Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 290(12): 7506-16, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25657005

ABSTRACT

Myostatin, a member of the TGF-ß family of ligands, is a strong negative regulator of muscle growth. As such, it is a prime therapeutic target for muscle wasting disorders. Similar to other TGF-ß family ligands, myostatin is neutralized by binding one of a number of structurally diverse antagonists. Included are the antagonists GASP-1 and GASP-2, which are unique in that they specifically antagonize myostatin. However, little is known from a structural standpoint describing the interactions of GASP antagonists with myostatin. Here, we present the First low resolution solution structure of myostatin-free and myostatin-bound states of GASP-1 and GASP-2. Our studies have revealed GASP-1, which is 100 times more potent than GASP-2, preferentially binds myostatin in an asymmetrical 1:1 complex, whereas GASP-2 binds in a symmetrical 2:1 complex. Additionally, C-terminal truncations of GASP-1 result in less potent myostatin inhibitors that form a 2:1 complex, suggesting that the C-terminal domains of GASP-1 are the primary mediators for asymmetric complex formation. Overall, this study provides a new perspective on TGF-ß antagonism, where closely related antagonists can utilize different ligand-binding strategies.


Subject(s)
Carrier Proteins/metabolism , Myostatin/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , Chromatography, Gel , Cricetinae , Cricetulus , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Protein Binding , Scattering, Radiation , Surface Plasmon Resonance , Ultracentrifugation
2.
J Biol Chem ; 290(8): 4759-4771, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25561725

ABSTRACT

Bone morphogenetic proteins (BMPs) are antagonized through the action of numerous extracellular protein antagonists, including members from the differential screening-selected gene aberrative in neuroblastoma (DAN) family. In vivo, misregulation of the balance between BMP signaling and DAN inhibition can lead to numerous disease states, including cancer, kidney nephropathy, and pulmonary arterial hypertension. Despite this importance, very little information is available describing how DAN family proteins effectively inhibit BMP ligands. Furthermore, our understanding for how differences in individual DAN family members arise, including affinity and specificity, remains underdeveloped. Here, we present the structure of the founding member of the DAN family, neuroblastoma suppressor of tumorigenicity 1 (NBL1). Comparing NBL1 to the structure of protein related to Dan and Cerberus (PRDC), a more potent BMP antagonist within the DAN family, a number of differences were identified. Through a mutagenesis-based approach, we were able to correlate the BMP binding epitope in NBL1 with that in PRDC, where introduction of specific PRDC amino acids in NBL1 (A58F and S67Y) correlated with a gain-of-function inhibition toward BMP2 and BMP7, but not GDF5. Although NBL1(S67Y) was able to antagonize BMP7 as effectively as PRDC, NBL1(S67Y) was still 32-fold weaker than PRDC against BMP2. Taken together, this data suggests that alterations in the BMP binding epitope can partially account for differences in the potency of BMP inhibition within the DAN family.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 7/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/chemistry , Mutation, Missense , Proteins/chemistry , Amino Acid Substitution , Animals , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/genetics , CHO Cells , Cell Cycle Proteins , Cricetinae , Cricetulus , Cytokines , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mutagenesis , Protein Structure, Tertiary , Proteins/genetics , Structure-Activity Relationship
3.
J Biomol Screen ; 18(7): 837-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23543431

ABSTRACT

Myostatin, a member of the transforming growth factor (TGF)-ß family of secreted ligands, is a strong negative regulator of muscle growth. As such, therapeutic inhibitors of myostatin are actively being investigated for their potential in the treatment of muscle-wasting diseases such as muscular dystrophy and sarcopenia. Here, we sought to develop a high-throughput screening (HTS) method for small-molecule inhibitors that target myostatin. We created a HEK293 stable cell line that expresses the (CAGA)12-luciferase reporter construct and robustly responds to signaling of certain classes of TGF-ß family ligands. After optimization and miniaturization of the assay to a 384-well format, we successfully screened a library of compounds for inhibition of myostatin and the closely related activin A. Selection of some of the tested compounds was directed by in silico screening against myostatin, which led to an enrichment of target hits as compared with random selection. Altogether, we present an HTS method that will be useful for screening potential inhibitors of not only myostatin but also many other ligands of the TGF-ß family.


