Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(7): 102076, 2022 07.
Article in English | MEDLINE | ID: mdl-35643319

ABSTRACT

BMPR2 is a type II Transforming Growth Factor (TGF)-ß family receptor that is fundamentally associated with pulmonary arterial hypertension (PAH) in humans. BMPR2 shares functional similarities with the type II activin receptors ACVR2A and ACVR2B, as it interacts with an overlapping group of TGF-ß family growth factors (GFs). However, how BMPR2 recognizes GFs remains poorly understood. Here, we solved crystal structures of BMPR2 in complex with the GF activin B and of ACVR2A in complex with the related GF activin A. We show that both BMPR2 and ACVR2A bind GFs with nearly identical geometry using a conserved hydrophobic hot spot, while differences in contacting residues are predominantly found in loop areas. Upon further exploration of the GF-binding spectrum of the two receptors, we found that although many GFs bind both receptors, the high-affinity BMPR2 GFs comprise BMP15, BMP10, and Nodal, whereas those of ACVR2A are activin A, activin B, and GDF11. Lastly, we evaluated GF-binding domain BMPR2 variants found in human PAH patients. We demonstrate that mutations within the GF-binding interface resulted in loss of GF binding, while mutations in loop areas allowed BMPR2 to retain the ability to bind cognate GFs with high affinity. In conclusion, the in vitro activities of BMPR2 variants and the crystal structures reported here indicate biochemically relevant complexes that explain how some GF-binding domain variants can lead to PAH.


Subject(s)
Activin Receptors, Type II/metabolism , Bone Morphogenetic Protein Receptors, Type II , Activin Receptors/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Proteins/genetics , Growth Differentiation Factors , Humans , Mutation , Transforming Growth Factor beta/genetics
2.
Commun Biol ; 5(1): 157, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197550

ABSTRACT

Fortilin is a 172-amino acid multifunctional protein present in both intra- and extracellular spaces. Although fortilin binds and regulates various cellular proteins, the biological role of extracellular fortilin remains unknown. Here we report that fortilin specifically interacts with TGF-ß1 and prevents it from activating the TGF-ß1 signaling pathway. In a standard immunoprecipitation-western blot assay, fortilin co-immunoprecipitates TGF-ß1 and its isoforms. The modified ELISA assay shows that TGF-ß1 remains complexed with fortilin in human serum. Both bio-layer interferometry and surface plasmon resonance (SPR) reveal that fortilin directly bind TGF-ß1. The SPR analysis also reveals that fortilin and the TGF-ß receptor II (TGFßRII) compete for TGF-ß1. Both luciferase and secreted alkaline phosphatase reporter assays show that fortilin prevents TGF-ß1 from activating Smad3 binding to Smad-binding element. Fortilin inhibits the phosphorylation of Smad3 in both quantitative western blot assays and ELISA. Finally, fortilin inhibits TGFß-1-induced differentiation of C3H10T1/2 mesenchymal progenitor cells to smooth muscle cells. A computer-assisted virtual docking reveals that fortilin occupies the pocket of TGF-ß1 that is normally occupied by TGFßRII and that TGF-ß1 can bind either fortilin or TGFßRII at any given time. These data support the role of extracellular fortilin as a negative regulator of the TGF-ß1 signaling pathway.


Subject(s)
Receptors, Transforming Growth Factor beta , Transforming Growth Factor beta1 , Tumor Protein, Translationally-Controlled 1 , Humans , Phosphorylation , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Tumor Protein, Translationally-Controlled 1/metabolism
3.
Mol Biotechnol ; 64(2): 156-170, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34550550

ABSTRACT

Recombinant human BMP-4 growth factor (GF) has significant commercial potential as therapeutic for regenerating bone and as cell culture supplement. However, its commercial utility has been limited as large-scale attempts to express and purify human BMP-4 GF have proved challenging. We have established a novel approach to obtain significant quantities of pure and bioactive BMP-4 GF from Chinese hamster ovary cell cultures by extracting the GF moiety from the extracellular matrix or cell pellet fraction. This approach increased yields approximately one 100-fold over BMP-4 GF purified from CM. The molecular activities of the two fractions are indistinguishable. We further analyzed binding of BMP-4 GF to the proteoglycan Heparin and showed that an N-terminal basic sequence is essential for this interaction. Taken together, these results provide novel insights into the purification, localization, and Heparin binding of human BMP-4 that have implications for its bioprocessing and biological function.


