Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 18341, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526551

ABSTRACT

Ligands of the transforming growth factor-ß (TGF-ß) superfamily are important targets for therapeutic intervention but present challenges because they signal combinatorially and exhibit overlapping activities in vivo. To obtain agents capable of sequestering multiple TGF-ß superfamily ligands with novel selectivity, we generated soluble, heterodimeric ligand traps by pairing the extracellular domain (ECD) of the native activin receptor type IIB (ActRIIB) alternately with the ECDs of native type I receptors activin receptor-like kinase 4 (ALK4), ALK7, or ALK3. Systematic analysis of these heterodimeric constructs by surface plasmon resonance, and comparison with their homodimeric counterparts, revealed that each type I receptor partner confers a distinct ligand-binding profile to the heterodimeric construct. Additional characterization in cell-based reporter gene assays confirmed that the heterodimeric constructs possessed different profiles of signaling inhibition in vitro, which translated into altered patterns of pharmacological activity when constructs were administered systemically to wild-type mice. Our results detail a versatile platform for the modular recombination of naturally occurring receptor domains, giving rise to inhibitory ligand traps that could aid in defining the physiological roles of TGF-ß ligand sets or be directed therapeutically to human diseases arising from dysregulated TGF-ß superfamily signaling.


Subject(s)
Activin Receptors/metabolism , Drug Discovery/methods , Protein Engineering/methods , Activin Receptors/chemistry , Activin Receptors/genetics , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Humans , Ligands , Mice , Mice, Inbred C57BL , Protein Binding , Protein Multimerization , Transforming Growth Factor beta/metabolism
2.
Bone ; 112: 71-89, 2018 07.
Article in English | MEDLINE | ID: mdl-29626545

ABSTRACT

Heterotopic ossification (HO), the pathological extraskeletal formation of bone, can arise from blast injuries, severe burns, orthopedic procedures and gain-of-function mutations in a component of the bone morphogenetic protein (BMP) signaling pathway, the ACVR1/ALK2 receptor serine-threonine (protein) kinase, causative of Fibrodysplasia Ossificans Progressiva (FOP). All three ALKs (-2, -3, -6) that play roles in bone morphogenesis contribute to trauma-induced HO, hence are well-validated pharmacological targets. That said, development of inhibitors, typically competitors of ATP binding, is inherently difficult due to the conserved nature of the active site of the 500+ human protein kinases. Since these enzymes are regulated via inherent plasticity, pharmacological chaperone-like drugs binding to another (allosteric) site could hypothetically modulate kinase conformation and activity. To test for such a mechanism, a surface pocket of ALK2 kinase formed largely by a key allosteric substructure was targeted by supercomputer docking of drug-like compounds from a virtual library. Subsequently, the effects of docked hits were further screened in vitro with purified recombinant kinase protein. A family of compounds with terminal hydrogen-bonding acceptor groups was identified that significantly destabilized the protein, inhibiting activity. Destabilization was pH-dependent, putatively mediated by ionization of a histidine within the allosteric substructure with decreasing pH. In vivo, nonnative proteins are degraded by proteolysis in the proteasome complex, or cellular trashcan, allowing for the emergence of therapeutics that inhibit through degradation of over-active proteins implicated in the pathology of diseases and disorders. Because HO is triggered by soft-tissue trauma and ensuing hypoxia, dependency of ALK destabilization on hypoxic pH imparts selective efficacy on the allosteric inhibitors, providing potential for safe prophylactic use.


Subject(s)
Activin Receptors/metabolism , Hypoxia/metabolism , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/prevention & control , Activin Receptors/chemistry , Allosteric Regulation , Animals , Binding Sites , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Drug Evaluation, Preclinical , Enzyme Stability , Humans , Hydrogen-Ion Concentration , Phosphorylation , Protein Structure, Secondary , Structure-Activity Relationship , Substrate Specificity , Tacrolimus Binding Protein 1A/metabolism , Temperature
3.
Biomater Sci ; 4(2): 346-54, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26626506

