Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Bone Miner Res ; 39(4): 382-398, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38477818

ABSTRACT

Single case studies of extraordinary disease resilience may provide therapeutic insight into conditions for which no definitive treatments exist. An otherwise healthy 35-year-old man (patient-R) with the canonical pathogenic ACVR1R206H variant and the classic congenital great toe malformation of fibrodysplasia ossificans progressiva (FOP) had extreme paucity of post-natal heterotopic ossification (HO) and nearly normal mobility. We hypothesized that patient-R lacked a sufficient post-natal inflammatory trigger for HO. A plasma biomarker survey revealed a reduction in total matrix metalloproteinase-9 (MMP-9) compared to healthy controls and individuals with quiescent FOP. Whole exome sequencing identified compound heterozygous variants in MMP-9 (c.59C > T, p.A20V and c.493G > A, p.D165N). Structural analysis of the D165N variant predicted both decreased MMP-9 secretion and activity that were confirmed by enzyme-linked immunosorbent assay and gelatin zymography. Further, human proinflammatory M1-like macrophages expressing either MMP-9 variant produced significantly less Activin A, an obligate ligand for HO in FOP, compared to wildtype controls. Importantly, MMP-9 inhibition by genetic, biologic, or pharmacologic means in multiple FOP mouse models abrogated trauma-induced HO, sequestered Activin A in the extracellular matrix (ECM), and induced regeneration of injured skeletal muscle. Our data suggest that MMP-9 is a druggable node linking inflammation to HO, orchestrates an existential role in the pathogenesis of FOP, and illustrates that a single patient's clinical phenotype can reveal critical molecular mechanisms of disease that unveil novel treatment strategies.


A healthy 35-year-old man (patient-R) with the classic fibrodysplasia ossificans progressiva (FOP) mutation and the congenital great toe malformation of FOP had extreme lack of heterotopic ossification (HO) and nearly normal mobility. We hypothesized that patient-R lacked a sufficient inflammatory trigger for HO. Blood tests revealed a reduction in the level of an inflammatory protein called matrix metalloproteinase-9 (MMP-9) compared to other individuals with FOP as well as healthy controls. DNA analysis in patient-R identified mutations in MMP-9, one of which predicted decreased activity of MMP-9 which was confirmed by further testing. Inflammatory cells (macrophages) expressing the MMP-9 mutations identified in patient-R produced significantly less Activin A, an obligate stimulus for HO in FOP. In order to determine if MMP-9 deficiency was a cause of HO prevention in FOP, we inhibited MMP-9 activity by genetic, biologic, or pharmacologic means in FOP mouse models and showed that MMP-9 inhibition prevented or dramatically decreased trauma-induced HO in FOP, locked-up Activin A in the extracellular matrix, and induced regeneration of injured skeletal muscle. Our data show that MMP-9 links inflammation to HO and illustrate that one patient's clinical picture can reveal critical molecular mechanisms of disease that unveil new treatment strategies.


Subject(s)
Activin Receptors, Type I , Matrix Metalloproteinase 9 , Myositis Ossificans , Adult , Animals , Humans , Male , Mice , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/deficiency , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Myositis Ossificans/genetics , Myositis Ossificans/pathology , Myositis Ossificans/metabolism , Ossification, Heterotopic/pathology , Ossification, Heterotopic/genetics , Ossification, Heterotopic/metabolism
2.
Biomolecules ; 13(7)2023 07 14.
Article in English | MEDLINE | ID: mdl-37509165

