misinterpretation of these signals results in skeletal defects. The brachydactylies, a group of overlapping inherited human hand malformation syndromes, are mainly caused by mutations in BMP signaling pathway components. Two closely related forms, Brachydactyly type B2 (BDB2) and BDB1 are caused by mutations in the BMP antagonist Noggin (NOG) and the atypical receptor tyrosine kinase ROR2 that acts as a receptor in the non-canonical Wnt pathway. Genetic analysis of Nog and Ror2 functional interaction via crossing Noggin and Ror2 mutant mice revealed a widening of skeletal elements in compound but not in any of the single mutants, thus indicating genetic interaction. Since ROR2 is a non-canonical Wnt co-receptor specific for Wnt-5a we speculated that this phenotype might be a result of deregulated Wnt-5a signaling activation, which is known to be essential for limb skeletal elements growth and patterning. We show that Noggin potentiates activation of the Wnt-5a-Ror2-Disheveled (Dvl) pathway in mouse embryonic fibroblast (MEF) cells in a Ror2-dependent fashion. Rat chondrosarcoma chondrocytes (RCS), however, are not able to respond to Noggin in this fashion unless growth arrest is induced by FGF2. In summary, our data demonstrate genetic interaction between Noggin and Ror2 and show that Noggin can sensitize cells to Wnt-5a/Ror2-mediated non-canonical Wnt signaling, a feature that in cartilage may depend on the presence of active FGF signaling. These findings indicate an unappreciated function of Noggin that will help to understand BMP and Wnt/PCP signaling pathway interactions.

3.
Proc Natl Acad Sci U S A ; 107(32): 14211-6, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660756

ABSTRACT

Elongation of the digit rays resulting in the formation of a defined number of phalanges is a process poorly understood in mammals, whereas in the chicken distal mesenchymal bone morphogenetic protein (BMP) signaling in the so-called phalanx-forming region (PFR) or digit crescent (DC) seems to be involved. The human brachydactylies (BDs) are inheritable conditions characterized by variable degrees of digit shortening, thus providing an ideal model to analyze the development and elongation of phalanges. We used a mouse model for BDB1 (Ror2(W749X/W749X)) lacking middle phalanges and show that a signaling center corresponding to the chick PFR exists in the mouse, which is diminished in BDB1 mice. This resulted in a strongly impaired elongation of the digit condensations due to reduced chondrogenic commitment of undifferentiated distal mesenchymal cells. We further show that a similar BMP-based mechanism accounts for digit shortening in a mouse model for the closely related condition BDA1 (Ihh(E95K/E95K)), altogether indicating the functional significance of the PFR in mammals. Genetic interaction experiments as well as pathway analysis in BDB1 mice suggest that Indian hedgehog and WNT/beta-catenin signaling, which we show is inhibited by receptor tyrosine kinase-like orphan receptor 2 (ROR2) in distal limb mesenchyme, are acting upstream of BMP signaling in the PFR.


Subject(s)
Hedgehog Proteins/physiology , Receptor Tyrosine Kinase-like Orphan Receptors/physiology , Toe Phalanges/growth & development , Toe Phalanges/physiology , Animals , Bone Morphogenetic Proteins , Extremities/growth & development , Hedgehog Proteins/genetics , Mice , Mice, Mutant Strains , Mutation, Missense , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction , Wnt Proteins
4.
FASEB J ; 24(7): 2417-26, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20215527

ABSTRACT

Dishevelled (Dvl) is a multifunctional effector of different Wnt cascades. Both canonical Wnt3a and noncanonical Wnt5a stimulate casein-kinase-1 (CK1) -mediated phosphorylation of Dvl, visualized as electrophoretic mobility shift [phosphorylated and shifted Dvl (ps-Dvl)]. However, the role of this phosphorylation remains obscure. Here we report the functional interaction of ps-Dvl with the receptor tyrosine kinase Ror2, which is an alternative Wnt receptor and is able to inhibit canonical Wnt signaling. We demonstrate interaction between Ror2 and ps-Dvl at the cell membrane after Wnt3a or Wnt5a stimulus dependent on CK1. Ps-Dvl interacts with the C-terminal proline-serine-threonine-rich domain of Ror2, which is required for efficient inhibition of canonical Wnt signaling. We further show that the Dvl C terminus, which seems to be exposed in ps-Dvl and efficiently binds Ror2, is an intrinsic negative regulator of the canonical Wnt pathway downstream of beta-catenin. The Dvl C terminus is necessary and sufficient to inhibit canonical Wnt/beta-catenin signaling, which is dependent on the presence of Ror2. Furthermore, both the Dvl C terminus and CK1epsilon can inhibit the Wnt5a/Ror2/ATF2 pathway in mammalian cells and Xenopus explant cultures. This suggests that phosphorylation of Dvl triggers negative feedback regulation for different branches of Wnt signaling in a Ror2-dependent manner.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Casein Kinase I/metabolism , Feedback, Physiological , Phosphoproteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dishevelled Proteins , Humans , Mice , Phosphorylation , Wnt-5a Protein , Wnt3 Protein , Wnt3A Protein , Xenopus , Xenopus Proteins
5.
Biochem Biophys Res Commun ; 390(2): 211-6, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19785987