ABSTRACT

C16-YEALRVANEVTLN, a peptide amphiphile (PA) incorporating a biologically active amino acid sequence found in lumican, has been examined for its influence upon collagen synthesis by human corneal fibroblasts in vitro, and the roles of supra-molecular assembly and activin receptor-like kinase ALK receptor signaling in this effect were assessed. Cell viability was monitored using the Alamar blue assay, and collagen synthesis was assessed using Sirius red. The role of ALK signaling was studied by receptor inhibition. Cultured human corneal fibroblasts synthesized significantly greater amounts of collagen in the presence of the PA over both 7-day and 21-day periods. The aggregation of the PA to form nanotapes resulted in a notable enhancement in this activity, with an approximately two-fold increase in collagen production per cell. This increase was reduced by the addition of an ALK inhibitor. The data presented reveal a stimulatory effect upon collagen synthesis by the primary cells of the corneal stroma, and demonstrate a direct influence of supra-molecular assembly of the PA upon the cellular response observed. The effects of PA upon fibroblasts were dependent upon ALK receptor function. These findings elucidate the role of self-assembled nanostructures in the biological activity of peptide amphiphiles, and support the potential use of a self-assembling lumican derived PA as a novel biomaterial, intended to promote collagen deposition for wound repair and tissue engineering purposes.


Subject(s)
Activin Receptors/chemistry , Biocompatible Materials/chemistry , Collagen/chemical synthesis , Corneal Stroma/chemistry , Fibroblasts/chemistry , Lumican/chemistry , Peptides/chemistry , Activin Receptors/metabolism , Amino Acid Sequence , Biocompatible Materials/metabolism , Cell Survival , Collagen/chemistry , Corneal Stroma/drug effects , Fibroblasts/drug effects , Humans , Lumican/metabolism , Lumican/pharmacology , Peptides/metabolism , Peptides/pharmacology , Signal Transduction , Tissue Engineering
4.
Trends Endocrinol Metab ; 25(9): 464-71, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25042839

ABSTRACT

The transforming growth factor beta (TGFß) superfamily comprises a large number of secreted proteins that regulate various fundamental biological processes underlying embryonic development and the postnatal regulation of many cell types and organs. Sequence similarities define two ligand subfamilies: the TGFß/activin subfamily and the bone morphogenetic protein (BMP) subfamily. The discovery that myostatin, a member of the TGFß/activin subfamily, negatively controls muscle mass attracted attention to this pathway. However, recent findings of a positive role for BMP-mediated signaling in muscle have challenged the model of how the TGFß network regulates skeletal muscle phenotype. This review illustrates how this complex network integrates crosstalk among members of the TGFß superfamily and downstream signaling elements to regulate muscle in health and disease.


Subject(s)
Bone Morphogenetic Protein Receptors/agonists , Bone Morphogenetic Proteins/metabolism , Models, Biological , Muscle, Skeletal/metabolism , Receptors, Transforming Growth Factor beta/agonists , Signal Transduction , Transforming Growth Factor beta/metabolism , Activin Receptors/chemistry , Activin Receptors/genetics , Activin Receptors/metabolism , Activins/genetics , Activins/metabolism , Animals , Autophagy , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Humans , Hypertrophy , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics
5.
Vitam Horm ; 85: 1-27, 2011.
Article in English | MEDLINE | ID: mdl-21353873

ABSTRACT

The biological responses of the transforming growth factor-ß (TGF-ß) superfamily, which includes Activins and Nodal, are induced by activation of a receptor complex and Smads. A type I receptor, which is a component of the complex, is known as an activin receptor-like kinase (ALK); currently seven ALKs (ALK1-ALK7) have been identified in humans. Activins signaling, which is mediated by ALK4 and 7 together with ActRIIA and IIB, plays a critical role in glucose-stimulated insulin secretion, development/neogenesis, and glucose homeostatic control of pancreatic endocrine cells; the insulin gene is regulated by these signaling pathways via ALK7, which is a receptor for Activins AB and B and Nodal. This review discusses signal transduction of ALKs in pancreatic endocrine cells and the role of ALKs in insulin gene regulation.