ABSTRACT

Although structurally similar to type II counterparts, type I or activin receptor-like kinases (ALKs) are set apart by a metastable helix-loop-helix (HLH) element preceding the protein kinase domain that, according to a longstanding paradigm, serves passive albeit critical roles as an inhibitor-to-substrate-binding switch. A single recurrent mutation in the codon of the penultimate residue, directly adjacent the position of a constitutively activating substitution, causes milder activation of ACVR1/ALK2 leading to sporadic heterotopic bone deposition in patients presenting with fibrodysplasia ossificans progressiva, or FOP. To determine the protein structural-functional basis for the gain of function, R206H mutant, Q207D (aspartate-substituted caALK2) and HLH subdomain-truncated (208 Ntrunc) forms were compared to one another and the wild-type enzyme through in vitro kinase and protein-protein interaction analyses that were complemented by signaling read-out (p-Smad) in primary mouse embryonic fibroblasts and Drosophila S2 cells. Contrary to the paradigm, the HLH subdomain actively suppressed the phosphotransferase activity of the enzyme, even in the absence of FKBP12. Unexpectedly, perturbation of the HLH subdomain elevated kinase activity at a distance, i.e., allosterically, at the ATP-binding and polypeptide-interacting active site cleft. Accessibility to polypeptide substrate (BMP Smad C-terminal tails) due to allosterically altered conformations of type I active sites within heterohexameric cytoplasmic signaling complexes-assembled noncanonically by activin-type II receptors extracellularly-is hypothesized to produce a gain of function of the R206H mutant protein responsible for episodic heterotopic ossification in FOP.


Subject(s)
Activin Receptors, Type I , Gain of Function Mutation , Animals , Mice , Activin Receptors/genetics , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Fibroblasts/metabolism , Mutation , Peptides/genetics
3.
J Bone Miner Res ; 37(11): 2058-2076, 2022 11.
Article in English | MEDLINE | ID: mdl-36153796

ABSTRACT

Bone morphogenetic protein (BMP) signaling is critical in skeletal development. Overactivation can trigger heterotopic ossification (HO) as in fibrodysplasia ossificans progressiva (FOP), a rare, progressive disease of massive HO formation. A small subset of FOP patients harboring the causative ACVR1R206H mutation show strikingly mild or delayed-onset HO, suggesting that genetic variants in the BMP pathway could act as disease modifiers. Whole-exome sequencing of one such patient identified BMPR1AR443C and ACVR2AV173I as candidate modifiers. Molecular modeling predicted significant structural perturbations. Neither variant decreased BMP signaling in ACVR1R206H HEK 293T cells at baseline or after stimulation with BMP4 or activin A (AA), ligands that activate ACVR1R206H signaling. Overexpression of BMPR1AR443C in a Tg(ACVR1-R206Ha) embryonic zebrafish model, in which overactive BMP signaling yields ventralized embryos, did not alter ventralization severity, while ACVR2AV173I exacerbated ventralization. Co-expression of both variants did not affect dorsoventral patterning. In contrast, BMPR1A knockdown in ACVR1R206H HEK cells decreased ligand-stimulated BMP signaling but did not affect dorsoventral patterning in Tg(ACVR1-R206Ha) zebrafish. ACVR2A knockdown decreased only AA-stimulated signaling in ACVR1R206H HEK cells and had no effect in Tg(ACVR1-R206Ha) zebrafish. Co-knockdown in ACVR1R206H HEK cells decreased basal and ligand-stimulated signaling, and co-knockdown/knockout (bmpr1aa/ab; acvr2aa/ab) decreased Tg(ACVR1-R206Ha) zebrafish ventralization phenotypes. Our functional studies showed that knockdown of wild-type BMPR1A and ACVR2A could attenuate ACVR1R206H signaling, particularly in response to AA, and that ACVR2AV173I unexpectedly increased ACVR1R206H -mediated signaling in zebrafish. These studies describe a useful strategy and platform for functionally interrogating potential genes and genetic variants that may impact the BMP signaling pathway. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Myositis Ossificans , Ossification, Heterotopic , Animals , Humans , Myositis Ossificans/genetics , Myositis Ossificans/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Exome Sequencing , Ligands , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Ossification, Heterotopic/metabolism , Mutation
4.
Am J Med Genet A ; 188(3): 806-817, 2022 03.
Article in English | MEDLINE | ID: mdl-34854557

ABSTRACT

Genetic variants are vital in informing clinical phenotypes, aiding physical diagnosis, guiding genetic counseling, understanding the molecular basis of disease, and potentially stimulating drug development. Here we describe two families with an ultrarare ACVR1 gain-of-function pathogenic variant (codon 375, Arginine > Proline; ACVR1R375P ) responsible for a mild nonclassic fibrodysplasia ossificans progressiva (FOP) phenotype. Both families include people with the ultrarare ACVR1R375P variant who exhibit features of FOP while other individuals currently do not express any clinical signs of FOP. Thus, the mild ACVR1R375P variant greatly expands the scope and understanding of this rare disorder.