Subject(s)
Activins/antagonists & inhibitors , Growth Substances/pharmacology , Myostatin/antagonists & inhibitors , Computer Simulation , Gene Expression/drug effects , Genes, Reporter , HEK293 Cells , High-Throughput Screening Assays , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Small Molecule Libraries
4.
Mol Endocrinol ; 26(7): 1167-78, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22593183

ABSTRACT

Follistatin (FST)-type proteins are important antagonists of some members of the large TGF-ß family of cytokines. These include myostatin, an important negative regulator of muscle growth, and the closely related activin A, which is involved in many physiological functions, including maintenance of a normal reproductive axis. FST-type proteins, including FST and FST-like 3 (FSTL3), differentially inhibit various TGF-ß family ligands by binding each ligand with two FST-type molecules. In this study, we sought to examine features that are important for ligand antagonism by FST-type proteins. Previous work has shown that a modified construct consisting of the FST N-terminal domain (ND) followed by two repeating follistatin domains (FSD), herein called FST ND-FSD1-FSD1, exhibits strong specificity for myostatin over activin A. Using cell-based assays, we show that FST ND-FSD1-FSD1 is unique in its specificity for myostatin as compared with similar constructs containing domains from FSTL3 and that the ND is critical to its activity. Furthermore, we demonstrate that FSD3 of FST provides affinity to ligand inhibition and confers resistance to perturbations in the ND and FSD2, likely through the interaction of FSD3 of one FST molecule with the ND of the other FST molecule. Additionally, our data suggest that this contact provides cooperativity to ligand antagonism. Cross-linking studies show that this interaction also potentiates formation of 1:2 ligand-FST complexes, whereas lack of FSD3 allows formation of 1:1 complexes. Altogether, these studies support that domain differences generate FST-type molecules that are each uniquely suited ligand antagonists.


Subject(s)
Activins/chemistry , Follistatin-Related Proteins/chemistry , Follistatin/chemistry , Myostatin/chemistry , Activins/antagonists & inhibitors , Animals , Cell Line , Follistatin/metabolism , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , HEK293 Cells , Humans , Mice , Myostatin/antagonists & inhibitors , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary
5.
J Biol Chem ; 287(2): 1043-53, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22052913

ABSTRACT

TGF-ß family ligands are involved in a variety of critical physiological processes. For instance, the TGF-ß ligand myostatin is a staunch negative regulator of muscle growth and a therapeutic target for muscle-wasting disorders. Therefore, it is important to understand the molecular mechanisms of TGF-ß family regulation. One form of regulation is through inhibition by extracellular antagonists such as the follistatin (Fst)-type proteins. Myostatin is tightly controlled by Fst-like 3 (Fstl3), which is the only Fst-type molecule that has been identified in the serum bound to myostatin. Here, we present the crystal structure of myostatin in complex with Fstl3. The structure reveals that the N-terminal domain (ND) of Fstl3 interacts uniquely with myostatin as compared with activin A, because it utilizes different surfaces on the ligand. This results in conformational differences in the ND of Fstl3 that alter its position in the type I receptor-binding site of the ligand. We also show that single point mutations in the ND of Fstl3 are detrimental to ligand binding, whereas corresponding mutations in Fst have little effect. Overall, we have shown that the NDs of Fst-type molecules exhibit distinctive modes of ligand binding, which may affect overall affinity of ligand·Fst-type protein complexes.


Subject(s)
Follistatin-Related Proteins/chemistry , Models, Molecular , Myostatin/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Humans , Myostatin/genetics , Myostatin/metabolism , Point Mutation , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary
6.
Cell ; 129(5): 879-90, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17540169

ABSTRACT

The heritability of B cell chronic lymphocytic leukemia (CLL) is relatively high; however, no predisposing mutation has been convincingly identified. We show that loss or reduced expression of death-associated protein kinase 1 (DAPK1) underlies cases of heritable predisposition to CLL and the majority of sporadic CLL. Epigenetic silencing of DAPK1 by promoter methylation occurs in almost all sporadic CLL cases. Furthermore, we defined a disease haplotype, which segregates with the CLL phenotype in a large family. DAPK1 expression of the CLL allele is downregulated by 75% in germline cells due to increased HOXB7 binding. In the blood cells from affected family members, promoter methylation results in additional loss of DAPK1 expression. Thus, reduced expression of DAPK1 can result from germline predisposition, as well as epigenetic or somatic events causing or contributing to the CLL phenotype.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Down-Regulation , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Allelic Imbalance , Animals , Apoptosis , B-Lymphocytes/cytology , Chromosomes, Human, Pair 9 , DNA Methylation , DNA Mutational Analysis , Death-Associated Protein Kinases , Epigenesis, Genetic , Female , Germ-Line Mutation , Homeodomain Proteins/metabolism , Humans , Hybrid Cells , Jurkat Cells , Male , Mice , Pedigree , Point Mutation , Promoter Regions, Genetic , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...