Subject(s)
Activin Receptors/physiology , Insulin/metabolism , Activin Receptors/chemistry , Animals , Gene Expression Regulation , Humans , Insulin/genetics , Isoenzymes/chemistry , Isoenzymes/physiology , Pancreas/metabolism
6.
Expert Opin Investig Drugs ; 19(8): 995-1005, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20629616

ABSTRACT

IMPORTANCE OF THE FIELD: Anabolic therapy, or stimulating the function of bone-forming osteoblasts, is the preferred pharmacological intervention for osteoporosis. AREAS COVERED IN THIS REVIEW: We reviewed bone anabolic agents currently under active investigation. The bone anabolic potential of IGF-I and parathyroid hormone-related protein is discussed in the light of animal data and human studies. We also discuss the use of antagonists of the calcium-sensing receptor (calcilytics) as orally administered small molecules capable of transiently elevating serum parathyroid hormone (PTH). Further, we reviewed novel anabolic agents targeting members of the wingless tail (Wnt) signaling family that regulate bone formation including DKK-1, sclerostin, Thp1, and glycogen synthase kinase 3beta. We have also followed up on the promise shown by beta-blockers in modulating the activity of sympathetic nervous system, thus affecting bone anabolism. We give critical consideration to neutralizing the activity of activin A, a negative regulator of bone mass by soluble activin receptor IIA, as a strategy to promote bone formation. WHAT THE READER WILL GAIN: Update on various strategies to promote osteoblast function currently under evaluation. TAKE HOME MESSAGE: In spite of favorable results in experimental models, none of these strategies has yet achieved the ultimate goal of providing an alternative to injectable PTH, the sole anabolic therapy in clinical use.


Subject(s)
Anabolic Agents/therapeutic use , Bone Density/drug effects , Drugs, Investigational , Insulin-Like Growth Factor I/therapeutic use , Osteoporosis/drug therapy , Activin Receptors/chemistry , Activin Receptors/metabolism , Adrenergic beta-Antagonists/chemistry , Adrenergic beta-Antagonists/metabolism , Adrenergic beta-Antagonists/therapeutic use , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Female , Humans , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/metabolism , Male , Osteoporosis/physiopathology , Parathyroid Hormone-Related Protein/chemistry , Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone-Related Protein/therapeutic use , Receptors, Calcium-Sensing/antagonists & inhibitors , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Signal Transduction/drug effects , Wnt1 Protein/metabolism
7.
Comp Biochem Physiol B Biochem Mol Biol ; 156(3): 158-67, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20226263

ABSTRACT

The signal transduction mechanisms in mollusks are still elusive since the genome information is incomplete and cell lines are not available. In previous study, we cloned a highly conserved Smad3 homolog (designated as Pf-Smad3) from the pearl oyster, Pinctada fucata. It seems that transforming growth factor beta (TGFbeta) signaling may play similar roles in the oyster as in vertebrate. Here we report a cDNA encoding an activin like receptor 1 homolog (designated as Pf-ALR1) of the oyster, another kind of TGFbeta superfamily member. Compared to the activin receptor-like kinases (ALK) in human, the amino acid sequence of Pf-ALR1 is more similar to that of ALK1, especially the L45 loop. Reverse transcription-polymerase chain reaction results indicate that Pf-ALR1 mRNA is expressed ubiquitously in the adult oyster. Thus, Pf-ALR1 may be important for many physiological processes in the oyster. To lay a basis for further investigation of the TGFbeta signal pathway functions in the oyster shell formation, in this report, the Pf-ALR1 mRNA expression in the oyster mantle was detected by in situ hybridization. The results show that Pf-ALR1 in the oyster mantle is mainly expressed at the inner epithelial cells of the outer fold and the outer epithelial cells of the middle fold, similarly as Pf-Smad3. The mRNA levels of Pf-ALR1 and Pf-Smad3 are all changed after shell notching. These results indicate that both Pf-ALR1 and Pf-Smad3 may take part in shell formation and repair. The results of drug treatment experiments with in-vitro cultured oyster mantle tissue cells demonstrate that the mRNA expression levels of Pf-Smad3, Pf-ALR1 and two oyster nuclear factor-kappaB (NF-kB) members can be adjusted and correlated. All our observations suggest that there should be similar TGFbeta signal pathways in the oyster and vertebrate. However, the potential functions of Pf-ALR1 and the relations of TGFbeta and NF-kB members in the oyster all need to be thoroughly investigated.