Subject(s)
Myositis Ossificans , Activin Receptors, Type I/genetics , Humans , Mutation , Myositis Ossificans/diagnosis , Myositis Ossificans/genetics , Myositis Ossificans/pathology , Phenotype
5.
Expert Opin Drug Discov ; 14(12): 1237-1253, 2019 12.
Article in English | MEDLINE | ID: mdl-31513432

ABSTRACT

Introduction: Protein kinases are a major target for small-molecule drug development. However, relatively few compounds are free of off-target toxicity and reach the clinic. Because the 500-plus kinases share conserved ATP-binding clefts, the site targeted by competitive inhibitors, generation of specific therapeutics remains a nearly intractable challenge. Areas covered: Inducing degradation, instead of inhibition by occupancy-driven drugs, is an emerging strategy that offers the long-sought specificity, as well as mechanistic benefits. Currently approved inhibitors require steady-state binding and leave proteins intact for interactions in multi-protein complexes. After a general background about induced protein degradation, perspectives on protein kinases are provided. Expert opinion: Induced degradation by state-of-the-art compounds (proteolysis-targeting chimeras, PROTACs) has been shown for protein kinases, albeit in early pre-clinical stages. Further work is required to expand the number of enzymes that could be exploited to direct proteins for degradation by ubiquitylation. In addition, despite the simple modularity of the chimeras, generation of hits will require empirical approaches due to the role of protein-protein interactions and distribution of tagging sites. However, given the advantages of degradation, drug discovery efforts targeting protein kinases should increasingly shift toward generation and screening of inducers of degradation and away from occupancy-based inhibitors of old.


Subject(s)
Drug Development/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinases/drug effects , Animals , Drug Design , Drug Discovery/methods , Humans , Protein Kinase Inhibitors/adverse effects , Protein Kinases/metabolism , Proteins/metabolism , Proteolysis/drug effects , Small Molecule Libraries
6.
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
7.
J Bone Miner Res ; 31(9): 1652-65, 2016 09.
Article in English | MEDLINE | ID: mdl-27027798

ABSTRACT

Hypoxia and inflammation are implicated in the episodic induction of heterotopic endochondral ossification (HEO); however, the molecular mechanisms are unknown. HIF-1α integrates the cellular response to both hypoxia and inflammation and is a prime candidate for regulating HEO. We investigated the role of hypoxia and HIF-1α in fibrodysplasia ossificans progressiva (FOP), the most catastrophic form of HEO in humans. We found that HIF-1α increases the intensity and duration of canonical bone morphogenetic protein (BMP) signaling through Rabaptin 5 (RABEP1)-mediated retention of Activin A receptor, type I (ACVR1), a BMP receptor, in the endosomal compartment of hypoxic connective tissue progenitor cells from patients with FOP. We further show that early inflammatory FOP lesions in humans and in a mouse model are markedly hypoxic, and inhibition of HIF-1α by genetic or pharmacologic means restores canonical BMP signaling to normoxic levels in human FOP cells and profoundly reduces HEO in a constitutively active Acvr1(Q207D/+) mouse model of FOP. Thus, an inflammation and cellular oxygen-sensing mechanism that modulates intracellular retention of a mutant BMP receptor determines, in part, its pathologic activity in FOP. Our study provides critical insight into a previously unrecognized role of HIF-1α in the hypoxic amplification of BMP signaling and in the episodic induction of HEO in FOP and further identifies HIF-1α as a therapeutic target for FOP and perhaps nongenetic forms of HEO. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Signal Transduction , Activin Receptors, Type I/metabolism , Animals , Cell Hypoxia , Chondrogenesis , Disease Models, Animal , Endosomes/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/pathology , Ligands , Male , Mice, Transgenic , Models, Biological , Myositis Ossificans/pathology , Stem Cells/metabolism , Stem Cells/pathology , Tooth Exfoliation/pathology , Tooth, Deciduous/pathology
8.
Am J Med Genet A ; 167A(10): 2265-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26097044