Subject(s)
Bone Morphogenetic Protein 4/isolation & purification , Bone Morphogenetic Protein 4/metabolism , Heparin/metabolism , Activin Receptors, Type II/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , CHO Cells , Cricetulus , Extracellular Matrix/chemistry , Hep G2 Cells , Humans , Protein Engineering/methods , Protein Multimerization , Surface Plasmon Resonance
4.
Int J Mol Sci ; 22(16)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34445177

ABSTRACT

Adipose tissues (AT) expand in response to energy surplus through adipocyte hypertrophy and hyperplasia. The latter, also known as adipogenesis, is a process by which multipotent precursors differentiate to form mature adipocytes. This process is directed by developmental cues that include members of the TGF-ß family. Our goal here was to elucidate, using the 3T3-L1 adipogenesis model, how TGF-ß family growth factors and inhibitors regulate adipocyte development. We show that ligands of the Activin and TGF-ß families, several ligand traps, and the SMAD1/5/8 signaling inhibitor LDN-193189 profoundly suppressed 3T3-L1 adipogenesis. Strikingly, anti-adipogenic traps and ligands engaged the same mechanism of action involving the simultaneous activation of SMAD2/3 and inhibition of SMAD1/5/8 signaling. This effect was rescued by the SMAD2/3 signaling inhibitor SB-431542. By contrast, although LDN-193189 also suppressed SMAD1/5/8 signaling and adipogenesis, its effect could not be rescued by SB-431542. Collectively, these findings reveal the fundamental role of SMAD1/5/8 for 3T3-L1 adipogenesis, and potentially identify a negative feedback loop that links SMAD2/3 activation with SMAD1/5/8 inhibition in adipogenic precursors.


Subject(s)
Adipogenesis , Smad2 Protein/metabolism , Smad3 Protein/metabolism , 3T3-L1 Cells , Animals , Mice , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism
5.
Cytokine Growth Factor Rev ; 57: 39-54, 2021 02.
Article in English | MEDLINE | ID: mdl-33087301

ABSTRACT

The transforming growth factor (TGF)-ß family is a group of structurally related, multifunctional growth factors, or ligands that are crucially involved in the development, regulation, and maintenance of animal tissues. In humans, the family counts over 33 members. These secreted ligands typically form multimeric complexes with two type I and two type II receptors to activate one of two distinct signal transduction branches. A striking feature of the family is its promiscuity, i.e., many ligands bind the same receptors and compete with each other for binding to these receptors. Although several explanations for this feature have been considered, its functional significance has remained puzzling. However, several recent reports have promoted the idea that ligand-receptor binding promiscuity and competition are critical features of the TGF-ß family that provide an essential regulating function. Namely, they allow a cell to read and process multi-ligand inputs. This capability may be necessary for producing subtle, distinctive, or adaptive responses and, possibly, for facilitating developmental plasticity. Here, we review the molecular basis for ligand competition, with emphasis on molecular structures and binding affinities. We give an overview of methods that were used to establish experimentally ligand competition. Finally, we discuss how the concept of ligand competition may be fundamentally tied to human physiology, disease, and therapy.


Subject(s)
Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Humans , Ligands , Protein Binding , Receptors, Transforming Growth Factor beta/metabolism
6.
Elife ; 92020 06 09.
Article in English | MEDLINE | ID: mdl-32515349

ABSTRACT

Activin A functions in BMP signaling in two ways: it either engages ACVR1B to activate Smad2/3 signaling or binds ACVR1 to form a non-signaling complex (NSC). Although the former property has been studied extensively, the roles of the NSC remain unexplored. The genetic disorder fibrodysplasia ossificans progressiva (FOP) provides a unique window into ACVR1/Activin A signaling because in that disease Activin can either signal through FOP-mutant ACVR1 or form NSCs with wild-type ACVR1. To explore the role of the NSC, we generated 'agonist-only' Activin A muteins that activate ACVR1B but cannot form the NSC with ACVR1. Using one of these muteins, we demonstrate that failure to form the NSC in FOP results in more severe disease pathology. These results provide the first evidence for a biological role for the NSC in vivo and pave the way for further exploration of the NSC's physiological role in corresponding knock-in mice.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Myositis Ossificans/genetics , Signal Transduction/genetics , Activin Receptors, Type I/genetics , Activins/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Proteins/genetics , Gene Knock-In Techniques , Mice , Mice, Transgenic , Mutation , Myositis Ossificans/pathology
7.
Biomolecules ; 10(4)2020 03 29.
Article in English | MEDLINE | ID: mdl-32235336