Subject(s)
Activin Receptors/genetics , Pinctada/genetics , Activin Receptors/chemistry , Activin Receptors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/chemistry , Gene Expression , Molecular Sequence Data , Pinctada/metabolism
8.
BMC Biol ; 7: 59, 2009 Sep 07.
Article in English | MEDLINE | ID: mdl-19735544

ABSTRACT

BACKGROUND: Transforming growth factor (TGF)beta superfamily members transduce signals by oligomerizing two classes of serine/threonine kinase receptors, termed type I and type II. In contrast to the large number of ligands only seven type I and five type II receptors have been identified in mammals, implicating a prominent promiscuity in ligand-receptor interaction. Since a given ligand can usually interact with more than one receptor of either subtype, differences in binding affinities and specificities are likely important for the generation of distinct ligand-receptor complexes with different signaling properties. RESULTS: In vitro interaction analyses showed two different prototypes of binding kinetics, 'slow on/slow off' and 'fast on/fast off'. Surprisingly, the binding specificity of ligands to the receptors of one subtype is only moderate. As suggested from the dimeric nature of the ligands, binding to immobilized receptors shows avidity due to cooperative binding caused by bivalent ligand-receptor interactions. To compare these in vitro observations to the situation in vivo, binding studies on whole cells employing homodimeric as well as heterodimeric bone morphogenetic protein 2 (BMP2) mutants were performed. Interestingly, low and high affinity binding sites were identified, as defined by the presence of either one or two BMP receptor (BMPR)-IA receptor chains, respectively. Both sites contribute to different cellular responses in that the high affinity sites allow a rapid transient response at low ligand concentrations whereas the low affinity sites facilitate sustained signaling but higher ligand concentrations are required. CONCLUSION: Binding of a ligand to a single high affinity receptor chain functioning as anchoring molecule and providing sufficient complex stability allows the subsequent formation of signaling competent complexes. Another receptor of the same subtype, and up to two receptors of the other subtype, can then be recruited. Thus, the resulting receptor arrangement can principally consist of four different receptors, which is consistent with our interaction analysis showing low ligand-receptor specificity within one subtype class. For BMP2, further complexity is added by the fact that heterooligomeric signaling complexes containing only one type I receptor chain can also be found. This indicates that despite prominent ligand receptor promiscuity a manifold of diverse signals might be generated in this receptor limited system.


Subject(s)
Bone Morphogenetic Protein Receptors/chemistry , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factor 5/metabolism , Protein Interaction Domains and Motifs/physiology , Activin Receptors/chemistry , Activin Receptors/genetics , Activin Receptors/isolation & purification , Activin Receptors/metabolism , Activins/chemistry , Activins/genetics , Activins/isolation & purification , Activins/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Binding Sites , Biosensing Techniques , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/isolation & purification , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/isolation & purification , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cholic Acids/chemistry , Detergents/chemistry , Growth Differentiation Factor 5/chemistry , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/isolation & purification , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Kinetics , Ligands , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Isoforms , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection
9.
Mech Dev ; 126(11-12): 950-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19766717

ABSTRACT

How TGF-beta-type ligands achieve signaling specificity during development is only partially understood. Here, we show that Dawdle, one of four Activin-type ligands in Drosophila, preferentially signals through Babo(c), one of three isoforms of the Activin Type-I receptor that are expressed during development. In cell culture, Dawdle signaling is active in the presence of the Type-II receptor Punt but not Wit, demonstrating that the Type-II receptor also contributes to the specificity of the signaling complex. During development, different larval tissues express unique combinations of these receptors, and ectopic expression of Babo(c) in a tissue where it is not normally expressed at high levels can make that tissue sensitive to Dawdle signaling. These results reveal a mechanism by which distinct cell types can discriminate between different Activin-type signals during development as a result of differential expression of Type-I receptor isoforms.