ABSTRACT

Wtip is a LIM domain protein of the Ajuba/Zyxin family involved in kidney and neural crest development; Ror2 is a receptor tyrosine kinase involved in the development of skeleton, heart, lung, genitalia and kidneys. Here we describe Wtip as an intracellular interaction partner of Ror2. Full-length Ror2 recruits Wtip to the cell membrane, a mutant involved in human disease fails to do so. Both genes and proteins show overlapping expression in the mouse embryo. We show that Wtip is able to inhibit canonical Wnt signalling in mammalian cells and in Xenopus embryos linking Wtip to a crucial developmental pathway.


Subject(s)
Carrier Proteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Co-Repressor Proteins , Cytoskeletal Proteins , Humans , Mice , Protein Structure, Tertiary , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction , Two-Hybrid System Techniques , Wnt Proteins/metabolism , Xenopus
6.
Hum Mol Genet ; 18(21): 4013-21, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19640924

ABSTRACT

Mutations in ROR2 cause dominant brachydactyly type B (BDB1) or recessive Robinow syndrome (RRS), each characterized by a distinct combination of phenotypic features. We here report a novel nonsense mutation in ROR2 (c.1324C>T; p.R441X) causing intracellular protein truncation in a patient exhibiting features of RRS in conjunction with severe recessive brachydactyly. The mutation is located at the same position as a previously described frame shift mutation causing dominant BDB1. To investigate the apparent discrepancy in phenotypic outcome, we analysed ROR2 protein stability and distribution in stably transfected cell lines expressing exact copies of several human RRS and BDB1 intracellular mutations. RRS mutant proteins were less abundant and retained intracellularly, although BDB1 mutants were stable and predominantly located at the cell membrane. The p.R441X mutation showed an intermediate pattern with membrane localization but also high endoplasmic reticulum retention. Furthermore, we observed a correlation between the severity of BDB1, the location of the mutation, and the amount of membrane-associated ROR2. Membrane protein fraction quantification revealed a gradient of distribution and stability correlating with the clinical phenotypes. This gradual model was confirmed by crossing mouse models for RRS and BDB1, yielding double heterozygous animals that exhibited an intermediate phenotype. We propose a model in which the RRS versus the BDB1 phenotype is determined by the relative degree of protein retention/degradation and the amount of mutant protein reaching the plasma membrane.


Subject(s)
Abnormalities, Multiple/genetics , Codon, Nonsense , Limb Deformities, Congenital/pathology , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Abnormalities, Multiple/pathology , Animals , Blotting, Western , Bone Diseases, Developmental/pathology , COS Cells , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Genes, Recessive , Humans , Immunohistochemistry , Phenotype , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Transfection
7.
Nature ; 458(7242): 1196-200, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19252479

ABSTRACT

Brachydactyly type A1 (BDA1) was the first recorded disorder of the autosomal dominant Mendelian trait in humans, characterized by shortened or absent middle phalanges in digits. It is associated with heterozygous missense mutations in indian hedgehog (IHH). Hedgehog proteins are important morphogens for a wide range of developmental processes. The capacity and range of signalling is thought to be regulated by its interaction with the receptor PTCH1 and antagonist HIP1. Here we show that a BDA1 mutation (E95K) in Ihh impairs the interaction of IHH with PTCH1 and HIP1. This is consistent with a recent paper showing that BDA1 mutations cluster in a calcium-binding site essential for the interaction with its receptor and cell-surface partners. Furthermore, we show that in a mouse model that recapitulates the E95K mutation, there is a change in the potency and range of signalling. The mice have digit abnormalities consistent with the human disorder.