ABSTRACT

Activins belong to the transforming growth factor (TGF)-ß family of multifunctional cytokines and signal via the activin receptors ALK4 or ALK7 to activate the SMAD2/3 pathway. In some cases, activins also signal via the bone morphogenetic protein (BMP) receptor ALK2, causing activation of the SMAD1/5/8 pathway. In this study, we aimed to dissect how activin A and activin B homodimers, and activin AB and AC heterodimers activate the two main SMAD branches. We compared the activin-induced signaling dynamics of ALK4/7-SMAD2/3 and ALK2-SMAD1/5 in a multiple myeloma cell line. Signaling via the ALK2-SMAD1/5 pathway exhibited greater differences between ligands than signaling via ALK4/ALK7-SMAD2/3. Interestingly, activin B and activin AB very potently activated SMAD1/5, resembling the activation commonly seen with BMPs. As SMAD1/5 was also activated by activins in other cell types, we propose that dual specificity is a general mechanism for activin ligands. In addition, we found that the antagonist follistatin inhibited signaling by all the tested activins, whereas the antagonist cerberus specifically inhibited activin B. Taken together, we propose that activins may be considered dual specificity TGF-ß family members, critically affecting how activins may be considered and targeted clinically.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Activins/chemistry , Cell Line, Tumor , Humans , Protein Multimerization , Protein Structure, Quaternary , Substrate Specificity
8.
Nat Commun ; 10(1): 4533, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586071

ABSTRACT

Multiple myeloma is an incurable, bone marrow-dwelling malignancy that disrupts bone homeostasis causing skeletal damage and pain. Mechanisms underlying myeloma-induced bone destruction are poorly understood and current therapies do not restore lost bone mass. Using transcriptomic profiling of isolated bone lining cell subtypes from a murine myeloma model, we find that bone morphogenetic protein (BMP) signalling is upregulated in stromal progenitor cells. BMP signalling has not previously been reported to be dysregulated in myeloma bone disease. Inhibition of BMP signalling in vivo using either a small molecule BMP receptor antagonist or a solubilized BMPR1a-FC receptor ligand trap prevents trabecular and cortical bone volume loss caused by myeloma, without increasing tumour burden. BMP inhibition directly reduces osteoclastogenesis, increases osteoblasts and bone formation, and suppresses bone marrow sclerostin levels. In summary we describe a novel role for the BMP pathway in myeloma-induced bone disease that can be therapeutically targeted.


Subject(s)
Bone Diseases/drug therapy , Bone Morphogenetic Proteins/metabolism , Multiple Myeloma/complications , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Stem Cells/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Density/drug effects , Bone Diseases/etiology , Bone Diseases/pathology , Bone Marrow/pathology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Cell Line, Tumor , Disease Models, Animal , Femur/cytology , Femur/drug effects , Femur/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred Strains , Multiple Myeloma/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , RNA-Seq , Signal Transduction/drug effects , Stem Cells/pathology , Tibia/cytology , Tibia/drug effects , Tibia/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Methods Mol Biol ; 1891: 37-49, 2019.
Article in English | MEDLINE | ID: mdl-30414125

ABSTRACT

Binding of a BMP to its cognate cell surface receptors is the initiating step in the BMP signaling cascade. Thus, knowing which BMP-receptor complexes form is vital for understanding the physiological activities of a particular BMP. Here, we describe a surface plasmon resonance (SPR)-based, high-throughput approach that allows fast identification and evaluation of BMP-receptor complexes. Briefly, the extracellular, BMP-binding domains of receptors are produced as human IgG1-Fc-fusion proteins. The Fc moiety enables simple capture of the Fc-receptor-fusion protein on the sensor chip, supports a highly reproducible, uniform approach of surface regeneration, and ensures full activity of the receptor moiety. BMPs are injected over the captured receptors at one concentration (approximately 60-100 nM), permitting stratification of high-affinity, medium-affinity, and low-affinity binders. Using this concentration range, equilibrium dissociation constants for high-affinity and medium-affinity binders can be estimated with good accuracy and with great precision from the single injection binding curves.