Subject(s)
Activin Receptors/metabolism , Activins/metabolism , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Signal Transduction , Activin Receptors/chemistry , Activin Receptors/genetics , Activin Receptors, Type II/metabolism , Amino Acid Sequence , Animals , Body Patterning , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Ligands , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Cell Surface/metabolism , Wings, Animal/embryology , Wings, Animal/metabolism
10.
J Biol Chem ; 283(47): 32831-8, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18768470

ABSTRACT

Transforming growth factor beta family ligands are neutralized by a number of structurally divergent antagonists. Follistatin-type antagonists, which include splice variants of follistatin (FS288 and FS315) and follistatin-like 3 (FSTL3), have high affinity for activin A but differ in their affinity for other ligands, particularly bone morphogenetic proteins. To understand the structural basis for ligand specificity within FS-type antagonists, we determined the x-ray structure of activin A in complex with FSTL3 to a resolution of 2.5 A. Similar to the previously resolved FS.activin A structures, the ligand is encircled by two antagonist molecules blocking all ligand receptor-binding sites. Recently, the significance of the FS N-terminal domain interaction at the ligand type I receptor site has been questioned; however, our data show that for FSTL3, the N-terminal domain forms a more intimate contact with activin A, implying that this interaction is stronger than that for FS. Furthermore, binding studies revealed that replacing the FSTL3 N-terminal domain with the corresponding FS domain considerably lowers activin A affinity. Therefore, both structural and biochemical evidence support a significant interaction of the N-terminal domain of FSTL3 with activin A. In addition, structural comparisons with bone morphogenetic proteins suggest that the interface where the N-terminal domain binds may be the key site for determining FS-type antagonist specificity.


Subject(s)
Activins/chemistry , Follistatin-Related Proteins/chemistry , Follistatin/chemistry , Activin Receptors/chemistry , Binding Sites , Cell Line , Crystallography, X-Ray , Electrons , Humans , Ligands , Models, Molecular , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Surface Properties
11.
Endocr J ; 55(1): 11-21, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17878607

ABSTRACT

Activin, myostatin and other members of the TGF-beta superfamily signal through a combination of type II and type I receptors, both of which are transmembrane serine/threonine kinases. Activin type II receptors, ActRIIA and ActRIIB, are primary ligand binding receptors for activins, nodal, myostatin and GDF11. ActRIIs also bind a subset of bone morphogenetic proteins (BMPs). Type I receptors that form complexes with ActRIIs are dependent on ligands. In the case of activins and nodal, activin receptor-like kinases 4 and 7 (ALK4 and ALK7) are the authentic type I receptors. Myostatin and GDF11 utilize ALK5, although ALK4 could also be activated by these growth factors. ALK4, 5 and 7 are structurally and functionally similar and activate receptor-regulated Smads for TGF-beta, Smad2 and 3. BMPs signal through a combination of three type II receptors, BMPRII, ActRIIA, and ActRIIB and four type I receptors, ALK1, 2, 3, and 6. BMPs activate BMP-specific Smads, Smad1, 5 and 8. Smad proteins undergo multimerization with co-mediator Smad, Smad4, and translocated into the nucleus to regulate the transcription of target genes in cooperation with nuclear cofactors. The signal transduction pathway through activin type II receptors, ActRIIA and ActRIIB, with type I receptors is involved in various human diseases. In this review, we discuss the role of signaling through activin receptors as therapeutic targets of intractable neuromuscular diseases, endocrine disorders and cancers.


Subject(s)
Activin Receptors/metabolism , Musculoskeletal Diseases/drug therapy , Neoplasms/drug therapy , Activin Receptors/antagonists & inhibitors , Activin Receptors/chemistry , Activin Receptors/physiology , Activins/physiology , Animals , Antineoplastic Agents/administration & dosage , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Drug Delivery Systems , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/physiology , GPI-Linked Proteins , Humans , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Models, Biological , Myostatin , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Protein Binding , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta/physiology
12.
Article in English | MEDLINE | ID: mdl-16611164

ABSTRACT

The transforming growth factor-beta (TGF-beta) superfamily regulates a multitude of cellular processes from fertilization to adulthood in vertebrates. Signaling by the TGF-beta superfamily occurs via formation of heteromeric complexes consisting of type I and type II receptors. The type I receptors, referred to as activin receptor-like kinases (ALK), lie at the epicenter of the signaling cascade as they transduce TGF-beta signals to intracellular regulators of transcription known as Smad proteins. Currently, seven ALKs have been identified in mammals. Structurally, ALKs possess an extracellular binding domain, a transmembrane domain, a GS domain that serves as the site of activation by type II receptors, and a kinase domain that activates downstream signaling molecules. ALKs mediate the effect of TGF-beta superfamily on a variety of cellular processes such as proliferation, differentiation, apoptosis, adhesion and migration, and therefore play important roles in many biological processes. Some ALKs have been implicated in several disorders, including tumorigenesis, hemorrhagic telangiectasia (HHT), immune and renal diseases, and skeletal malfunctions, suggesting that these receptors can be used as drug targets.