ABSTRACT

Severe variants of fibrodysplasia ossificans progressiva (FOP) affect <2% of all FOP patients worldwide, but provide an unprecedented opportunity to probe the phenotype-genotype relationships that propel the pathology of this disabling disease. We evaluated two unrelated children who had severe reduction deficits of the hands and feet with absence of nails, progressive heterotopic ossification, hypoplasia of the brain stem, motor and cognitive developmental delays, facial dysmorphology, small malformed teeth, and abnormal hair development. One child had sensorineural hearing loss, microcytic anemia, and a tethered spinal cord and the other had a patent ductus arteriosus and gonadal dysgenesis with sex reversal (karyotype 46, XY female). Both children had an identical mutation in ACVR1 c.772A>G; p.Arg258Gly (R258G), not previously described in FOP. Although many, if not most, FOP mutations directly perturb the structure of the GS regulatory subdomain and presumably the adjacent αC helix, substitution with glycine at R258 may directly alter the position of the helix in the kinase domain, eliminating a key aspect of the autoinhibitory mechanism intrinsic to the wild-type ACVR1 kinase. The high fidelity phenotype-genotype relationship in these unrelated children with the most severe FOP phenotype reported to date suggests that the shared features are due to the dysregulated activity of the mutant kinase during development and postnatally, and provides vital insight into the structural biology and function of ACVR1 as well as the design of small molecule inhibitors.


Subject(s)
Abnormalities, Multiple/pathology , Activin Receptors, Type I/genetics , Mutation , Myositis Ossificans/pathology , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/genetics , Activin Receptors, Type I/metabolism , Amino Acid Substitution , Female , Gene Expression , Genetic Association Studies , Genotype , Humans , Infant , Karyotype , Models, Molecular , Myositis Ossificans/diagnosis , Myositis Ossificans/enzymology , Myositis Ossificans/genetics , Phenotype , Protein Structure, Tertiary , Severity of Illness Index
10.
PLoS One ; 7(4): e35062, 2012.
Article in English | MEDLINE | ID: mdl-22529972

ABSTRACT

We identified an amino acid change (p.G92E) in the Bone Morphogenetic Protein antagonist NOGGIN in a 22-month-old boy who presented with a unilateral brachydactyly type B phenotype. Brachydactyly type B is a skeletal malformation that has been associated with increased Bone Morphogenetic Protein pathway activation in other patients. Previously, the amino acid change p.G92E in NOGGIN was described as causing fibrodysplasia ossificans progressiva, a rare genetic disorder characterized by limb malformations and progressive heterotopic bone formation in soft tissues that, like Brachydactyly type B, is caused by increased activation of Bone Morphogenetic Protein signaling. To determine whether G92E-NOGGIN shows impaired antagonism that could lead to increased Bone Morphogenetic Protein signaling, we performed functional assays to evaluate inhibition of BMP signaling. Interestingly, wt-NOGGIN shows different inhibition efficacies towards various Bone Morphogenetic Proteins that are known to be essential in limb development. However, comparing the biological activity of G92E-NOGGIN with wt-NOGGIN, we observed that G92E-NOGGIN inhibits activation of bone morphogenetic protein signaling with equal efficiency as wt-NOGGIN, supporting that G92E-NOGGIN does not cause pathological effects. Genetic testing of the child's parents revealed the same amino acid change in the healthy father, further supporting that p.G92E is a neutral amino acid substitution in NOGGIN. We conclude that p.G92E represents a rare polymorphism of the NOGGIN gene-- causing neither brachydactyly nor fibrodysplasia ossificans progressiva. This study highlights that a given genetic variation should not be considered pathogenic unless supported by functional analyses.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Mutation , Amino Acid Substitution , Animals , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/metabolism , Brachydactyly/diagnostic imaging , Brachydactyly/etiology , Carrier Proteins/chemistry , Cell Line , Chick Embryo , Humans , Infant , Male , Mice , Models, Molecular , Phenotype , Polymorphism, Genetic , Protein Binding , Protein Conformation , Radiography
11.
Biochem Genet ; 50(1-2): 12-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21882044