Subject(s)
Biosensing Techniques , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , High-Throughput Screening Assays , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Proteins/genetics , Protein Interaction Mapping , Surface Plasmon Resonance
10.
BMC Biol ; 15(1): 19, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28257634

ABSTRACT

BACKGROUND: Growth/differentiation factor 8 (GDF8) and GDF11 are two highly similar members of the transforming growth factor ß (TGFß) family. While GDF8 has been recognized as a negative regulator of muscle growth and differentiation, there are conflicting studies on the function of GDF11 and whether GDF11 has beneficial effects on age-related dysfunction. To address whether GDF8 and GDF11 are functionally identical, we compared their signaling and structural properties. RESULTS: Here we show that, despite their high similarity, GDF11 is a more potent activator of SMAD2/3 and signals more effectively through the type I activin-like receptor kinase receptors ALK4/5/7 than GDF8. Resolution of the GDF11:FS288 complex, apo-GDF8, and apo-GDF11 crystal structures reveals unique properties of both ligands, specifically in the type I receptor binding site. Lastly, substitution of GDF11 residues into GDF8 confers enhanced activity to GDF8. CONCLUSIONS: These studies identify distinctive structural features of GDF11 that enhance its potency, relative to GDF8; however, the biological consequences of these differences remain to be determined.


Subject(s)
Bone Morphogenetic Proteins/chemistry , Growth Differentiation Factors/chemistry , Myostatin/chemistry , Myostatin/metabolism , Amino Acid Sequence , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Cells, Cultured , Crystallography, X-Ray , Follistatin/metabolism , Genes, Reporter , Growth Differentiation Factors/antagonists & inhibitors , Growth Differentiation Factors/metabolism , Humans , Injections, Intravenous , Ligands , Luciferases/metabolism , Mice , Models, Molecular , Myoblasts/metabolism , Myocardium/metabolism , Myostatin/antagonists & inhibitors , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Sequence Alignment , Signal Transduction , Smad Proteins/metabolism , Structural Homology, Protein , Structure-Activity Relationship
11.
J Biol Chem ; 292(10): 4138-4151, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28126904

ABSTRACT

Transforming growth factor ß (TGF-ß) pathways are key determinants of cell fate in animals. Their basic mechanism of action is simple. However, to produce cell-specific responses, TGF-ß pathways are heavily regulated by secondary factors, such as membrane-associated EGF-CFC family proteins. Cellular activities of EGF-CFC proteins have been described, but their molecular functions, including how the mammalian homologs Cripto-1 and Cryptic recognize and regulate TGF-ß family ligands, are less clear. Here we use purified human Cripto-1 and mouse Cryptic produced in mammalian cells to show that these two EGF-CFC homologs have distinct, highly specific ligand binding activities. Cripto-1 interacts with BMP-4 in addition to its known partner Nodal, whereas Cryptic interacts only with Activin B. These interactions depend on the integrity of the protein, as truncated or deglycosylated Cripto-1 lacked BMP-4 binding activity. Significantly, Cripto-1 and Cryptic blocked binding of their cognate ligands to type I and type II TGF-ß receptors, indicating that Cripto-1 and Cryptic contact ligands at their receptor interaction surfaces and, thus, that they could inhibit their ligands. Indeed, soluble Cripto-1 and Cryptic inhibited ligand signaling in various cell-based assays, including SMAD-mediated luciferase reporter gene expression, and differentiation of a multipotent stem cell line. But in agreement with previous work, the membrane bound form of Cripto-1 potentiated signaling, revealing a critical role of membrane association for its established cellular activity. Thus, our studies provide new insights into the mechanism of ligand recognition by this enigmatic family of membrane-anchored TGF-ß family signaling regulators and link membrane association with their signal potentiating activities.


Subject(s)
Cell Membrane/metabolism , GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Cell Differentiation , Hep G2 Cells , Humans , Ligands , Protein Binding , Receptor, Transforming Growth Factor-beta Type II , Sequence Homology, Amino Acid , Signal Transduction
12.
Nat Commun ; 7: 13602, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27934856