Subject(s)
Activin Receptors/physiology , Phosphotransferases/physiology , Activin Receptors/antagonists & inhibitors , Activin Receptors/chemistry , Animals , Humans , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/chemistry , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/physiology
13.
J Biol Chem ; 280(48): 40177-86, 2005 Dec 02.
Article in English | MEDLINE | ID: mdl-16186117

ABSTRACT

Within the transforming growth factor beta superfamily, the agonist-antagonist relationship between activin and inhibin is unique and critical to integrated reproductive function. Activin acts in the pituitary to stimulate follicle-stimulating hormone, and is antagonized by endocrine acting, gonadally derived inhibin. We have undertaken a mutational analysis of the activin betaA subunit to determine the precise structural aspects that contribute to inhibin antagonism of activin. By substituting specific amino acid residues in the activin betaA subunit with similarly aligned amino acids from the alpha subunit, we have pinpointed the residues required for activin receptor binding and activity, as well as for inhibin antagonism of activin through its receptors. Additionally, we have identified an activin mutant with a higher affinity for the activin type I receptor that provides structural evidence for the evolution of ligand-receptor interactions within the transforming growth factor beta superfamily.


Subject(s)
Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/physiology , Activin Receptors/chemistry , Activins/antagonists & inhibitors , Activins/chemistry , Amino Acid Sequence , Animals , Binding, Competitive , Blotting, Western , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , DNA Mutational Analysis , Glycosylation , Immunoprecipitation , Inhibin-beta Subunits/chemistry , Inhibins/chemistry , Ligands , Luciferases/metabolism , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Multigene Family , Mutagenesis , Mutation , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta/chemistry
14.
J Mol Recognit ; 18(5): 385-403, 2005.
Article in English | MEDLINE | ID: mdl-15948132

ABSTRACT

The structure-function properties of the pleiotropic activins and their relationship to other members of the transforming growth factor-beta superfamily of proteins are described. In order to highlight the molecular promiscuity of these growth factors, emphasis has been placed on molecular features associated with the recognition by activin A and the bone morphogenic proteins of the corresponding extracellular domains of the ActRI and ActRII receptors. The available evidence suggests that the homodimeric activin A in its various functional roles has the propensity to fulfill key tasks in the regulation of mammalian cell behaviour, through coordination of numerous transcriptional and translational processes. Because of these profound effects, under physiologically normal conditions, activin A levels are closely controlled by a variety of binding partners, such as follistatin-288 and follistatin-315, alpha(2)-macroglobulin and other proteins. Moreover, the subunits of other members of the activin subfamily, such as activin B or activin C, are able to form heterodimers with the activin A subunit, thus providing a further avenue to positively or negatively control the physiological concentrations of activin A that are available for interaction with specific receptors and induction of cell signaling events. Based on data from X-ray crystallographic studies and homology modeling experiments, the molecular architecture of the ternary receptor-activin ligand complexes has been dissected, permitting rationalization in structural terms of the pattern of interactions that are the hallmark of this protein family.


Subject(s)
Activin Receptors/chemistry , Activins/chemistry , Bone Morphogenetic Proteins/chemistry , Inhibin-beta Subunits/chemistry , Receptors, Transforming Growth Factor beta/chemistry , Activin Receptors/metabolism , Activins/metabolism , Amino Acid Sequence , Animals , Bone Morphogenetic Proteins/metabolism , Crystallography, X-Ray , Inhibin-beta Subunits/metabolism , Ligands , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Structure-Activity Relationship
15.
J Am Soc Mass Spectrom ; 16(1): 38-45, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15653362