ABSTRACT

Noggin (NOG) is an important regulator for the signaling of bone morphogenetic proteins. In this study, we sequenced the complete coding sequence of the canine NOG gene and characterized the nucleotide polymorphisms. The sequence length varied from 717 to 729 bp, depending on the number of a 6-bp tandem repeat unit (GGCGCG), an insertion that has not been observed in other mammalian NOG genes investigated to date. It results in extensions of (Gly-Ala)3-5 in the putative NOG protein. To survey the distribution of these tandem repeat polymorphisms, we analyzed 126 individuals in seven dog breeds. We identified only three alleles: (GGCGCG)3, (GGCGCG)4, and (GGCGCG)5. Although the allele frequencies were remarkably different among the breeds, the three alleles were present in all seven of the breeds and did not show any deviation from Hardy-Weinberg equilibrium.


Subject(s)
Carrier Proteins/genetics , Dogs/genetics , Polymorphism, Genetic , Amino Acid Sequence , Animals , Gene Frequency , Minisatellite Repeats , Molecular Sequence Data , Tandem Repeat Sequences
12.
J Mol Biol ; 412(4): 601-18, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21821041

ABSTRACT

Transforming growth factor ß isoforms (TGF-ß) are among the most recently evolved members of a signaling superfamily with more than 30 members. TGF-ß play vital roles in regulating cellular growth and differentiation, and they signal through a highly restricted subset of receptors known as TGF-ß type I receptor (TßR-I) and TGF-ß type II receptor (TßR-II). TGF-ß's specificity for TßR-I has been proposed to arise from its pre-helix extension, a five-residue loop that binds in the cleft between TGF-ß and TßR-II. The structure and backbone dynamics of the unbound form of the TßR-I extracellular domain were determined using NMR to investigate the extension's role in binding. This showed that the unbound form is highly similar to the bound form in terms of both the ß-strand framework that defines the three-finger toxin fold and the extension and its characteristic cis-Ile54-Pro55 peptide bond. The NMR data further showed that the extension and two flanking 3(10) helices are rigid on the nanosecond-to-picosecond timescale. The functional significance of several residues within the extension was investigated by binding studies and reporter gene assays in cultured epithelial cells. These demonstrated that the pre-helix extension is essential for binding, with Pro55 and Pro59 each playing a major role. These findings suggest that the pre-helix extension and its flanking prolines evolved to endow the TGF-ß signaling complex with its unique specificity, departing from the ancestral promiscuity of the bone morphogenetic protein subfamily, where the binding interface of the type I receptor is highly flexible.


Subject(s)
Proline/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Humans , Isoleucine/chemistry , Isoleucine/physiology , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Proline/chemistry , Proline/physiology , Protein Binding/physiology , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Secondary/physiology , Receptor, Transforming Growth Factor-beta Type I , Sequence Homology, Amino Acid , Transforming Growth Factor beta1/metabolism
13.
Cells Tissues Organs ; 194(2-4): 291-5, 2011.
Article in English | MEDLINE | ID: mdl-21525719

ABSTRACT

A single recurrent mutation in the regulatory subdomain of a bone morphogenetic protein type I receptor kinase has been linked to heterotopic ossification in classic fibrodysplasia ossificans progressiva (FOP). As a result of a substitution at 1 residue by only 1 other side chain (Arg206His) in just 1 of the 4 type I BMP receptors (ALK2/ACVR1), soft connective tissues progressively metamorphose through an endochondral process into cartilage that is replaced by bone. The substitution of arginine for histidine, also a basic residue yet with the singular property of ionization/protonation over the physiological pH range, led to the hypothesis of an aberrant, pH-sensitive switch mechanism for the ligand-independent activation of BMP signaling through the mutant receptor kinase in patients presenting with classic FOP. To test a potential aspect of the putative pH-dependent mechanism, i.e. loss of autoinhibition of the kinase mediated by the inhibitory protein FKBP12, in vitrointeraction analyses with purified wild-type and R206H ALK2 kinase and FKBP12 proteins were performed. Interactions between the kinases and inhibitory proteins were analyzed qualitatively and quantitatively by native gel electrophoresis and HPLC size exclusion chromatography and with an optical biosensor (Octet; ForteBio). Binding of inhibitory protein by the R206H mutant was diminished 3-fold relative to the wild type kinase at a physiological pH, yet below this value (<~7.5) pronounced nonspecific interactions, particularly with the mutant, prevented comparative evaluations. In conclusion, substitution with histidine leads to partial loss of inhibition of the mutant type I receptor through diminished binding of FKBP12, which may act as a gradient reader in morphogenetic contexts.