ABSTRACT

In vitro differentiation of human pluripotent stem cells (hPSCs) recapitulates early aspects of human embryogenesis, but the underlying processes are poorly understood and controlled. Here we show that modulating the bulk cell density (BCD: cell number per culture volume) deterministically alters anteroposterior patterning of primitive streak (PS)-like priming. The BCD in conjunction with the chemical WNT pathway activator CHIR99021 results in distinct paracrine microenvironments codifying hPSCs towards definitive endoderm, precardiac or presomitic mesoderm within the first 24 h of differentiation, respectively. Global gene expression and secretome analysis reveals that TGFß superfamily members, antagonist of Nodal signalling LEFTY1 and CER1, are paracrine determinants restricting PS progression. These data result in a tangible model disclosing how hPSC-released factors deflect CHIR99021-induced lineage commitment over time. By demonstrating a decisive, functional role of the BCD, we show its utility as a method to control lineage-specific differentiation. Furthermore, these findings have profound consequences for inter-experimental comparability, reproducibility, bioprocess optimization and scale-up.


Subject(s)
Cell Count , Pluripotent Stem Cells/physiology , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , Oligonucleotide Array Sequence Analysis , Protein Array Analysis , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering , Signal Transduction/physiology , Transcriptome , Transforming Growth Factor beta/genetics , Wnt Proteins/genetics
13.
Anal Biochem ; 508: 97-103, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27365221

ABSTRACT

Half-maximal inhibitory concentration (IC50) is the most widely used and informative measure of a drug's efficacy. It indicates how much drug is needed to inhibit a biological process by half, thus providing a measure of potency of an antagonist drug in pharmacological research. Most approaches to determine IC50 of a pharmacological compound are based on assays that utilize whole cell systems. While they generally provide outstanding potency information, results can depend on the experimental cell line used and may not differentiate a compound's ability to inhibit specific interactions. Here we show using the secreted Transforming Growth Factor-ß (TGF-ß) family ligand BMP-4 and its receptors as example that surface plasmon resonance can be used to accurately determine IC50 values of individual ligand-receptor pairings. The molecular resolution achievable wih this approach can help distinguish inhibitors that specifically target individual complexes, or that can inhibit multiple functional interactions at the same time.


Subject(s)
Chemistry Techniques, Analytical/methods , Receptors, Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta1/chemistry , Biological Assay , Bone Morphogenetic Protein 4/chemistry , Bone Morphogenetic Protein 4/metabolism , Humans , Inhibitory Concentration 50 , Protein Binding , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transforming Growth Factor beta1/metabolism
14.
J Biol Chem ; 291(20): 10792-804, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26961869

ABSTRACT

Transforming growth factor-ß (TGF-ß) family ligands are pleiotropic cytokines. Their physiological activities are not determined by a simple coupling of stimulus and response, but depend critically on context, i.e. the interplay of receptors, ligands, and regulators that form the TGF-ß signal transduction system of a cell or tissue. How these different components combine to regulate signaling activities remains poorly understood. Here, we describe a ligand-mediated mechanism of signaling regulation. Based on the observation that the type II TGF-ß family receptors ActRIIA, ActRIIB, and BMPRII interact with a large group of overlapping ligands at overlapping epitopes, we hypothesized high affinity ligands compete with low affinity ligands for receptor binding and signaling. We show activin A and other high affinity ligands directly inhibited signaling by the low affinity ligands BMP-2, BMP-7, and BMP-9. We demonstrate activin A functions as a competitive inhibitor that blocks the ligand binding epitope on type II receptors. We propose binding competition and signaling antagonism are integral functions of the TGF-ß signal transduction system. These functions could help explain how activin A modulates physiological signaling during extraordinary cellular responses, such as injury and wound healing, and how activin A could elicit disease phenotypes such as cancer-related muscle wasting and fibrosis.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/metabolism , Bone Morphogenetic Protein Receptors, Type II/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type II/metabolism , Transforming Growth Factor beta/metabolism , Activins/antagonists & inhibitors , Activins/metabolism , Activins/pharmacology , Binding, Competitive , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Line, Tumor , Follistatin/pharmacology , Growth Differentiation Factor 2 , Growth Differentiation Factors/metabolism , Hep G2 Cells , Humans , Ligands , Recombinant Proteins/metabolism , Signal Transduction/drug effects
15.
J Mol Biol ; 428(3): 590-602, 2016 Feb 13.
Article in English | MEDLINE | ID: mdl-26802359