ABSTRACT

Limited proteolysis is an important and widely used method for analyzing the tertiary structure and determining the domain boundaries of proteins. Here we describe a novel method for determining the N- and C-terminal boundary amino acid sequences of products derived from limited proteolysis using semi-specific and/or non-specific enzymes, with mass spectrometry as the only analytical tool. The core of this method is founded on the recognition that cleavage of proteins with non-specific proteases is not random, but patterned. Based on this recognition, we have the ability to determine the sequence of each proteolytic fragment by extracting a common association between data sets containing multiple potential sequences derived from two or more different mass spectral molecular weight measurements. Proteolytic product sequences derived from specific and non-specific enzymes can be accurately determined without resorting to the conventional time-consuming and laborious methods of SDS-PAGE and N-terminal sequencing analysis. Because of the sensitivity of mass spectrometry, multiple transient proteolysis intermediates can also be identified and analyzed by this method, which allows the ability to monitor the progression of proteolysis and thereby gain insight into protein structures.


Subject(s)
Activin Receptors/chemistry , Peptide Fragments/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Amino Acid Sequence , Chromatography, High Pressure Liquid , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Protein Structure, Tertiary , Thermolysin/chemistry
16.
J Biol Chem ; 279(51): 53126-35, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15475360

ABSTRACT

Activins and inhibins compose a heterogeneous subfamily within the transforming growth factor-beta (TGF-beta) superfamily of growth and differentiation factors with critical biological activities in embryos and adults. They signal through a heteromeric complex of type II, type I, and for inhibin, type III receptors. To characterize the affinity, specificity, and activity of these receptors (alone and in combination) for the inhibin/activin subfamily, we developed a cell-free assay system using soluble receptor-Fc fusion proteins. The soluble activin type II receptor (sActRII)-Fc fusion protein had a 7-fold higher affinity for activin A compared with sActRIIB-Fc, whereas both receptors had a marked preference for activin A over activin B. Although inhibin A and B binding was 20-fold lower compared with activin binding to either type II receptor alone, the mixture of either type II receptor with soluble TGF-beta type III receptor (TbetaRIII; betaglycan)-Fc reconstituted a soluble high affinity inhibin receptor. In contrast, mixing either soluble activin type II receptor with soluble activin type I receptors did not substantially enhance activin binding. Our results support a cooperative model of binding for the inhibin receptor (ActRII.sTbetaRIII complex) but not for activin receptors (type II + type I) and demonstrate that a complex composed of activin type II receptors and TbetaRIII is both necessary and sufficient for high affinity inhibin binding. This study also illustrates the utility of this cell-free system for investigating hypotheses of receptor complex mechanisms resulting from crystal structure analyses.


Subject(s)
Activin Receptors/chemistry , Inhibins/chemistry , Cell Line , Cell-Free System , Crystallography, X-Ray , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Glycosylation , Humans , Iodine/chemistry , Kinetics , Ligands , Protein Binding , Protein Structure, Tertiary , Receptors, Fc/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Transforming Growth Factor beta/metabolism
17.
Dev Growth Differ ; 45(2): 113-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12752499

ABSTRACT

Studies have shown that the activin type IB receptor is specific for activin/nodal signaling. Activin is produced by follicle cells in the ovary, and is incorporated into the oocytes. Antisera against three peptides were prepared, encompassing the extracellular, intracellular and serine/threonine kinase domains of the Xenopus type IB activin receptor (XALK4). Immunocytochemistry was done using these antisera to investigate the distribution of XALK4 in the Xenopus ovary. All three antisera stained the mitochondrial cloud of Xenopus previtellogenic oocytes. Purified antibody against the intracellular domain also recognized the mitochondrial cloud. Immunoelectron microscopy localized XALK4 on the endoplasmic reticulum of the mitochondrial cloud, although not on mitochondria.


Subject(s)
Activin Receptors, Type II/analysis , Activin Receptors/analysis , Ovary/cytology , Xenopus Proteins/analysis , Xenopus/physiology , Activin Receptors/chemistry , Activin Receptors/genetics , Activin Receptors, Type I , Activin Receptors, Type II/chemistry , Activin Receptors, Type II/genetics , Amino Acid Sequence , Animals , Binding Sites , Female , Immunohistochemistry , Microscopy, Immunoelectron , Molecular Sequence Data , Oocytes/cytology , Peptide Fragments/chemistry , RNA/genetics , Xenopus/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...