Subject(s)
Activin Receptors, Type I/metabolism , Amino Acid Substitution/genetics , Myositis Ossificans/complications , Myositis Ossificans/enzymology , Ossification, Heterotopic/complications , Ossification, Heterotopic/enzymology , Tacrolimus Binding Protein 1A/metabolism , Biosensing Techniques , Chromatography, Gel , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Interferometry , Protein Binding
14.
PLoS Genet ; 5(11): e1000747, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19956691

ABSTRACT

Signaling output of bone morphogenetic proteins (BMPs) is determined by two sets of opposing interactions, one with heterotetrameric complexes of cell surface receptors, the other with secreted antagonists that act as ligand traps. We identified two mutations (N445K,T) in patients with multiple synostosis syndrome (SYM1) in the BMP-related ligand GDF5. Functional studies of both mutants in chicken micromass culture demonstrated a gain of function caused by a resistance to the BMP-inhibitor NOGGIN and an altered signaling effect. Residue N445, situated within overlapping receptor and antagonist interfaces, is highly conserved among the BMP family with the exception of BMP9 and BMP10, in which it is substituted with lysine. Like the mutant GDF5, both BMPs are insensitive to NOGGIN and show a high chondrogenic activity. Ectopic expression of BMP9 or the GDF5 mutants resulted in massive induction of cartilage in an in vivo chick model presumably by bypassing the feedback inhibition imposed by endogenous NOGGIN. Swapping residues at the mutation site alone was not sufficient to render Bmp9 NOG-sensitive; however, successive introduction of two additional substitutions imparted high to total sensitivity on customized variants of Bmp9. In conclusion, we show a new mechanism for abnormal joint development that interferes with a naturally occurring regulatory mechanism of BMP signaling.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Carrier Proteins/physiology , Growth Differentiation Factor 5/genetics , Mutation , Animals , Cartilage , Cells, Cultured , Chickens , Feedback, Physiological , Humans , Mice , Synostosis/genetics
15.
Cytokine Growth Factor Rev ; 20(5-6): 367-77, 2009.
Article in English | MEDLINE | ID: mdl-19926516

ABSTRACT

The TGF-beta superfamily exhibits a feature making it distinct from many other growth factor families in that the inadequate number of ligands and receptors premises a high degree of promiscuity in ligand-receptor interaction. This highlights the importance of even small differences in affinities and specificities between different binding partners to maintain the broad spectrum of their well defined biological functions. Despite the promiscuous interactions recent data reveal differences in receptor recruitment, architectures of these assemblies and specific modulation by a multitude of extracellular as well as membrane-associated factors. These modulatory mechanisms might possibly add specificity towards defined biological functions despite the overlapping usage of receptors by various ligands.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Protein Receptors/physiology , Protein Multimerization/physiology , Animals , Bone Morphogenetic Protein Receptors/chemistry , Humans , Models, Biological , Models, Molecular , Structure-Activity Relationship , TGF-beta Superfamily Proteins/chemistry , TGF-beta Superfamily Proteins/metabolism , TGF-beta Superfamily Proteins/physiology
16.
Hum Mol Genet ; 18(15): 2863-74, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19429910