ABSTRACT

Cerberus is a key regulator of vertebrate embryogenesis. Its biological function has been studied extensively in frog and mouse embryos. Its ability to bind and antagonize the transforming growth factor-ß (TGF-ß) family ligand Nodal is well established. Strikingly, the molecular function of Cerberus remains poorly understood. The underlying reason is that Cerberus is a complex, multifunctional protein: It binds and inhibits multiple TGF-ß family ligands, it may bind and inhibit some Wnt family members, and two different forms with distinct activities have been described. In addition, sequence homology between frog and mammalian Cerberus is low, suggesting that previous studies, which analyzed frog Cerberus function, may not accurately describe the function of mammalian Cerberus. We therefore undertook to determine the molecular activities of human Cerberus in TGF-ß family signaling. Using purified proteins, surface plasmon resonance, and reporter gene assays, we discovered that human Cerberus bound and inhibited the TGF-ß family ligands Activin B, BMP-6, and BMP-7, but not the frog Cerberus ligand BMP-2. Notably, full-length Cerberus successfully blocked ligand binding to type II receptors, but the short form was less effective. In addition, full-length Cerberus suppressed breast cancer cell migration but the short form did not. Thus, our findings expand the roles of Cerberus as TGF-ß family signaling inhibitor, provide a molecular rationale for the function of the N-terminal region, and support the idea that Cerberus could have regulatory activities beyond direct inhibition of TGF-ß family signaling.


Subject(s)
Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Cytokines/metabolism , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cytokines/chemistry , Female , Humans , Molecular Sequence Data , Proteolysis , Sequence Alignment , Signal Transduction
16.
PLoS One ; 10(1): e0114954, 2015.
Article in English | MEDLINE | ID: mdl-25603319

ABSTRACT

The Transforming Growth Factor-ß (TGFß) family ligand Nodal is an essential embryonic morphogen that is associated with progression of breast and other cancers. It has therefore been suggested that Nodal inhibitors could be used to treat breast cancers where Nodal plays a defined role. As secreted antagonists, such as Cerberus, tightly regulate Nodal signaling during embryonic development, we undertook to produce human Cerberus, characterize its biochemical activities, and determine its effect on human breast cancer cells. Using quantitative methods, we investigated the mechanism of Nodal signaling, we evaluated binding of human Cerberus to Nodal and other TGFß family ligands, and we characterized the mechanism of Nodal inhibition by Cerberus. Using cancer cell assays, we examined the ability of Cerberus to suppress aggressive breast cancer cell phenotypes. We found that human Cerberus binds Nodal with high affinity and specificity, blocks binding of Nodal to its signaling partners, and inhibits Nodal signaling. Moreover, we showed that Cerberus profoundly suppresses migration, invasion, and colony forming ability of Nodal expressing and Nodal supplemented breast cancer cells. Taken together, our studies provide mechanistic insights into Nodal signaling and Nodal inhibition with Cerberus and highlight the potential value of Cerberus as anti-Nodal therapeutic.


Subject(s)
Cytokines/metabolism , Nodal Protein/metabolism , Phenotype , Signal Transduction , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Proliferation , Female , GPI-Linked Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Kinetics , Ligands , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Protein Binding
17.
Biochem J ; 465(2): 325-35, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25330773

ABSTRACT

Many human pathogens have strict host specificity, which affects not only their epidemiology but also the development of animal models and vaccines. Complement Factor H (FH) is recruited to pneumococcal cell surface in a human-specific manner via the N-terminal domain of the pneumococcal protein virulence factor choline-binding protein A (CbpAN). FH recruitment enables Streptococcus pneumoniae to evade surveillance by human complement system and contributes to pneumococcal host specificity. The molecular determinants of host specificity of complement evasion are unknown. In the present study, we show that a single human FH (hFH) domain is sufficient for tight binding of CbpAN, present the crystal structure of the complex and identify the critical structural determinants for host-specific FH recruitment. The results offer new approaches to the development of better animal models for pneumococcal infection and redesign of the virulence factor for pneumococcal vaccine development and reveal how FH recruitment can serve as a mechanism for both pneumococcal complement evasion and adherence.