Subject(s)
Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/metabolism , Mutation/genetics , Signal Transduction , Animals , Chickens , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Patched Receptors , Patched-1 Receptor , Protein Binding , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
8.
Gene Expr Patterns ; 9(4): 215-23, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19185060

ABSTRACT

Wnt signalling plays important roles in patterning and outgrowth of the vertebrate limb. Different mutations in Wnt genes, their antagonists or (co-)receptors result in patterning and outgrowth defects as well as chondrocyte and bone phenotypes in mouse and human. Understanding Wnt activity during mouse limb development and chondrogenesis requires a temporal and spatial overview of Wnt signalling key factor expression. Here we present a comparative expression analysis of all 19 Wnt genes and their major secreted antagonists of the Dickkopf (Dkk), Wisp and the secreted frizzled related protein (Sfrp) families during mouse limb development. Our study reveals new domains of expression for Wnt2, Wnt2b, Wnt5b, Wnt6, Wnt7b, Wnt9a, Wnt10a, Wnt10b, Wnt11 and Wnt16, in the limb. We also identified novel expression domains for the Wnt antagonists Sfrp1, Sfrp3, Sfrp5, Wisp1 as well as Dkk2 and Dkk3. We provide a full expression pattern for Wif1 in limb development, for which no limb expression had been documented so far.


Subject(s)
Cartilage/metabolism , Forelimb/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice/genetics , Wnt Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , CCN Intercellular Signaling Proteins , Cartilage/embryology , Chondrocytes/metabolism , Ectoderm/metabolism , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Extracellular Matrix Proteins/genetics , Forelimb/embryology , Glycoproteins/genetics , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mesoderm/metabolism , Mice/embryology , Oncogene Proteins/genetics , Proto-Oncogene Proteins , Time Factors , Wnt2 Protein
9.
J Clin Invest ; 119(1): 146-56, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19075394

ABSTRACT

Individuals with the birth defect synpolydactyly (SPD) have 1 or more digit duplicated and 2 or more digits fused together. One form of SPD is caused by polyalanine expansions in homeobox d13 (Hoxd13). Here we have used the naturally occurring mouse mutant that has the same mutation, the SPD homolog (Spdh) allele, and a similar phenotype, to investigate the molecular pathogenesis of SPD. A transgenic approach and crossing experiments showed that the Spdh allele is a combination of loss and gain of function. Here we identify retinaldehyde dehydrogenase 2 (Raldh2), the rate-limiting enzyme for retinoic acid (RA) synthesis in the limb, as a direct Hoxd13 target and show decreased RA production in limbs from Spdh/Spdh mice. Intrauterine treatment with RA restored pentadactyly in Spdh/Spdh mice. We further show that RA and WT Hoxd13 suppress chondrogenesis in mesenchymal progenitor cells, whereas Hoxd13 encoded by Spdh promotes cartilage formation in primary cells isolated from Spdh/Spdh limbs, and that this was associated with increased expression of Sox6/9. Increased Sox9 expression and ectopic cartilage formation in the interdigital mesenchyme of limbs from Spdh/Spdh mice suggest uncontrolled differentiation of these cells into the chondrocytic lineage. Thus, we propose that mutated Hoxd13 causes polydactyly in SPD by inducing extraneous interdigital chondrogenesis, both directly and indirectly, via a reduction in RA levels.


Subject(s)
Disease Models, Animal , Homeodomain Proteins , Mutation , Polydactyly/genetics , Syndactyly/genetics , Toes/abnormalities , Transcription Factors , Tretinoin/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Cells, Cultured , Chondrogenesis/physiology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Transgenic , Phenotype , Polydactyly/metabolism , Syndactyly/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/administration & dosage
10.
Development ; 135(9): 1713-23, 2008 May.
Article in English | MEDLINE | ID: mdl-18353862

ABSTRACT

Mutations in ROR2 result in a spectrum of genetic disorders in humans that are classified, depending on the nature of the mutation and the clinical phenotype, as either autosomal dominant brachydactyly type B (BDB, MIM 113000) or recessive Robinow syndrome (RRS, MIM 268310). In an attempt to model BDB in mice, the mutation W749X was engineered into the mouse Ror2 gene. In contrast to the human situation, mice heterozygous for Ror2(W749FLAG) are normal and do not develop brachydactyly, whereas homozygous mice exhibit features resembling RRS. Furthermore, both Ror2(W749FLAG/W749FLAG) and a previously engineered mutant, Ror2(TMlacZ/TMlacZ), lack the P2/P3 joint. Absence of Gdf5 expression at the corresponding interzone suggests that the defect is in specification of the joint. As this phenotype is absent in mice lacking the entire Ror2 gene, it appears that specification of the P2/P3 joint is affected by ROR2 activity. Finally, Ror2(W749FLAG/W749FLAG) mice survive to adulthood and exhibit phenotypes (altered body composition, reduced male fertility) not observed in Ror2 knockout mice, presumably due to the perinatal lethality of the latter. Therefore, Ror2(W749FLAG/W749FLAG) mice represent a postnatal model for RRS, provide insight into the mechanism of joint specification, and uncover novel roles of Ror2 in the mouse.