ABSTRACT

Mutations in the paired-domain transcription factor PAX9 are associated with non-syndromic tooth agenesis that preferentially affects posterior dentition. Of the 18 mutations identified to date, eight are phenotypically well-characterized missense mutations within the DNA-binding paired domain. We determined the structural and functional consequences of these paired domain missense mutations and correlated our findings with the associated dental phenotype variations. In vitro testing included subcellular localization, protein-protein interactions between MSX1 and mutant PAX9 proteins, binding of PAX9 mutants to a DNA consensus site and transcriptional activation from the Pax9 effector promoters Bmp4 and Msx1 with and without MSX1 as co-activator. All mutant PAX9 proteins were localized in the nucleus of transfected cells and physically interacted with MSX1 protein. Three of the mutants retained the ability to bind the consensus paired domain recognition sequence; the others were unable or only partly able to interact with this DNA fragment and also showed a similarly impaired capability for activation of transcription from the Msx1 and Bmp4 promoters. For seven of the eight mutants, the degree of loss of DNA-binding and promoter activation correlated quite well with the severity of the tooth agenesis pattern seen in vivo. One of the mutants however showed neither reduction in DNA-binding nor decrease in transactivation; instead, a loss of responsiveness to synergism with MSX1 in target promoter activation and a dominant negative effect when expressed together with wild-type PAX9 could be observed. Our structure-based studies, which modeled DNA binding and subdomain stability, were able to predict functional consequences quite reliably.


Subject(s)
Mutation , PAX9 Transcription Factor/chemistry , PAX9 Transcription Factor/genetics , Tooth/growth & development , Tooth/pathology , Amino Acid Sequence , Animals , Binding Sites , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , COS Cells , Chlorocebus aethiops , Humans , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mice , Molecular Sequence Data , PAX9 Transcription Factor/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Tooth/chemistry , Tooth/metabolism , Transcriptional Activation
17.
Clin Orthop Relat Res ; 462: 87-92, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17572636

ABSTRACT

Individuals with fibrodysplasia ossificans progressiva are born with malformations of the great toes and develop a heterotopic skeleton during childhood because of an identical heterozygous mutation in the glycine-serine activation domain of ACVR1, a bone morphogenetic protein type I receptor. Substitution of adenine for guanine at nucleotide 617 replaces an evolutionarily conserved arginine with histidine at residue 206 of ACVR1 in all classically affected individuals, making this one of the most highly conserved disease-causing mutations in the human genome. To better understand the molecular constraints and physiological implications of this mutation, we performed in silico modeling of wild-type and mutant ACVR1. In both the wild-type ACVR1 model and template crystal structures (TbetaRI), the conserved arginine appears to form a salt bridge with an invariant aspartate residue. Although lysine, a conservative substitution in BMPRIA and BMPRIB, can be readily accommodated, histidine at residue 206 (like in fibrodysplasia ossificans progressiva) would participate in a salt bridge with the aspartate only at decreased intracellular pH and with extensive structural rearrangement. Protein modeling predicts that substitution with histidine, and only histidine, creates a pH-sensitive switch within the activation domain of the receptor that leads to ligand-independent activation of ACVR1 in fibrodysplasia ossificans progressiva.


Subject(s)
Activin Receptors, Type I/genetics , Models, Genetic , Myositis Ossificans/genetics , Point Mutation , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Amino Acid Sequence , Amino Acid Substitution , Arginine/chemistry , Computer Simulation , Histidine/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Myositis Ossificans/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
18.
J Mol Biol ; 354(5): 1052-68, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16289576