Subject(s)
Bacterial Proteins/chemistry , Complement Factor H/chemistry , Multiprotein Complexes/chemistry , Streptococcus pneumoniae/chemistry , Virulence Factors/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
18.
J Biol Chem ; 287(33): 27313-25, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22718755

ABSTRACT

Activin receptor-like kinase 1 (ALK1), an endothelial cell-specific type I receptor of the TGF-ß superfamily, is an important regulator of normal blood vessel development as well as pathological tumor angiogenesis. As such, ALK1 is an important therapeutic target. Thus, several ALK1-directed agents are currently in clinical trials as anti-angiogenic cancer therapeutics. Given the biological and clinical importance of the ALK1 signaling pathway, we sought to elucidate the biophysical and structural basis underlying ALK1 signaling. The TGF-ß family ligands BMP9 and BMP10 as well as the three type II TGF-ß family receptors ActRIIA, ActRIIB, and BMPRII have been implicated in ALK1 signaling. Here, we provide a kinetic and thermodynamic analysis of BMP9 and BMP10 interactions with ALK1 and type II receptors. Our data show that BMP9 displays a significant discrimination in type II receptor binding, whereas BMP10 does not. We also report the crystal structure of a fully assembled ternary complex of BMP9 with the extracellular domains of ALK1 and ActRIIB. The structure reveals that the high specificity of ALK1 for BMP9/10 is determined by a novel orientation of ALK1 with respect to BMP9, which leads to a unique set of receptor-ligand interactions. In addition, the structure explains how BMP9 discriminates between low and high affinity type II receptors. Taken together, our findings provide structural and mechanistic insights into ALK1 signaling that could serve as a basis for novel anti-angiogenic therapies.


Subject(s)
Activin Receptors, Type II/chemistry , Bone Morphogenetic Proteins/chemistry , Growth Differentiation Factors/chemistry , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Crystallography, X-Ray , Growth Differentiation Factor 2 , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , HEK293 Cells , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Signal Transduction/drug effects , Signal Transduction/genetics
19.
Nature ; 471(7338): 336-40, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21317882

ABSTRACT

The TrkH/TrkG/KtrB proteins mediate K(+) uptake in bacteria and probably evolved from simple K(+) channels by multiple gene duplications or fusions. Here we present the crystal structure of a TrkH from Vibrio parahaemolyticus. TrkH is a homodimer, and each protomer contains an ion permeation pathway. A selectivity filter, similar in architecture to those of K(+) channels but significantly shorter, is lined by backbone and side-chain oxygen atoms. Functional studies showed that TrkH is selective for permeation of K(+) and Rb(+) over smaller ions such as Na(+) or Li(+). Immediately intracellular to the selectivity filter are an intramembrane loop and an arginine residue, both highly conserved, which constrict the permeation pathway. Substituting the arginine with an alanine significantly increases the rate of K(+) flux. These results reveal the molecular basis of K(+) selectivity and suggest a novel gating mechanism for this large and important family of membrane transport proteins.


Subject(s)
Potassium Channels/chemistry , Potassium Channels/metabolism , Vibrio parahaemolyticus/chemistry , ATP-Binding Cassette Transporters/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Ion Channel Gating , Ion Transport , Models, Molecular , Molecular Sequence Data , Potassium/metabolism , Structure-Activity Relationship , Substrate Specificity
20.
J Mol Biol ; 407(3): 450-64, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21262232

ABSTRACT

In the cell, protein folding is mediated by folding catalysts and chaperones. The two functions are often linked, especially when the catalytic module forms part of a multidomain protein, as in Methanococcus jannaschii peptidyl-prolyl cis/trans isomerase FKBP26. Here, we show that FKBP26 chaperone activity requires both a 50-residue insertion in the catalytic FKBP domain, also called 'Insert-in-Flap' or IF domain, and an 80-residue C-terminal domain. We determined FKBP26 structures from four crystal forms and analyzed chaperone domains in light of their ability to mediate protein-protein interactions. FKBP26 is a crescent-shaped homodimer. We reason that folding proteins are bound inside the large crescent cleft, thus enabling their access to inward-facing peptidyl-prolyl cis/trans isomerase catalytic sites and ipsilateral chaperone domain surfaces. As these chaperone surfaces participate extensively in crystal lattice contacts, we speculate that the observed lattice contacts reflect a proclivity for protein associations and represent substrate interactions by FKBP26 chaperone domains. Finally, we find that FKBP26 is an exceptionally flexible molecule, suggesting a mechanism for nonspecific substrate recognition.


Subject(s)
Archaeal Proteins/chemistry , Methanococcus/enzymology , Molecular Chaperones/chemistry , Peptidylprolyl Isomerase/chemistry , Archaeal Proteins/metabolism , Binding Sites , Catalysis , Models, Molecular , Molecular Chaperones/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Folding , Protein Structure, Tertiary , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...