Subject(s)
Abnormalities, Multiple/genetics , Genes, Recessive , Musculoskeletal Abnormalities/genetics , Receptor Protein-Tyrosine Kinases/genetics , Abnormalities, Multiple/embryology , Animals , Body Mass Index , Bone Morphogenetic Proteins/metabolism , Fertility/genetics , Growth Differentiation Factor 5 , Humans , Joints/abnormalities , Joints/embryology , Limb Deformities, Congenital/embryology , Limb Deformities, Congenital/genetics , Male , Mice , Mice, Mutant Strains , Musculoskeletal Abnormalities/embryology , Mutation , Receptor Protein-Tyrosine Kinases/physiology , Receptor Tyrosine Kinase-like Orphan Receptors , Syndrome
11.
Dev Dyn ; 235(12): 3456-65, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17061261

ABSTRACT

Ror2 is a receptor tyrosine kinase mutated in the human syndromes Brachydactyly type B (BDB) and recessive Robinow syndrome (RS). In this study, we used the chick as a model to investigate the role of Ror2 in skeletogenesis and to elucidate the functional consequences of Ror2 mutations. For this purpose, we cloned chicken Ror2 and analyzed its expression pattern at various embryonic stages by in situ hybridization and immunolabeling. We document expression of cRor2 in several organs, including mesonephros, heart, nervous system, intestine and cartilage. The high conservation of expression when compared with the mouse underlines the validity of the chick as a model system. Using replication-competent retroviral vector-mediated overexpression, we analyzed the functional consequences of truncating BDB and RS mutations in the developing chick limb. Overexpression of Ror2 mutants led to a disturbance of growth plate architecture and a severe block of chondrocyte differentiation, demonstrating the functional importance of Ror2 in skeletogenesis.


Subject(s)
Bone Development/genetics , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Amino Acid Sequence , Animals , Base Sequence , Bone Development/physiology , Caenorhabditis elegans Proteins , Cell Differentiation , Chick Embryo , Chondrocytes/cytology , Chondrocytes/enzymology , Cloning, Molecular , DNA Probes/genetics , Gene Expression , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Limb Deformities, Congenital/enzymology , Limb Deformities, Congenital/genetics , Molecular Sequence Data , Phylogeny , Receptor Tyrosine Kinase-like Orphan Receptors , Sequence Deletion , Sequence Homology, Amino Acid , Syndrome
12.
Eur J Cell Biol ; 85(6): 519-28, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16584807

ABSTRACT

The high mobility group proteins A2 (HMGA2) have been implicated in the control of cell proliferation and differentiation, in particular during embryogenesis. Here, we used Xenopus laevis to analyze HMGA2 gene expression patterns during oogenesis and early embryogenesis. We found two functional XlHMGA2 isoforms, which we named XlHMGA2alpha and XlHMGA2beta. As revealed by RT-PCR, real-time PCR and whole-mount in situ hybridization both mRNAs are maternally produced and stored in eggs. Whole-mount in situ hybridizations revealed a conspicuous redistribution of the XlHMGA2 transcripts during early embryogenesis. Initially, during oogenesis and in eggs, the transcripts are uniformly distributed in the cytoplasm. With activation of the eggs the transcripts accumulate near the animal pole and remain in the juxtanuclear regions of animal pole blastomeres until midblastula transition. According to real-time PCR data, XlHMGA2alpha appears to be preferentially expressed during oogenesis and after midblastula transition, whereas XlHMGA2beta expression predominates after neurulation, suggesting an individual transcriptional regulation.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , HMGA2 Protein/genetics , Oogenesis/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Embryo, Nonmammalian/embryology , Gene Expression Profiling , HMGA2 Protein/chemistry , Models, Genetic , Molecular Sequence Data , Oocytes/cytology , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xenopus Proteins
SELECTION OF CITATIONS
SEARCH DETAIL