ABSTRACT

Transforming growth factor-beta (TGFbeta) isoforms initiate signaling by assembling a heterotetrameric complex of paired type I (TbetaRI) and type II (TbetaRII) receptors on the cell surface. Because two of the ligand isoforms (TGFbetas 1, 3) must first bind TbetaRII to recruit TbetaRI into the complex, and a third (TGFbeta2) requires a co-receptor, assembly is known to be sequential, cooperative and isoform-dependent. However the source of the cooperativity leading to recruitment of TbetaRI and the universality of the assembly mechanism with respect to isoforms remain unclear. Here, we show that the extracellular domain of TbetaRI (TbetaRI-ED) binds in vitro with high affinity to complexes of the extracellular domain of TbetaRII (TbetaRII-ED) and TGFbetas 1 or 3, but not to either ligand or receptor alone. Thus, recruitment of TbetaRI requires combined interactions with TbetaRII-ED and ligand, but not membrane attachment of the receptors. Cell-based assays show that TbetaRI-ED, like TbetaRII-ED, acts as an antagonist of TGFbeta signaling, indicating that receptor-receptor interaction is sufficient to compete against endogenous, membrane-localized receptors. On the other hand, neither TbetaRII-ED, nor TbetaRII-ED and TbetaRI-ED combined, form a complex with TGFbeta2, showing that receptor-receptor interaction is insufficient to compensate for weak ligand-receptor interaction. However, TbetaRII-ED does bind with high affinity to TGFbeta2-TM, a TGFbeta2 variant substituted at three positions to mimic TGFbetas 1 and 3 at the TbetaRII binding interface. This proves both necessary and sufficient for recruitment of TbetaRI-ED, suggesting that the three different TGFbeta isoforms induce assembly of the heterotetrameric receptor complex in the same general manner.


Subject(s)
Activin Receptors, Type I/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/isolation & purification , Amino Acid Sequence , Animals , Cattle , Cell Division/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Escherichia coli/genetics , Female , Genes, Reporter , Genetic Variation , Humans , In Vitro Techniques , Ligands , Luciferases/metabolism , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Molecular Weight , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Smad2 Protein/analysis , Smad2 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
19.
J Mol Biol ; 352(2): 338-54, 2005 Sep 16.
Article in English | MEDLINE | ID: mdl-16083906

ABSTRACT

km23 (96 residues, 11 kDa) is the mammalian ortholog of Drosophila roadblock, the founding member of LC7/robl/km23 class of dynein light chains. km23 has been shown to be serine-phosphorylated following TGFbeta receptor activation and to bind the dynein intermediate chain in response to such phosphorylation. Here, we report the three-dimensional solution structure of km23, which is shown to be that of a homodimer, similar to that observed for the heterodimeric complex formed between p14 and MP1, two distantly related members of the MglB/robl superfamily, but distinct from the LC8 and Tctex-1 classes of dynein light chains, which also adopt homodimeric structures. The conserved surface residues of km23, including three serine residues, are located predominantly on a single face of the molecule. Adjacent to this face is a large cleft formed by the incomplete overlap of loops from opposite monomers. As shown by NMR relaxation data collected at two fields, several cleft residues are flexible on the ns-ps and ms-mus timescales. Based on these observations, we propose that the patch of conserved residues on the central face of the molecule corresponds to the site at which km23 binds the dynein intermediate chain and that the flexible cleft formed between the overlap of loops from the two monomers corresponds to the site at which km23 binds other partners, such as the TGFbeta type II receptor or Smad2.


Subject(s)
Dyneins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/genetics , Crystallography, X-Ray , Cytoplasmic Dyneins , Dimerization , Drosophila , Drosophila Proteins/genetics , Dyneins/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Serine/chemistry
20.
Rouxs Arch Dev Biol ; 205(1-2): 97-101, 1995 Sep.
Article in English | MEDLINE | ID: mdl-28306070

ABSTRACT

In the non-feeding larva of the marine gastropod, Haliotis rufescens, gut morphogenesis is initiated at metamorphosis. Intestine-specific chymotrypsin gene expression begins in amoebocytes located in the dorsoposterior region of the undifferentiated digestive gland prior to metamorphosis, 5 d post-fertilization. Transcript accumulates steadily in these cells over the next 6 d while the amoebocytes migrate slowly dorsally. Induction of metamorphosis dramatically accelerates the rates of chymotrypsin mRNA accumulation and amoebocyte migration, and is required for homing of the amoebocytes to the hindgut region. Induction of chymotrypsin gene expression occurs only in larvae that had developed competence to recognize an exogenous morphogenetic cue and initiate metamorphosis, with a more pronounced increase in chymotrypsin mRNA accumulation in occurring older larvae. Chymotrypsin mRNA accumulation patterns suggest that hindgut cell specification occurs prior to metamorphosis, but that completion of the morphogenetic program requires signaling events associated with metamorphosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...